1
|
Lei L, Wang YF, Chen CY, Wang YT, Zhang Y. Novel insight into astrocyte-mediated gliotransmission modulates the synaptic plasticity in major depressive disorder. Life Sci 2024; 355:122988. [PMID: 39153595 DOI: 10.1016/j.lfs.2024.122988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/23/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Major depressive disorder (MDD) is a form of glial cell-based synaptic dysfunction disease in which glial cells interact closely with neuronal synapses and perform synaptic information processing. Glial cells, particularly astrocytes, are active components of the brain and are responsible for synaptic activity through the release gliotransmitters. A reduced density of astrocytes and astrocyte dysfunction have both been identified the brains of patients with MDD. Furthermore, gliotransmission, i.e., active information transfer mediated by gliotransmitters between astrocytes and neurons, is thought to be involved in the pathogenesis of MDD. However, the mechanism by which astrocyte-mediated gliotransmission contributes to depression remains unknown. This review therefore summarizes the alterations in astrocytes in MDD, including astrocyte marker, connexin 43 (Cx43) expression, Cx43 gap junctions, and Cx43 hemichannels, and describes the regulatory mechanisms of astrocytes involved in synaptic plasticity. Additionally, we investigate the mechanisms acting of the glutamatergic, gamma-aminobutyric acidergic, and purinergic systems that modulate synaptic function and the antidepressant mechanisms of the related receptor antagonists. Further, we summarize the roles of glutamate, gamma-aminobutyric acid, d-serine, and adenosine triphosphate in depression, providing a basis for the identification of diagnostic and therapeutic targets for MDD.
Collapse
Affiliation(s)
- Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yu-Fei Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cong-Ya Chen
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ya-Ting Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
2
|
Peracchia C. Gap Junction Channel Regulation: A Tale of Two Gates-Voltage Sensitivity of the Chemical Gate and Chemical Sensitivity of the Fast Voltage Gate. Int J Mol Sci 2024; 25:982. [PMID: 38256055 PMCID: PMC10815820 DOI: 10.3390/ijms25020982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Gap junction channels are regulated by gates sensitive to cytosolic acidification and trans-junctional voltage (Vj). We propose that the chemical gate is a calmodulin (CaM) lobe. The fast-Vj gate is made primarily by the connexin's NH2-terminus domain (NT). The chemical gate closes the channel slowly and completely, while the fast-Vj gate closes the channel rapidly but incompletely. The chemical gate closes with increased cytosolic calcium concentration [Ca2+]i and with Vj gradients at Vj's negative side. In contrast, the fast-Vj gate closes at the positive or negative side of Vj depending on the connexin (Cx) type. Cxs with positively charged NT close at Vj's negative side, while those with negatively charged NT close at Vj's positive side. Cytosolic acidification alters in opposite ways the sensitivity of the fast-Vj gate: it increases the Vj sensitivity of negative gaters and decreases that of positive gaters. While the fast-Vj gate closes and opens instantaneously, the chemical gate often shows fluctuations, likely to reflect the shifting of the gate (CaM's N-lobe) in and out of the channel's pore.
Collapse
Affiliation(s)
- Camillo Peracchia
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University Rochester, Rochester, NY 14642-8711, USA
| |
Collapse
|
3
|
von Bernhardi R, Eugenín J. Aging Microglia and Their Impact in the Nervous System. ADVANCES IN NEUROBIOLOGY 2024; 37:379-395. [PMID: 39207703 DOI: 10.1007/978-3-031-55529-9_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Aging is the greatest risk factor for neurodegenerative diseases. Microglia are the resident immune cells in the central nervous system (CNS), playing key roles in its normal functioning, and as mediators for age-dependent changes of the CNS, condition at which they generate a hostile environment for neurons. Transforming Growth Factor β1 (TGFβ1) is a regulatory cytokine involved in immuneregulation and neuroprotection, affecting glial cell inflammatory activation, neuronal survival, and function. TGFβ1 signaling undergoes age-dependent changes affecting the regulation of microglial cells and can contribute to the pathophysiology of neurodegenerative diseases. This chapter focuses on assessing the role of age-related changes on the regulation of microglial cells and their impact on neuroinflammation and neuronal function, for understanding age-dependent changes of the nervous system.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Faculty of Odontology and Rehabilitation Sciences, Universidad San Sebastian, Santiago, Chile.
| | - Jaime Eugenín
- Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
4
|
van Megen WH, Canki E, Wagenaar VHA, van Waes CRMM, Peters DJM, Van Asbeck-Van der Wijst J, Hoenderop JGJ. Fluid shear stress stimulates ATP release without regulating purinergic gene expression in the renal inner medullary collecting duct. FASEB J 2023; 37:e23232. [PMID: 37819258 DOI: 10.1096/fj.202301434r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
In the kidney, the flow rate of the pro-urine through the renal tubules is highly variable. The tubular epithelial cells sense these variations in pro-urinary flow rate in order to regulate various physiological processes, including electrolyte reabsorption. One of the mechanosensitive pathways activated by flow is the release of ATP, which can then act as a autocrine or paracrine factor. Increased ATP release is observed in various kidney diseases, among others autosomal dominant polycystic kidney disease (ADPKD). However, the mechanisms underlying flow-induced ATP release in the collecting duct, especially in the inner medullary collecting duct, remain understudied. Using inner medullary collecting duct 3 (IMCD3) cells in a microfluidic setup, we show here that administration of a high flow rate for 1 min results in an increased ATP release compared to a lower flow rate. Although the ATP release channel pannexin-1 contributed to flow-induced ATP release in Pkd1-/- IMCD3 cells, it did not in wildtype IMCD3 cells. In addition, flow application increased the expression of the putative ATP release channel connexin-30.3 (CX30.3) in wildtype and Pkd1-/- IMCD3 cells. However, CX30.3 knockout IMCD3 cells exhibited a similar flow-induced ATP release as wildtype IMCD3 cells, suggesting that CX30.3 does not drive flow-induced ATP release in wildtype IMDC3 cells. Collectively, our results show differential mechanisms underlying flow-induced ATP release in wildtype and Pkd1-/- IMCD3 cells and further strengthen the link between ADPKD and pannexin-1-dependent ATP release.
Collapse
Affiliation(s)
- Wouter H van Megen
- Department of Medical Biosciences, Radboudumc, Nijmegen, The Netherlands
| | - Esra Canki
- Department of Medical Biosciences, Radboudumc, Nijmegen, The Netherlands
| | - Vera H A Wagenaar
- Department of Medical Biosciences, Radboudumc, Nijmegen, The Netherlands
| | | | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
5
|
Zhang X, Wang Y, Cai Z, Wan Z, Aihemaiti Y, Tu H. A gonadal gap junction INX-14/Notch GLP-1 signaling axis suppresses gut defense through an intestinal lysosome pathway. Front Immunol 2023; 14:1249436. [PMID: 37928537 PMCID: PMC10620905 DOI: 10.3389/fimmu.2023.1249436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
Gap junctions mediate intercellular communications across cellular networks in the nervous and immune systems. Yet their roles in intestinal innate immunity are poorly understood. Here, we show that the gap junction/innexin subunit inx-14 acts in the C. elegans gonad to attenuate intestinal defenses to Pseudomonas aeruginosa PA14 infection through the PMK-1/p38 pathway. RNA-Seq analyses revealed that germline-specific inx-14 RNAi downregulated Notch/GLP-1 signaling, while lysosome and PMK-1/p38 pathways were upregulated. Consistently, disruption of inx-14 or glp-1 in the germline enhanced resistance to PA14 infection and upregulated lysosome and PMK-1/p38 activity. We show that lysosome signaling functions downstream of the INX-14/GLP-1 signaling axis and upstream of PMK-1/p38 pathway to facilitate intestinal defense. Our findings expand the understanding of the links between the reproductive system and intestinal defense, which may be evolutionarily conserved in higher organism.
Collapse
Affiliation(s)
| | | | | | | | | | - Haijun Tu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| |
Collapse
|
6
|
Li H, Guo A, Salgado M, Sáez JC, Lau CG. The connexin hemichannel inhibitor D4 produces rapid antidepressant-like effects in mice. J Neuroinflammation 2023; 20:191. [PMID: 37599352 PMCID: PMC10440914 DOI: 10.1186/s12974-023-02873-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023] Open
Abstract
Depression is a common mood disorder characterized by a range of clinical symptoms, including prolonged low mood and diminished interest. Although many clinical and animal studies have provided significant insights into the pathophysiology of depression, current treatment strategies are not sufficient to manage this disorder. It has been suggested that connexin (Cx)-based hemichannels are candidates for depression intervention by modifying the state of neuroinflammation. In this study, we investigated the antidepressant-like effect of a recently discovered selective Cx hemichannel inhibitor, a small organic molecule called D4. We first showed that D4 reduced hemichannel activity following systemic inflammation after LPS injections. Next, we found that D4 treatment prevented LPS-induced inflammatory response and depressive-like behaviors. These behavioral effects were accompanied by reduced astrocytic activation and hemichannel activity in depressive-like mice induced by repeated low-dose LPS challenges. D4 treatment also reverses depressive-like symptoms in mice subjected to chronic restraint stress (CRS). To test whether D4 broadly affected neural activity, we measured c-Fos expression in depression-related brain regions and found a reduction in c-Fos+ cells in different brain regions. D4 significantly normalized CRS-induced hypoactivation in several brain regions, including the hippocampus, entorhinal cortex, and lateral septum. Together, these results indicate that blocking Cx hemichannels using D4 can normalize neuronal activity and reduce depressive-like symptoms in mice by reducing neuroinflammation. Our work provides evidence of the antidepressant-like effect of D4 and supports glial Cx hemichannels as potential therapeutic targets for depression.
Collapse
Affiliation(s)
- Huanhuan Li
- Department of Neuroscience, City University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| | - Anni Guo
- Department of Neuroscience, City University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| | - Magdiel Salgado
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, 2381850, Valparaíso, Chile
| | - Juan C Sáez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, 2381850, Valparaíso, Chile
| | - Chunyue Geoffrey Lau
- Department of Neuroscience, City University of Hong Kong, Hong Kong SAR, China.
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China.
| |
Collapse
|
7
|
Wang Y, Jin Y, Zhang Q, Xiong Y, Gu X, Zeng S, Chen W. Research progress in delineating the pathological mechanisms of GJB2-related hearing loss. Front Cell Neurosci 2023; 17:1208406. [PMID: 37333892 PMCID: PMC10272732 DOI: 10.3389/fncel.2023.1208406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Hearing loss is the most common congenital sensory impairment. Mutations or deficiencies of the GJB2 gene are the most common genetic cause of congenital non-syndromic deafness. Pathological changes such as decreased potential in the cochlea, active cochlear amplification disorders, cochlear developmental disorders and macrophage activation have been observed in various GJB2 transgenic mouse models. In the past, researchers generally believed that the pathological mechanisms underlying GJB2-related hearing loss comprised a K+ circulation defect and abnormal ATP-Ca2+ signals. However, recent studies have shown that K+ circulation is rarely associated with the pathological process of GJB2-related hearing loss, while cochlear developmental disorders and oxidative stress play an important, even critical, role in the occurrence of GJB2-related hearing loss. Nevertheless, these research has not been systematically summarized. In this review, we summarize the pathological mechanisms of GJB2-related hearing loss, including aspects of K+ circulation, developmental disorders of the organ of Corti, nutrition delivery, oxidative stress and ATP-Ca2+ signals. Clarifying the pathological mechanism of GJB2-related hearing loss can help develop new prevention and treatment strategies.
Collapse
Affiliation(s)
- Yujun Wang
- Department of Intensive Care Unit, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Jin
- Department of Otorhinolaryngology–Head and Neck Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiong Zhang
- Department of Otorhinolaryngology–Head and Neck Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Xiong
- Department of Otorhinolaryngology–Head and Neck Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Gu
- Department of Otorhinolaryngology–Head and Neck Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Zeng
- Department of Otorhinolaryngology–Head and Neck Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Chen
- Department of Otorhinolaryngology–Head and Neck Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Man JHK, van Gelder CAGH, Breur M, Okkes D, Molenaar D, van der Sluis S, Abbink T, Altelaar M, van der Knaap MS, Bugiani M. Cortical Pathology in Vanishing White Matter. Cells 2022; 11:cells11223581. [PMID: 36429009 PMCID: PMC9688115 DOI: 10.3390/cells11223581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/24/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Vanishing white matter (VWM) is classified as a leukodystrophy with astrocytes as primary drivers in its pathogenesis. Magnetic resonance imaging has documented the progressive thinning of cortices in long-surviving patients. Routine histopathological analyses, however, have not yet pointed to cortical involvement in VWM. Here, we provide a comprehensive analysis of the VWM cortex. We employed high-resolution-mass-spectrometry-based proteomics and immunohistochemistry to gain insight into possible molecular disease mechanisms in the cortices of VWM patients. The proteome analysis revealed 268 differentially expressed proteins in the VWM cortices compared to the controls. A majority of these proteins formed a major protein interaction network. A subsequent gene ontology analysis identified enrichment for terms such as cellular metabolism, particularly mitochondrial activity. Importantly, some of the proteins with the most prominent changes in expression were found in astrocytes, indicating cortical astrocytic involvement. Indeed, we confirmed that VWM cortical astrocytes exhibit morphological changes and are less complex in structure than control cells. Our findings also suggest that these astrocytes are immature and not reactive. Taken together, we provide insights into cortical involvement in VWM, which has to be taken into account when developing therapeutic strategies.
Collapse
Affiliation(s)
- Jodie H. K. Man
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Charlotte A. G. H. van Gelder
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CS Utrecht, The Netherlands
- Netherlands Proteomics Center, 3584 CS Utrecht, The Netherlands
| | - Marjolein Breur
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Daniel Okkes
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Douwe Molenaar
- Department of Systems Bioinformatics, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sophie van der Sluis
- Department of Child and Adolescent Psychology and Psychiatry, Complex Trait Genetics, Amsterdam Neuroscience, VU University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Truus Abbink
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CS Utrecht, The Netherlands
- Netherlands Proteomics Center, 3584 CS Utrecht, The Netherlands
| | - Marjo S. van der Knaap
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Marianna Bugiani
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
- Department of Pathology, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-6-48517239
| |
Collapse
|
9
|
Rieck J, Skatchkov SN, Derst C, Eaton MJ, Veh RW. Unique Chemistry, Intake, and Metabolism of Polyamines in the Central Nervous System (CNS) and Its Body. Biomolecules 2022; 12:biom12040501. [PMID: 35454090 PMCID: PMC9025450 DOI: 10.3390/biom12040501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Polyamines (PAs) are small, versatile molecules with two or more nitrogen-containing positively charged groups and provide widespread biological functions. Most of these aspects are well known and covered by quite a number of excellent surveys. Here, the present review includes novel aspects and questions: (1) It summarizes the role of most natural and some important synthetic PAs. (2) It depicts PA uptake from nutrition and bacterial production in the intestinal system following loss of PAs via defecation. (3) It highlights the discrepancy between the high concentrations of PAs in the gut lumen and their low concentration in the blood plasma and cerebrospinal fluid, while concentrations in cellular cytoplasm are much higher. (4) The present review provides a novel and complete scheme for the biosynthesis of Pas, including glycine, glutamate, proline and others as PA precursors, and provides a hypothesis that the agmatine pathway may rescue putrescine production when ODC knockout seems to be lethal (solving the apparent contradiction in the literature). (5) It summarizes novel data on PA transport in brain glial cells explaining why these cells but not neurons preferentially accumulate PAs. (6) Finally, it provides a novel and complete scheme for PA interconversion, including hypusine, putreanine, and GABA (unique gliotransmitter) as end-products. Altogether, this review can serve as an updated contribution to understanding the PA mystery.
Collapse
Affiliation(s)
- Julian Rieck
- Institut für Zell- und Neurobiologie, Centrum 2, Charité—Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany;
| | - Serguei N. Skatchkov
- Department of Physiology, Universidad Central del Caribe, Bayamón, PR 00956, USA
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00956, USA;
- Correspondence: (S.N.S.); (R.W.V.)
| | - Christian Derst
- Institut für Integrative Neuroanatomie, Centrum 2, Charité—Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany;
| | - Misty J. Eaton
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00956, USA;
| | - Rüdiger W. Veh
- Institut für Zell- und Neurobiologie, Centrum 2, Charité—Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany;
- Correspondence: (S.N.S.); (R.W.V.)
| |
Collapse
|
10
|
Unal YC, Yavuz B, Ozcivici E, Mese G. The role of connexins in breast cancer: from misregulated cell communication to aberrant intracellular signaling. Tissue Barriers 2022; 10:1962698. [PMID: 34355641 PMCID: PMC8794248 DOI: 10.1080/21688370.2021.1962698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 12/24/2022] Open
Abstract
In spite of clinical advancements and improved diagnostic techniques, breast cancers are the leading cause of cancer-associated deaths in women worldwide. Although 70% of early breast cancers can be cured, there are no efficient therapies against metastatic breast cancers. Several factors including connexins and gap junctions play roles in breast tumorigenesis. Connexins are critical for cellular processes as a linkage between connexin mutations and hereditary disorders demonstrated their importance for tissue homeostasis. Further, alterations in their expression, localization and channel activities were observed in many cancers including breast cancer. Both channel-dependent and independent functions of connexins were reported in initiation and progression of cancers. Unlike initial reports suggesting tumor suppressor functions, connexins and gap junctions have stage, context and isoform dependent effects in breast cancers similar to other cancers. In this review, we tried to describe the current understanding of connexins in tumorigenesis specifically in breast cancers.
Collapse
Affiliation(s)
- Yagmur Ceren Unal
- Faculty of Science, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Busra Yavuz
- Faculty of Science, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Engin Ozcivici
- Department of Bioengineering, Faculty of Engineering, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Gulistan Mese
- Faculty of Science, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, Turkey
| |
Collapse
|
11
|
Shen XY, Gao ZK, Han Y, Yuan M, Guo YS, Bi X. Activation and Role of Astrocytes in Ischemic Stroke. Front Cell Neurosci 2021; 15:755955. [PMID: 34867201 PMCID: PMC8635513 DOI: 10.3389/fncel.2021.755955] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022] Open
Abstract
Ischemic stroke refers to the disorder of blood supply of local brain tissue caused by various reasons. It has high morbidity and mortality worldwide. Astrocytes are the most abundant glial cells in the central nervous system (CNS). They are responsible for the homeostasis, nutrition, and protection of the CNS and play an essential role in many nervous system diseases’ physiological and pathological processes. After stroke injury, astrocytes are activated and play a protective role through the heterogeneous and gradual changes of their gene expression, morphology, proliferation, and function, that is, reactive astrocytes. However, the position of reactive astrocytes has always been a controversial topic. Many studies have shown that reactive astrocytes are a double-edged sword with both beneficial and harmful effects. It is worth noting that their different spatial and temporal expression determines astrocytes’ various functions. Here, we comprehensively review the different roles and mechanisms of astrocytes after ischemic stroke. In addition, the intracellular mechanism of astrocyte activation has also been involved. More importantly, due to the complex cascade reaction and action mechanism after ischemic stroke, the role of astrocytes is still difficult to define. Still, there is no doubt that astrocytes are one of the critical factors mediating the deterioration or improvement of ischemic stroke.
Collapse
Affiliation(s)
- Xin-Ya Shen
- Graduate School of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhen-Kun Gao
- Graduate School of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Han
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Mei Yuan
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Yi-Sha Guo
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
12
|
Talukdar S, Emdad L, Das SK, Fisher PB. GAP junctions: multifaceted regulators of neuronal differentiation. Tissue Barriers 2021; 10:1982349. [PMID: 34651545 DOI: 10.1080/21688370.2021.1982349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Gap junctions are intercellular membrane channels consisting of connexin proteins, which contribute to direct cytoplasmic exchange of small molecules, substrates and metabolites between adjacent cells. These channels play important roles in neuronal differentiation, maintenance, survival and function. Gap junctions regulate differentiation of neurons from embryonic, neural and induced pluripotent stem cells. In addition, they control transdifferentiation of neurons from mesenchymal stem cells. The expression and levels of several connexins correlate with cell cycle changes and different stages of neurogenesis. Connexins such as Cx36, Cx45, and Cx26, play a crucial role in neuronal function. Several connexin knockout mice display lethal or severely impaired phenotypes. Aberrations in connexin expression is frequently associated with various neurodegenerative disorders. Gap junctions also act as promising therapeutic targets for neuronal regenerative medicine, because of their role in neural stem cell integration, injury and remyelination.
Collapse
Affiliation(s)
- Sarmistha Talukdar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.,Vcu Institute of Molecular Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.,Vcu Institute of Molecular Medicine, Richmond, VA, United States.,Vcu Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.,Vcu Institute of Molecular Medicine, Richmond, VA, United States.,Vcu Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.,Vcu Institute of Molecular Medicine, Richmond, VA, United States.,Vcu Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| |
Collapse
|
13
|
Baer SM, Chang S, Crook SM, Gardner CL, Jones JR, Ringhofer C, Nelson RF. A multiscale continuum model of the vertebrate outer retina: The temporal dynamics of background-induced flicker enhancement. J Theor Biol 2021; 525:110763. [PMID: 34000285 PMCID: PMC11385586 DOI: 10.1016/j.jtbi.2021.110763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 04/28/2021] [Accepted: 05/07/2021] [Indexed: 11/25/2022]
Abstract
The retina is a part of the central nervous system that is accessible, well documented, and studied by researchers spanning the clinical, experimental, and theoretical sciences. Here, we mathematically model the subcircuits of the outer plexiform layer of the retina on two spatial scales: that of an individual synapse and that of the scale of the receptive field (hundreds to thousands of synapses). To this end we formulate a continuum spine model (a partial differential equation system) that incorporates the horizontal cell syncytium and its numerous processes (spines) within cone pedicles. With this multiscale modeling approach, detailed biophysical mechanisms at the synaptic level are retained while scaling up to the receptive field level. As an example of its utility, the model is applied to study background-induced flicker enhancement in which the onset of a dim background enhances the center flicker response of horizontal cells. Simulation results, in comparison with flicker enhancement data for square, slit, and disk test regions, suggest that feedback mechanisms that are voltage-axis modulators of cone calcium channels (for example, ephaptic and/or pH feedback) are robust in capturing the temporal dynamics of background-induced flicker enhancement. The value and potential of this continuum spine approach is that it provides a framework for mathematically modeling the input-output properties of the entire receptive field of the outer retina while implementing the latest models for transmission mechanisms at the synaptic level.
Collapse
Affiliation(s)
- Steven M Baer
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States.
| | - Shaojie Chang
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States; The High School Affiliated to Beijing Normal University, Beijing 100052, PR China
| | - Sharon M Crook
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States
| | - Carl L Gardner
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States
| | - Jeremiah R Jones
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States
| | - Christian Ringhofer
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States
| | - Ralph F Nelson
- Neural Circuits Unit, Basic Neuroscience Program, NINDS, NIH, Bethesda, MD 20892, United States
| |
Collapse
|
14
|
Uptake of Biotinylated Spermine in Astrocytes: Effect of Cx43 siRNA, HIV-Tat Protein and Polyamine Transport Inhibitor on Polyamine Uptake. Biomolecules 2021; 11:biom11081187. [PMID: 34439853 PMCID: PMC8391674 DOI: 10.3390/biom11081187] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/06/2021] [Accepted: 08/08/2021] [Indexed: 12/14/2022] Open
Abstract
Polyamines (PAs) are polycationic biomolecules containing multiple amino groups. Patients with HIV-associated neurocognitive disorder (HAND) have high concentrations of the polyamine N-acetylated spermine in their brain and cerebral spinal fluid (CSF) and have increased PA release from astrocytes. These effects are due to the exposure to HIV-Tat. In healthy adult brain, PAs are accumulated but not synthesized in astrocytes, suggesting that PAs must enter astrocytes to be N-acetylated and released. Therefore, we tested if Cx43 hemichannels (Cx43-HCs) are pathways for PA flux in control and HIV-Tat-treated astrocytes. We used biotinylated spermine (b-SPM) to examine polyamine uptake. We found that control astrocytes and those treated with siRNA-Cx43 took up b-SPM, similarly suggesting that PA uptake is via a transporter/channel other than Cx43-HCs. Surprisingly, astrocytes pretreated with both HIV-Tat and siRNA-Cx43 showed increased accumulation of b-SPM. Using a novel polyamine transport inhibitor (PTI), trimer 44NMe, we blocked b-SPM uptake, showing that PA uptake is via a PTI-sensitive transport mechanism such as organic cation transporter. Our data suggest that Cx43 HCs are not a major pathway for b-SPM uptake in the condition of normal extracellular calcium concentration but may be involved in the release of PAs to the extracellular space during viral infection.
Collapse
|
15
|
Mazaud D, Capano A, Rouach N. The many ways astroglial connexins regulate neurotransmission and behavior. Glia 2021; 69:2527-2545. [PMID: 34101261 DOI: 10.1002/glia.24040] [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: 08/16/2020] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 12/18/2022]
Abstract
Astrocytes have emerged as major players in the brain, contributing to many functions such as energy supply, neurotransmission, and behavior. They accomplish these functions in part via their capacity to form widespread intercellular networks and to release neuroactive factors, which can modulate neurotransmission at different levels, from individual synapses to neuronal networks. The extensive network communication of astrocytes is primarily mediated by gap junction channels composed of two connexins, Cx30 and Cx43, which present distinct temporal and spatial expression patterns. Yet, astroglial connexins are also involved in direct exchange with the extracellular space via hemichannels, as well as in adhesion and signaling processes via unconventional nonchannel functions. Accumulating evidence indicate that astrocytes modulate neurotransmission and behavior through these diverse connexin functions. We here review the many ways astroglial connexins regulate neuronal activity from the molecular level to behavior.
Collapse
Affiliation(s)
- David Mazaud
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | - Anna Capano
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France.,Doctoral School N°158, Sorbonne University, Paris, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| |
Collapse
|
16
|
González-Casanova J, Schmachtenberg O, Martínez AD, Sanchez HA, Harcha PA, Rojas-Gomez D. An Update on Connexin Gap Junction and Hemichannels in Diabetic Retinopathy. Int J Mol Sci 2021; 22:ijms22063194. [PMID: 33801118 PMCID: PMC8004116 DOI: 10.3390/ijms22063194] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/06/2021] [Accepted: 03/10/2021] [Indexed: 01/10/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the main causes of vision loss in the working age population. It is characterized by a progressive deterioration of the retinal microvasculature, caused by long-term metabolic alterations inherent to diabetes, leading to a progressive loss of retinal integrity and function. The mammalian retina presents an orderly layered structure that executes initial but complex visual processing and analysis. Gap junction channels (GJC) forming electrical synapses are present in each retinal layer and contribute to the communication between different cell types. In addition, connexin hemichannels (HCs) have emerged as relevant players that influence diverse physiological and pathological processes in the retina. This article highlights the impact of diabetic conditions on GJC and HCs physiology and their involvement in DR pathogenesis. Microvascular damage and concomitant loss of endothelial cells and pericytes are related to alterations in gap junction intercellular communication (GJIC) and decreased connexin 43 (Cx43) expression. On the other hand, it has been shown that the expression and activity of HCs are upregulated in DR, becoming a key element in the establishment of proinflammatory conditions that emerge during hyperglycemia. Hence, novel connexin HCs blockers or drugs to enhance GJIC are promising tools for the development of pharmacological interventions for diabetic retinopathy, and initial in vitro and in vivo studies have shown favorable results in this regard.
Collapse
Affiliation(s)
- Jorge González-Casanova
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910060, Chile;
| | - Oliver Schmachtenberg
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Biología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile;
| | - Agustín D. Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.D.M.); (H.A.S.); (P.A.H.)
| | - Helmuth A. Sanchez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.D.M.); (H.A.S.); (P.A.H.)
| | - Paloma A. Harcha
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.D.M.); (H.A.S.); (P.A.H.)
| | - Diana Rojas-Gomez
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Santiago 8370146, Chile
- Correspondence: ; Tel.: +56-2-26618559
| |
Collapse
|
17
|
Buckley C, Zhang X, Wilson C, McCarron JG. Carbenoxolone and 18β-glycyrrhetinic acid inhibit inositol 1,4,5-trisphosphate-mediated endothelial cell calcium signalling and depolarise mitochondria. Br J Pharmacol 2021; 178:896-912. [PMID: 33269468 PMCID: PMC9328419 DOI: 10.1111/bph.15329] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/08/2020] [Accepted: 09/19/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Coordinated endothelial control of cardiovascular function is proposed to occur by endothelial cell communication via gap junctions and connexins. To study intercellular communication, the pharmacological agents carbenoxolone (CBX) and 18β-glycyrrhetinic acid (18βGA) are used widely as connexin inhibitors and gap junction blockers. EXPERIMENTAL APPROACH We investigated the effects of CBX and 18βGA on intercellular Ca2+ waves, evoked by inositol 1,4,5-trisphosphate (IP3 ) in the endothelium of intact mesenteric resistance arteries. KEY RESULTS Acetycholine-evoked IP3 -mediated Ca2+ release and propagated waves were inhibited by CBX (100 μM) and 18βGA (40 μM). Unexpectedly, the Ca2+ signals were inhibited uniformly in all cells, suggesting that CBX and 18βGA reduced Ca2+ release. Localised photolysis of caged IP3 (cIP3 ) was used to provide precise spatiotemporal control of site of cell activation. Local cIP3 photolysis generated reproducible Ca2+ increases and Ca2+ waves that propagated across cells distant to the photolysis site. CBX and 18βGA each blocked Ca2+ waves in a time-dependent manner by inhibiting the initiating IP3 -evoked Ca2+ release event rather than block of gap junctions. This effect was reversed on drug washout and was unaffected by small or intermediate K+ -channel blockers. Furthermore, CBX and 18βGA each rapidly and reversibly collapsed the mitochondrial membrane potential. CONCLUSION AND IMPLICATIONS CBX and 18βGA inhibit IP3 -mediated Ca2+ release and depolarise the mitochondrial membrane potential. These results suggest that CBX and 18βGA may block cell-cell communication by acting at sites that are unrelated to gap junctions.
Collapse
Affiliation(s)
- Charlotte Buckley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
18
|
Andelova K, Egan Benova T, Szeiffova Bacova B, Sykora M, Prado NJ, Diez ER, Hlivak P, Tribulova N. Cardiac Connexin-43 Hemichannels and Pannexin1 Channels: Provocative Antiarrhythmic Targets. Int J Mol Sci 2020; 22:ijms22010260. [PMID: 33383853 PMCID: PMC7795512 DOI: 10.3390/ijms22010260] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiac connexin-43 (Cx43) creates gap junction channels (GJCs) at intercellular contacts and hemi-channels (HCs) at the peri-junctional plasma membrane and sarcolemmal caveolae/rafts compartments. GJCs are fundamental for the direct cardiac cell-to-cell transmission of electrical and molecular signals which ensures synchronous myocardial contraction. The HCs and structurally similar pannexin1 (Panx1) channels are active in stressful conditions. These channels are essential for paracrine and autocrine communication through the release of ions and signaling molecules to the extracellular environment, or for uptake from it. The HCs and Panx1 channel-opening profoundly affects intracellular ionic homeostasis and redox status and facilitates via purinergic signaling pro-inflammatory and pro-fibrotic processes. These conditions promote cardiac arrhythmogenesis due to the impairment of the GJCs and selective ion channel function. Crosstalk between GJCs and HCs/Panx1 channels could be crucial in the development of arrhythmogenic substrates, including fibrosis. Despite the knowledge gap in the regulation of these channels, current evidence indicates that HCs and Panx1 channel activation can enhance the risk of cardiac arrhythmias. It is extremely challenging to target HCs and Panx1 channels by inhibitory agents to hamper development of cardiac rhythm disorders. Progress in this field may contribute to novel therapeutic approaches for patients prone to develop atrial or ventricular fibrillation.
Collapse
Affiliation(s)
- Katarina Andelova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 84104 Bratislava, Slovakia; (K.A.); (T.E.B.); (B.S.B.); (M.S.)
| | - Tamara Egan Benova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 84104 Bratislava, Slovakia; (K.A.); (T.E.B.); (B.S.B.); (M.S.)
| | - Barbara Szeiffova Bacova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 84104 Bratislava, Slovakia; (K.A.); (T.E.B.); (B.S.B.); (M.S.)
| | - Matus Sykora
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 84104 Bratislava, Slovakia; (K.A.); (T.E.B.); (B.S.B.); (M.S.)
| | - Natalia Jorgelina Prado
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, M5500 Mendoza, Argentina; (N.J.P.); (E.R.D.)
| | - Emiliano Raul Diez
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, M5500 Mendoza, Argentina; (N.J.P.); (E.R.D.)
| | - Peter Hlivak
- Department of Arrhythmias and Pacing, National Institute of Cardiovascular Diseases, Pod Krásnou Hôrkou 1, 83348 Bratislava, Slovakia;
| | - Narcis Tribulova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 84104 Bratislava, Slovakia; (K.A.); (T.E.B.); (B.S.B.); (M.S.)
- Correspondence: ; Tel.: +421-2-32295-423
| |
Collapse
|
19
|
Connexins-Therapeutic Targets in Cancers. Int J Mol Sci 2020; 21:ijms21239119. [PMID: 33266154 PMCID: PMC7730856 DOI: 10.3390/ijms21239119] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022] Open
Abstract
Connexins (Cx) are members of a protein family that forms intercellular channels localised in gap junction (GJ) plaques and single transmembrane channels called hemichannels. They participate in intercellular communication or communication between the intracellular and extracellular environments. Connexins affect cell homeostasis, growth and differentiation by enabling the exchange of metabolites or by interfering with various signalling pathways. Alterations in the functionality and the expression of connexins have been linked to the occurrence of many diseases. Connexins have been already linked to cancers, cardiac and brain disorders, chronic lung and kidney conditions and wound healing processes. Connexins have been shown either to suppress cancer tumour growth or to increase tumorigenicity by promoting cancer cell growth, migration and invasiveness. A better understanding of the complexity of cancer biology related to connexins and intercellular communication could result in the design of novel therapeutic strategies. The modulation of connexin expression may be an effective therapeutic approach in some types of cancers. Therefore, one important challenge is the search for mechanisms and new drugs, selectively modulating the expression of various connexin isoforms. We performed a systematic literature search up to February 2020 in the electronic databases PubMed and EMBASE. Our search terms were as follows: connexins, hemichannels, cancer and cancer treatment. This review aims to provide information about the role of connexins and gap junctions in cancer, as well as to discuss possible therapeutic options that are currently being studied.
Collapse
|
20
|
Maulik M, Vasan L, Bose A, Dutta Chowdhury S, Sengupta N, Das Sarma J. Amyloid-β regulates gap junction protein connexin 43 trafficking in cultured primary astrocytes. J Biol Chem 2020; 295:15097-15111. [PMID: 32868453 DOI: 10.1074/jbc.ra120.013705] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/06/2020] [Indexed: 11/06/2022] Open
Abstract
Altered expression and function of astroglial gap junction protein connexin 43 (Cx43) has increasingly been associated to neurotoxicity in Alzheimer disease (AD). Although earlier studies have examined the effect of increased β-amyloid (Aβ) on Cx43 expression and function leading to neuronal damage, underlying mechanisms by which Aβ modulates Cx43 in astrocytes remain elusive. Here, using mouse primary astrocyte cultures, we have examined the cellular processes by which Aβ can alter Cx43 gap junctions. We show that Aβ25-35 impairs functional gap junction coupling yet increases hemichannel activity. Interestingly, Aβ25-35 increased the intracellular pool of Cx43 with a parallel decrease in gap junction assembly at the surface. Intracellular Cx43 was found to be partly retained in the endoplasmic reticulum-associated cell compartments. However, forward trafficking of the newly synthesized Cx43 that already reached the Golgi was not affected in Aβ25-35-exposed astrocytes. Supporting this, treatment with 4-phenylbutyrate, a well-known chemical chaperone that improves trafficking of several transmembrane proteins, restored Aβ-induced impaired gap junction coupling between astrocytes. We further show that interruption of Cx43 endocytosis in Aβ25-35-exposed astrocytes resulted in their retention at the cell surface in the form of functional gap junctions indicating that Aβ25-35 causes rapid internalization of Cx43 gap junctions. Additionally, in silico molecular docking suggests that Aβ can bind favorably to Cx43. Our study thus provides novel insights into the cellular mechanisms by which Aβ modulates Cx43 function in astrocytes, the basic understanding of which is vital for the development of alternative therapeutic strategy targeting connexin channels in AD.
Collapse
Affiliation(s)
- Mahua Maulik
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India.
| | - Lakshmy Vasan
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Abhishek Bose
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Saikat Dutta Chowdhury
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Neelanjana Sengupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| |
Collapse
|
21
|
Leroy K, Pieters A, Tabernilla A, Cooreman A, Van Campenhout R, Cogliati B, Vinken M. Targeting gap junctional intercellular communication by hepatocarcinogenic compounds. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2020; 23:255-275. [PMID: 32568623 DOI: 10.1080/10937404.2020.1781010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Gap junctions in liver, as in other organs, play a critical role in tissue homeostasis. Inherently, these cellular constituents are major targets for systemic toxicity and diseases, including cancer. This review provides an overview of chemicals that compromise liver gap junctions, in particular biological toxins, organic solvents, pesticides, pharmaceuticals, peroxides, metals and phthalates. The focus in this review is placed upon the mechanistic scenarios that underlie these adverse effects. Further, the potential use of gap junctional activity as an in vitro biomarker to identify non-genotoxic hepatocarcinogenic chemicals is discussed.
Collapse
Affiliation(s)
- Kaat Leroy
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , Brussels, Belgium
| | - Alanah Pieters
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , Brussels, Belgium
| | - Andrés Tabernilla
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , Brussels, Belgium
| | - Axelle Cooreman
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , Brussels, Belgium
| | - Raf Van Campenhout
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , Brussels, Belgium
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Cidade Universitária , São Paulo, Brazil
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , Brussels, Belgium
| |
Collapse
|
22
|
Liang Z, Wang X, Hao Y, Qiu L, Lou Y, Zhang Y, Ma D, Feng J. The Multifaceted Role of Astrocyte Connexin 43 in Ischemic Stroke Through Forming Hemichannels and Gap Junctions. Front Neurol 2020; 11:703. [PMID: 32849190 PMCID: PMC7411525 DOI: 10.3389/fneur.2020.00703] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022] Open
Abstract
Ischemic stroke is a multi-factorial cerebrovascular disease with high worldwide morbidity and mortality. In the past few years, multiple studies have revealed the underlying mechanism of ischemia/reperfusion injury, including calcium overload, amino acid toxicity, oxidative stress, and inflammation. Connexin 43 (Cx43), the predominant connexin protein in astrocytes, has been recently proven to display non-substitutable roles in the pathology of ischemic stroke development and progression through forming gap junctions and hemichannels. Under normal conditions, astrocytic Cx43 could be found in hemichannels or in the coupling with other hemichannels on astrocytes, neurons, or oligodendrocytes to form the neuro-glial syncytium, which is involved in metabolites exchange between communicated cells, thus maintaining the homeostasis of the CNS environment. In ischemic stroke, the phosphorylation of Cx43 might cause the degradation of gap junctions and the opening of hemichannels, contributing to the release of inflammatory mediators. However, the remaining gap junctions could facilitate the exchange of protective and harmful metabolites between healthy and injured cells, protecting the injured cells to some extent or damaging the healthy cells depending on the balance of the exchange of protective and harmful metabolites. In this study, we review the changes in astrocytic Cx43 expression and distribution as well as the influence of these changes on the function of astrocytes and other cells in the CNS, providing new insight into the pathology of ischemic stroke injury; we also discuss the potential of astrocytic Cx43 as a target for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zhen Liang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Lin Qiu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yingyue Lou
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yaoting Zhang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Inoue A, Nakao-Kuroishi K, Kometani-Gunjigake K, Mizuhara M, Shirakawa T, Ito-Sago M, Yasuda K, Nakatomi M, Matsubara T, Tada-Shigeyama Y, Morikawa K, Kokabu S, Kawamoto T. VNUT/SLC17A9, a vesicular nucleotide transporter, regulates osteoblast differentiation. FEBS Open Bio 2020; 10:1612-1623. [PMID: 32592329 PMCID: PMC7396442 DOI: 10.1002/2211-5463.12918] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/04/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Osteoblasts release adenosine triphosphate (ATP) out of the cell following mechanical stress. Although it is well established that extracellular ATP affects bone metabolism via P2 receptors [such as purinergic receptor P2X7 (P2X7R) and purinergic receptor P2Y2 (P2Y2R)], the mechanism of ATP release from osteoblasts remains unknown. Recently, a vesicular nucleotide transporter [VNUT, solute carrier family 17 member 9 (SLC17A9)] that preserves ATP in vesicles has been identified. The purpose of this study was to elucidate the role of VNUT in osteoblast bone metabolism. mRNA and protein expression of VNUT were confirmed in mouse bone and in osteoblasts by quantitative real-time PCR (qPCR) and immunohistochemistry. Next, when compressive force was applied to MC3T3-E1 cells by centrifugation, the expression of Slc17a9, P2x7r, and P2y2r was increased concomitant with an increase in extracellular ATP levels. Furthermore, compressive force decreased the osteoblast differentiation capacity of MC3T3-E1 cells. shRNA knockdown of Slc17a9 in MC3T3-E1 cells reduced levels of extracellular ATP and also led to increased osteoblast differentiation after the application of compressive force as assessed by qPCR analysis of osteoblast markers such as Runx2, Osterix, and alkaline phosphatase (ALP) as well as ALP activity. Consistent with these observations, knockdown of P2x7r or P2y2r by siRNA partially rescued the downregulation of osteoblast differentiation markers, caused by mechanical loading. In conclusion, our results demonstrate that VNUT is expressed in osteoblasts and that VNUT inhibits osteoblast differentiation in response to compressive force by mechanisms related to ATP release and P2X7R and/or P2Y2R activity.
Collapse
Affiliation(s)
- Asako Inoue
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Kayoko Nakao-Kuroishi
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Kaori Kometani-Gunjigake
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Masahiro Mizuhara
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Tomohiko Shirakawa
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Misa Ito-Sago
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Kazuma Yasuda
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Mitsushiro Nakatomi
- Division of Anatomy, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Yukiyo Tada-Shigeyama
- Division of Dental Anesthesiology, Department of Science of Physical Functions, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Kazumasa Morikawa
- Division of Pediatric and Special Care Dentistry, Department of Developmental Oral Health Science, School of Dentistry, Iwate Medical University, Morioka, Japan
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| | - Tatsuo Kawamoto
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu-shi, Japan
| |
Collapse
|
24
|
Giaume C, Naus CC, Sáez JC, Leybaert L. Glial Connexins and Pannexins in the Healthy and Diseased Brain. Physiol Rev 2020; 101:93-145. [PMID: 32326824 DOI: 10.1152/physrev.00043.2018] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Over the past several decades a large amount of data have established that glial cells, the main cell population in the brain, dynamically interact with neurons and thus impact their activity and survival. One typical feature of glia is their marked expression of several connexins, the membrane proteins forming intercellular gap junction channels and hemichannels. Pannexins, which have a tetraspan membrane topology as connexins, are also detected in glial cells. Here, we review the evidence that connexin and pannexin channels are actively involved in dynamic and metabolic neuroglial interactions in physiological as well as in pathological situations. These features of neuroglial interactions open the way to identify novel non-neuronal aspects that allow for a better understanding of behavior and information processing performed by neurons. This will also complement the "neurocentric" view by facilitating the development of glia-targeted therapeutic strategies in brain disease.
Collapse
Affiliation(s)
- Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Christian C Naus
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Juan C Sáez
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Luc Leybaert
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
25
|
Establishment and functional characterization of a murine primary Sertoli cell line deficient of connexin43. Cell Tissue Res 2020; 381:309-326. [PMID: 32328805 PMCID: PMC7369266 DOI: 10.1007/s00441-020-03203-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 03/12/2020] [Indexed: 12/12/2022]
Abstract
The Sertoli cell (SC) specific connexin43 (Cx43) knockout (SCCx43KO) mouse line is ideal to gain insight into the mechanistic gap junction formation in SC and the seminiferous epithelium. A method for developing primary SC cultures from these mice was established, validated and successfully characterized via polymerase chain reaction, immunohistochemistry, immunofluorescence (IF), and Western blots (WB). It was evident that both knockout (KO) and wild-type (WT) primary cell cultures were similar in morphology. These highly pure SC cultures were subjected to cell proliferation assays indicating no notable proliferation in cultures of both genotypes. Measurements of cell monolayer integrity indicated significant increases in transepithelial electrical resistance and consequently in tight junction expression of the KO cultures. Using semi-quantitative WB and IF, tight junction protein claudin-11 was analyzed. These results support a role for Cx43 in regulating blood-testis barrier (BTB) function, composition, and dynamics in vitro. Thus, the SC deficient Cx43 cell cultures may provide a valuable in vitro tool for a better understanding of the mechanistic role of Cx43 in spermatogenesis and BTB assembly.
Collapse
|
26
|
Nakamura T, Iwamoto T, Nakamura HM, Shindo Y, Saito K, Yamada A, Yamada Y, Fukumoto S, Nakamura T. Regulation of miR-1-Mediated Connexin 43 Expression and Cell Proliferation in Dental Epithelial Cells. Front Cell Dev Biol 2020; 8:156. [PMID: 32258035 PMCID: PMC7089876 DOI: 10.3389/fcell.2020.00156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
Many genes encoding growth factors, receptors, and transcription factors are induced by the epithelial-mesenchymal interaction during tooth development. Recently, numerous functions of microRNAs (miRNAs) are reportedly involved in organogenesis and disease. miRNAs regulate gene expression by inhibiting translation and destabilizing mRNAs. However, the expression and function of miRNAs in tooth development remain poorly understood. This study aimed to analyze the expression of miRNAs produced during tooth development using a microarray system to clarify the role of miRNAs in dental development. miR-1 showed a unique expression pattern in the developing tooth. miR-1 expression in the tooth germ peaked on embryonic day 16.5, decreasing gradually on postnatal days 1 and 3. An in situ hybridization assay revealed that miR-1 is expressed at the cervical loop of the dental epithelium. The expression of miR-1 and connexin (Cx) 43, a target of miR-1, were inversely correlated both in vitro and in vivo. Knockdown of miR-1 induced the expression of Cx43 in dental epithelial cells. Interestingly, cells with miR-1 downregulation proliferated slower than the control cells. Immunocytochemistry revealed that Cx43 in cells with miR-1 knockdown formed both cell-cell gap junctions and hemichannels at the plasma membrane. Furthermore, the rate of ATP release was higher in cells with miR-1 knockdown than in control cells. Furthermore, Cx43 downregulation in developing molars was observed in Epiprofin-knockout mice, along with the induction of miR-1 expression. These results suggest that the expression pattern of Cx43 is modulated by miR-1 to control cell proliferation activity during dental epithelial cell differentiation.
Collapse
Affiliation(s)
- Tomoaki Nakamura
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Tsutomu Iwamoto
- Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hannah M Nakamura
- Division of Nephrology and Endocrinology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yuki Shindo
- Division of Molecular Pharmacology and Cell Biophysics, Department of Oral Biology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Kan Saito
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Aya Yamada
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Yoshihiko Yamada
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, United States
| | - Satoshi Fukumoto
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Takashi Nakamura
- Division of Molecular Pharmacology and Cell Biophysics, Department of Oral Biology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
27
|
Nielsen BS, Toft-Bertelsen TL, Lolansen SD, Anderson CL, Nielsen MS, Thompson RJ, MacAulay N. Pannexin 1 activation and inhibition is permeant-selective. J Physiol 2020; 598:361-379. [PMID: 31698505 DOI: 10.1113/jp278759] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/05/2019] [Indexed: 01/14/2023] Open
Abstract
KEY POINTS The large-pore channel pannexin 1 (Panx1) is expressed in many cell types and can open upon different, yet not fully established, stimuli. Panx1 permeability is often inferred from channel permeability to fluorescent dyes, but it is currently unknown whether dye permeability translates to permeability to other molecules. Cell shrinkage and C-terminal cleavage led to a Panx1 open-state with increased permeability to atomic ions (current), but did not alter ethidium uptake. Panx1 inhibitors affected Panx1-mediated ion conduction differently from ethidium permeability, and inhibitor efficiency towards a given molecule therefore cannot be extrapolated to its effects on the permeability of another. We conclude that ethidium permeability does not reflect equal permeation of other molecules and thus is no measure of general Panx1 activity. ABSTRACT Pannexin 1 (Panx1) is a large-pore membrane channel connecting the extracellular milieu with the cell interior. While several activation regimes activate Panx1 in a variety of cell types, the selective permeability of an open Panx1 channel remains unresolved: does a given activation paradigm increase Panx1's permeability towards all permeants equally and does fluorescent dye flux serve as a proxy for biological permeation through an open channel? To explore permeant-selectivity of Panx1 activation and inhibition, we employed Panx1-expressing Xenopus laevis oocytes and HEK293T cells. We report that different mechanisms of activation of Panx1 differentially affected ethidium and atomic ion permeation. Most notably, C-terminal truncation or cell shrinkage elevated Panx1-mediated ion conductance, but had no effect on ethidium permeability. In contrast, extracellular pH changes predominantly affected ethidium permeability but not ionic conductance. High [K+ ]o did not increase the flux of either of the two permeants. Once open, Panx1 demonstrated preference for anionic permeants, such as Cl- , lactate and glutamate, while not supporting osmotic water flow. Panx1 inhibitors displayed enhanced potency towards Panx1-mediated currents compared to that of ethidium uptake. We conclude that activation or inhibition of Panx1 display permeant-selectivity and that permeation of ethidium does not necessarily reflect an equal permeation of smaller biological molecules and atomic ions.
Collapse
Affiliation(s)
- Brian Skriver Nielsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trine Lisberg Toft-Bertelsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sara Diana Lolansen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Connor L Anderson
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Morten Schak Nielsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Roger J Thompson
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Heterogeneity of Astrocytes in Grey and White Matter. Neurochem Res 2019; 46:3-14. [PMID: 31797158 DOI: 10.1007/s11064-019-02926-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/21/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023]
Abstract
Astrocytes are a diverse and heterogeneous type of glial cells. The major task of grey and white matter areas in the brain are computation of information at neuronal synapses and propagation of action potentials along axons, respectively, resulting in diverse demands for astrocytes. Adapting their function to the requirements in the local environment, astrocytes differ in morphology, gene expression, metabolism, and many other properties. Here we review the differential properties of protoplasmic astrocytes of grey matter and fibrous astrocytes located in white matter in respect to glutamate and energy metabolism, to their function at the blood-brain interface and to coupling via gap junctions. Finally, we discuss how this astrocytic heterogeneity might contribute to the different susceptibility of grey and white matter to ischemic insults.
Collapse
|
29
|
Nielsen BS, Zonta F, Farkas T, Litman T, Nielsen MS, MacAulay N. Structural determinants underlying permeant discrimination of the Cx43 hemichannel. J Biol Chem 2019; 294:16789-16803. [PMID: 31554662 DOI: 10.1074/jbc.ra119.007732] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 09/24/2019] [Indexed: 02/03/2023] Open
Abstract
Connexin (Cx) gap junction channels comprise two hemichannels in neighboring cells, and their permeability is well-described, but permeabilities of the single Cx hemichannel remain largely unresolved. Moreover, determination of isoform-specific Cx hemichannel permeability is challenging because of concurrent expression of other channels with similar permeability profiles and inhibitor sensitivities. The mammalian Cx hemichannels Cx30 and Cx43 are gated by extracellular divalent cations, removal of which promotes fluorescent dye uptake in both channels but atomic ion conductance only through Cx30. To determine the molecular determinants of this difference, here we employed chimeras and mutagenesis of predicted pore-lining residues in Cx43. We expressed the mutated channels in Xenopus laevis oocytes to avoid background activity of alternative channels. Oocytes expressing a Cx43 hemichannel chimera containing the N terminus or the first extracellular loop from Cx30 displayed ethidium uptake and, unlike WT Cx43, ion conduction, an observation further supported by molecular dynamics simulations. Additional C-terminal truncation of the chimeric Cx43 hemichannel elicited an even greater ion conductance with a magnitude closer to that of Cx30. The inhibitory profile for the connexin hemichannels depended on the permeant, with conventional connexin hemichannel inhibitors having a higher potency toward the ion conductance pathway than toward fluorescent dye uptake. Our results demonstrate a permeant-dependent, isoform-specific inhibition of connexin hemichannels. They further reveal that the outer segments of the pore-lining region, including the N terminus and the first extracellular loop, together with the C terminus preclude ion conductance of the open Cx43 hemichannel.
Collapse
Affiliation(s)
- Brian Skriver Nielsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Francesco Zonta
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Thomas Farkas
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Thomas Litman
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Morten Schak Nielsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
30
|
Liu C, Huang L, Li C, Shen Y, Wang J. [Blocking pannexin-1 alleviates cisplatin-induced acute kidney injury in mice by reducing renal inflammatory cell infiltration]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:508-514. [PMID: 31140412 DOI: 10.12122/j.issn.1673-4254.2019.05.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate the effect of blocking pannexin-1 against acute kidney injury induced by cisplatin. METHODS Twenty-six male C57BL/6 mice aged 6-8 weeks were randomly divided into control group, cisplatin model (Cis) group and cisplatin + carbenoxolone treatment group (Cis + CBX). In Cis group and Cis + CBX group, the mice were injected intraperitoneally with 20 mg/kg of cisplatin and with CBX (20 mg/kg) at 30 min before and 24 and 48 h after cisplatin inhjection, respectively. All the mice were sacrificed at 72 h after cisplatin injection, and plasma and kidney samples were collected for testing mRNA and protein expression levels of pannexin-1 in the renal tissue using RT-qPCR and Western blotting and for detecting plasma creatinine and BUN levels; the pathological changes in the renal tissues were observed using Periodic Acid-Schiff staining. The expression of kidney injury molecule 1 (KIM-1) was examined using immunohistochemistry and the mRNA expressions of KIM-1 and neutrophil gelatinase- related lipid transport protein (NGAL) were detected by RT-qPCR to evaluate the injuries of the renal tubules. The infiltration of F4/80-positive macrophages and CD4-positive T cells were observed by immunofluorescence. In the in vitro experiment, human proximal tubule epithelial cell line HK-2 was stimulated with 50 μmol/L cisplatin to establish a cell model of acute kidney injury, and the mRNA and protein expressions of pannexin-1 were detected by RT-qPCR and Western blotting at 4, 6, 12, 18 and 24 h after the stimulation. RESULTS Compared with the control mice, the cisplatin-treated mice showed significantly up-regulated protein levels (P < 0.05) and mRNA levels (P < 0.005) of pannexin-1 in the kidney tissue. Cisplatin stimulation also caused significant increases in the protein levels (P < 0.005) and mRNA levels (P < 0.005) of pannexin-1 in cultured HK-2 cells. Compared with cisplatin-treated mice, the mice treated with both cisplatin and the pannexin-1 inhibitor CBX showed obviously lessened kidney pathologies and milder renal tubular injuries with significantly reduced plasma BUN and Scr levels (P < 0.01), expressions of KIM-1 and NGAL in the kidney (P < 0.05), and infiltration of F4/80-positive macrophages (P < 0.01) and CD4- positive T cells (P < 0.05) in the kidney tissues. CONCLUSIONS In cisplatin induced acute kidney injury mice model, Pannexin-1 expression is up-regulated in the kidneys tissue, and blocking pannexin-1 alleviates the acute kidney injury via reducing renal inflammatory cell infiltration.
Collapse
Affiliation(s)
- Chongbin Liu
- National Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Liuwei Huang
- National Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Caizhen Li
- National Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanting Shen
- National Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jun Wang
- National Clinical Research Center of Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
31
|
Dispelling myths about connexins, pannexins and P2X7 in hypoxic-ischemic central nervous system. Neurosci Lett 2019; 695:76-85. [PMID: 29195910 DOI: 10.1016/j.neulet.2017.11.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 10/07/2017] [Accepted: 11/21/2017] [Indexed: 01/17/2023]
Abstract
In membrane physiology, as in other fields, myths or speculations may be repeated so often and so widely that they are perceived as facts. To some extent, this has occurred with regard to gap junctions, hemichannels, pannexin channels and P2X7 (ionotropic receptors), especially concerning the interpretation of the individual role of these channels in hypoxic-ischemic CNS since these channels may be closed by the same pharmacological blockers. Significance of existing controversial data are highlighted and contradictory views from different groups are critically discussed herein.
Collapse
|
32
|
Spray DC, Iglesias R, Shraer N, Suadicani SO, Belzer V, Hanstein R, Hanani M. Gap junction mediated signaling between satellite glia and neurons in trigeminal ganglia. Glia 2019; 67:791-801. [PMID: 30715764 DOI: 10.1002/glia.23554] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/28/2018] [Accepted: 10/03/2018] [Indexed: 01/07/2023]
Abstract
Peripheral sensory ganglia contain the somata of neurons mediating mechanical, thermal, and painful sensations from somatic, visceral, and oro-facial organs. Each neuronal cell body is closely surrounded by satellite glial cells (SGCs) that have properties and functions similar to those of central astrocytes, including expression of gap junction proteins and functional dye coupling. As shown in other pain models, after systemic pain induction by intra-peritoneal injection of lipopolysaccharide, dye coupling among SGCs in intact trigeminal ganglion was enhanced. Moreover, neuron-neuron and neuron-SGC coupling was also detected. To verify the presence of gap junction-mediated coupling between SGCs and sensory neurons, we performed dual whole cell patch clamp recordings from both freshly isolated and short term cultured cell pairs dissociated from mouse trigeminal ganglia. Bidirectional gap junction mediated electrical responses were frequently recorded between SGCs, between neurons and between neurons and SGCs. Polarization of SGC altered neuronal excitability, providing evidence that gap junction-mediated interactions between neurons and glia within sensory ganglia may contribute to integration of peripheral sensory responses, and to the modulation and coordinaton of neuronal activity.
Collapse
Affiliation(s)
- David C Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| | - Rodolfo Iglesias
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| | - Nathanael Shraer
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| | - Sylvia O Suadicani
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York.,Department of Urology, Albert Einstein College of Medicine, Bronx, New York
| | - Vitali Belzer
- Laboratory of Experimental Surgery, Department of Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Regina Hanstein
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| | - Menachem Hanani
- Laboratory of Experimental Surgery, Department of Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
33
|
Abudara V, Retamal MA, Del Rio R, Orellana JA. Synaptic Functions of Hemichannels and Pannexons: A Double-Edged Sword. Front Mol Neurosci 2018; 11:435. [PMID: 30564096 PMCID: PMC6288452 DOI: 10.3389/fnmol.2018.00435] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/08/2018] [Indexed: 01/18/2023] Open
Abstract
The classical view of synapses as the functional contact between presynaptic and postsynaptic neurons has been challenged in recent years by the emerging regulatory role of glial cells. Astrocytes, traditionally considered merely supportive elements are now recognized as active modulators of synaptic transmission and plasticity at the now so-called "tripartite synapse." In addition, an increasing body of evidence indicates that beyond immune functions microglia also participate in various processes aimed to shape synaptic plasticity. Release of neuroactive compounds of glial origin, -process known as gliotransmission-, constitute a widespread mechanism through which glial cells can either potentiate or reduce the synaptic strength. The prevailing vision states that gliotransmission depends on an intracellular Ca2+/exocytotic-mediated release; notwithstanding, growing evidence is pointing at hemichannels (connexons) and pannexin channels (pannexons) as alternative non-vesicular routes for gliotransmitters efflux. In concurrence with this novel concept, both hemichannels and pannexons are known to mediate the transfer of ions and signaling molecules -such as ATP and glutamate- between the cytoplasm and the extracellular milieu. Importantly, recent reports show that glial hemichannels and pannexons are capable to perceive synaptic activity and to respond to it through changes in their functional state. In this article, we will review the current information supporting the "double edge sword" role of hemichannels and pannexons in the function of central and peripheral synapses. At one end, available data support the idea that these channels are chief components of a feedback control mechanism through which gliotransmitters adjust the synaptic gain in either resting or stimulated conditions. At the other end, we will discuss how the excitotoxic release of gliotransmitters and [Ca2+]i overload linked to the opening of hemichannels/pannexons might impact cell function and survival in the nervous system.
Collapse
Affiliation(s)
- Verónica Abudara
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.,Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Santiago, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Envejecimiento y Regeneración, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes, Universidad de Magallanes, Punta Arenas, Chile
| | - Juan A Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes, Santiago, Chile
| |
Collapse
|
34
|
Watanabe N, Sasaki S, Masamoto K, Hotta H. Vascular Gap Junctions Contribute to Forepaw Stimulation-Induced Vasodilation Differentially in the Pial and Penetrating Arteries in Isoflurane-Anesthetized Rats. Front Mol Neurosci 2018; 11:446. [PMID: 30559647 PMCID: PMC6286957 DOI: 10.3389/fnmol.2018.00446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/19/2018] [Indexed: 12/25/2022] Open
Abstract
Somatosensory stimulation causes dilation of the pial and penetrating arteries and an increase in cerebral blood flow (CBF) in the representative region of the somatosensory cortex. As an underlying mechanism for such stimulation-induced increases in CBF, cerebral artery dilation has been thought to propagate in the vascular endothelium from the parenchyma to the brain surface. Vascular gap junctions may propagate vasodilation. However, the contribution of vascular gap junctions to cerebrovascular regulation induced by somatosensory stimulation is largely unknown. The aim of the present study was to investigate the contribution of vascular gap junctions to the regulation of the pial and penetrating arteries during neuronal activity attributed to somatosensory stimulation. Experiments were performed on male Wistar rats (age: 7-10 weeks) with artificial ventilation under isoflurane anesthesia. For somatosensory stimulation, the left forepaw was electrically stimulated (1.5 mA, 0.5 ms and 10 Hz, for 5 s). The artery in the forelimb area of the right somatosensory cortex was imaged through a cranial window using a two-photon microscope and the diameter was measured. Carbenoxolone (CBX) was intravenously (i.v.) administered, at a dose of 100 mg/kg, to block vascular gap junctions. The forepaw electrical stimulation increased the diameter of the pial and penetrating arteries by 7.0% and 5.0% of the pre-stimulus diameter, respectively, without changing the arterial pressure. After CBX administration, the change in pial artery diameter during forepaw stimulation was attenuated to 3.2%. However, changes in the penetrating artery were not significantly affected. CBF was measured using a laser speckle flowmeter, together with somatosensory-evoked potential (SEP) recorded in the somatosensory cortex. The extent of CBF increase (by 24.1% of the pre-stimulus level) and amplitude of SEP were not affected by CBX administration. The present results suggest that vascular gap junctions, possibly on the endothelium, contribute to pial artery dilation during neuronal activity induced by somatosensory stimulation.
Collapse
Affiliation(s)
- Nobuhiro Watanabe
- Department of Autonomic Neuroscience, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Satoshi Sasaki
- Department of Autonomic Neuroscience, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan
| | - Kazuto Masamoto
- Department of Autonomic Neuroscience, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan.,Brain Science Inspired Life Support Research Center, The University of Electro-Communications, Tokyo, Japan
| | - Harumi Hotta
- Department of Autonomic Neuroscience, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
35
|
Loss of connexin 43 in Sertoli cells provokes postnatal spermatogonial arrest, reduced germ cell numbers and impaired spermatogenesis. Reprod Biol 2018; 18:456-466. [PMID: 30243528 DOI: 10.1016/j.repbio.2018.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 07/31/2018] [Accepted: 08/20/2018] [Indexed: 01/27/2023]
Abstract
For the reason that adult Sertoli cell specific connexin 43 knockout (SCCx43KO) mice show arrested spermatogenesis at spermatogonial level or Sertoli cell only tubules and significantly reduced germ cell (GC) numbers, the aims of the present study were (1) to characterize the remaining GC population and (2) to elucidate possible mechanisms of their fading. Apoptosis was analyzed in both, KO and wild type (WT) male littermates during postnatal development and in adulthood using TUNEL. Although GC numbers were significantly reduced in KO at 2 and 8 days postpartum (dpp) when compared to WT, no differences were found concerning apoptotic incidence between genotypes. From 10 dpp, the substantial GC deficiency became more obvious. However, significantly higher apoptotic GC numbers were seen in WT during this period, possibly related to the first wave of spermatogenesis, a known phenomenon in normal pubertal testes associated with increased apoptosis. Characterization of residual spermatogonia in postnatal to adult KO and WT mice was performed by immunohistochemical reaction against VASA (marker of GCs in general), Lin28 and Fox01 (markers for undifferentiated spermatogonia) and Stra8 (marker for differentiating spermatogonia and early spermatocytes). During puberty, the GC component in SCCx43KO mice consisted likely of undifferentiated spermatogonia, few differentiating spermatogonia and very few early spermatocytes, which seemed to be rapidly cleared by apoptosis. In adult KOs, spermatogenesis was arrested at the level of undifferentiated spermatogonia. Overall, our data indicate that Cx43 gap junctions in SCs influence male GC development and differentiation rather than their survival.
Collapse
|
36
|
Cerebrospinal Fluid-Contacting Neurons Sense pH Changes and Motion in the Hypothalamus. J Neurosci 2018; 38:7713-7724. [PMID: 30037834 DOI: 10.1523/jneurosci.3359-17.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 07/04/2018] [Accepted: 07/15/2018] [Indexed: 01/20/2023] Open
Abstract
CSF-contacting (CSF-c) cells are present in the walls of the brain ventricles and the central canal of the spinal cord and found throughout the vertebrate phylum. We recently identified ciliated somatostatin-/GABA-expressing CSF-c neurons in the lamprey spinal cord that act as pH sensors as well as mechanoreceptors. In the same neuron, acidic and alkaline responses are mediated through ASIC3-like and PKD2L1 channels, respectively. Here, we investigate the functional properties of the ciliated somatostatin-/GABA-positive CSF-c neurons in the hypothalamus by performing whole-cell recordings in hypothalamic slices. Depolarizing current pulses readily evoked action potentials, but hypothalamic CSF-c neurons had no or a very low level of spontaneous activity at pH 7.4. They responded, however, with membrane potential depolarization and trains of action potentials to small deviations in pH in both the acidic and alkaline direction. Like in spinal CSF-c neurons, the acidic response in hypothalamic cells is mediated via ASIC3-like channels. In contrast, the alkaline response appears to depend on connexin hemichannels, not on PKD2L1 channels. We also show that hypothalamic CSF-c neurons respond to mechanical stimulation induced by fluid movements along the wall of the third ventricle, a response mediated via ASIC3-like channels. The hypothalamic CSF-c neurons extend their processes dorsally, ventrally, and laterally, but as yet, the effects exerted on hypothalamic circuits are unknown. With similar neurons being present in rodents, the pH- and mechanosensing ability of hypothalamic CSF-c neurons is most likely conserved throughout vertebrate phylogeny.SIGNIFICANCE STATEMENT CSF-contacting neurons are present in all vertebrates and are located mainly in the hypothalamic area and the spinal cord. Here, we report that the somatostatin-/GABA-expressing CSF-c neurons in the lamprey hypothalamus sense bidirectional deviations in the extracellular pH and do so via different molecular mechanisms. They also serve as mechanoreceptors. The hypothalamic CSF-c neurons have extensive axonal ramifications and may decrease the level of motor activity via release of somatostatin. In conclusion, hypothalamic somatostatin-/GABA-expressing CSF-c neurons, as well as their spinal counterpart, represent a novel homeostatic mechanism designed to sense any deviation from physiological pH and thus constitute a feedback regulatory system intrinsic to the CNS, possibly serving a protective role from damage caused by changes in pH.
Collapse
|
37
|
Vicario N, Zappalà A, Calabrese G, Gulino R, Parenti C, Gulisano M, Parenti R. Connexins in the Central Nervous System: Physiological Traits and Neuroprotective Targets. Front Physiol 2017; 8:1060. [PMID: 29326598 PMCID: PMC5741605 DOI: 10.3389/fphys.2017.01060] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/04/2017] [Indexed: 01/22/2023] Open
Abstract
Cell-to-cell interaction and cell-to-extracellular environment communication are emerging as new therapeutic targets in neurodegenerative disorders. Dynamic expression of connexins leads to distinctive hemichannels and gap junctions, characterized by cell-specific conduction, exchange of stimuli or metabolites, and particular channel functions. Herein, we briefly reviewed classical physiological traits and functions of connexins, hemichannels, and gap junctions, in order to discuss the controversial role of these proteins and their mediated interactions during neuroprotection, with a particular focus on Cx43-based channels. We pointed out the contribution of connexins in neural cells populations during neurodegenerative processes to explore potential neuroprotective therapeutic applications.
Collapse
Affiliation(s)
- Nunzio Vicario
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Agata Zappalà
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giovanna Calabrese
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosario Gulino
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Carmela Parenti
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Massimo Gulisano
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Department of Drug Sciences, University of Catania, Catania, Italy
| | - Rosalba Parenti
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
38
|
Leybaert L, Lampe PD, Dhein S, Kwak BR, Ferdinandy P, Beyer EC, Laird DW, Naus CC, Green CR, Schulz R. Connexins in Cardiovascular and Neurovascular Health and Disease: Pharmacological Implications. Pharmacol Rev 2017; 69:396-478. [PMID: 28931622 PMCID: PMC5612248 DOI: 10.1124/pr.115.012062] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Connexins are ubiquitous channel forming proteins that assemble as plasma membrane hemichannels and as intercellular gap junction channels that directly connect cells. In the heart, gap junction channels electrically connect myocytes and specialized conductive tissues to coordinate the atrial and ventricular contraction/relaxation cycles and pump function. In blood vessels, these channels facilitate long-distance endothelial cell communication, synchronize smooth muscle cell contraction, and support endothelial-smooth muscle cell communication. In the central nervous system they form cellular syncytia and coordinate neural function. Gap junction channels are normally open and hemichannels are normally closed, but pathologic conditions may restrict gap junction communication and promote hemichannel opening, thereby disturbing a delicate cellular communication balance. Until recently, most connexin-targeting agents exhibited little specificity and several off-target effects. Recent work with peptide-based approaches has demonstrated improved specificity and opened avenues for a more rational approach toward independently modulating the function of gap junctions and hemichannels. We here review the role of connexins and their channels in cardiovascular and neurovascular health and disease, focusing on crucial regulatory aspects and identification of potential targets to modify their function. We conclude that peptide-based investigations have raised several new opportunities for interfering with connexins and their channels that may soon allow preservation of gap junction communication, inhibition of hemichannel opening, and mitigation of inflammatory signaling.
Collapse
Affiliation(s)
- Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Paul D Lampe
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Stefan Dhein
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Brenda R Kwak
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Peter Ferdinandy
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Eric C Beyer
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Dale W Laird
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Christian C Naus
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Colin R Green
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| |
Collapse
|
39
|
Role of Connexin and Pannexin containing channels in HIV infection and NeuroAIDS. Neurosci Lett 2017; 695:86-90. [PMID: 28886986 DOI: 10.1016/j.neulet.2017.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 07/27/2017] [Accepted: 09/01/2017] [Indexed: 01/31/2023]
Abstract
Neuron-Glia crosstalk is essential for efficient synaptic communication, cell growth and differentiation, neuronal activity, neurotransmitter recycling, and brain immune response. The master regulators of this neuron-glia communication are connexin containing Gap Junctions (GJs) and Hemichannels (HCs) as well as pannexin HCs. However, the role of these channels under pathological conditions, especially in infectious diseases is still in exploratory stages. Human Immunodeficiency Virus-1 (HIV) is one such infectious agent that takes advantage of the host intercellular communication systems, GJs and HCs, to exacerbate viral pathogenesis in the brain in spite of the antiretroviral therapy effectively controlling viral replication in the periphery. Although most infectious agents lead to total "shutdown" of gap junctional communication in parenchymal cells, HIV infection maintains and "hijacks" GJs and HCs to enable few infected cells to spread toxic intracellular agents to neighboring uninfected cells aggravating viral neuropathology even in the absence of viral replication. In this mini-review, we present a comprehensive overview of the role of GJs and HCs in augmenting HIV neuropathogenesis.
Collapse
|
40
|
Maes M, Crespo Yanguas S, Willebrords J, Weemhoff JL, da Silva TC, Decrock E, Lebofsky M, Pereira IVA, Leybaert L, Farhood A, Jaeschke H, Cogliati B, Vinken M. Connexin hemichannel inhibition reduces acetaminophen-induced liver injury in mice. Toxicol Lett 2017; 278:30-37. [PMID: 28687253 PMCID: PMC5800489 DOI: 10.1016/j.toxlet.2017.07.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/27/2017] [Accepted: 07/01/2017] [Indexed: 02/07/2023]
Abstract
Historically, connexin hemichannels have been considered as structural precursors of gap junctions. However, accumulating evidence points to independent roles for connexin hemichannels in cellular signaling by connecting the intracellular compartment with the extracellular environment. Unlike gap junctions, connexin hemichannels seem to be mainly activated in pathological processes. The present study was set up to test the potential involvement of hemichannels composed of connexin32 and connexin43 in acute hepatotoxicity induced by acetaminophen. Prior to this, in vitro testing was performed to confirm the specificity and efficacy of TAT-Gap24 and TAT-Gap19 in blocking connexin32 and connexin43 hemichannels, respectively. Subsequently, mice were overdosed with acetaminophen followed by treatment with TAT-Gap24 or TAT-Gap19 or a combination of both after 1.5h. Sampling was performed 3, 6, 24 and 48h following acetaminophen administration. Evaluation of the effects of connexin hemichannel inhibition was based on a series of clinically relevant read-outs, measurement of inflammatory cytokines and oxidative stress. Subsequent treatment of acetaminophen-overdosed mice with TAT-Gap19 only marginally affected liver injury. In contrast, a significant reduction in serum alanine aminotransferase activity was found upon administration of TAT-Gap24 to intoxicated animals. Furthermore, co-treatment of acetaminophen-overdosed mice with both peptides revealed an additive effect as even lower serum alanine aminotransferase activity was observed. Blocking of connexin32 or connexin43 hemichannels individually was found to decrease serum quantities of pro-inflammatory cytokines, while no effects were observed on the occurrence of hepatic oxidative stress. This study shows for the first time a role for connexin hemichannels in acetaminophen-induced acute liver failure.
Collapse
Affiliation(s)
- Michaël Maes
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Sara Crespo Yanguas
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Joost Willebrords
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium.
| | - James L Weemhoff
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, United States.
| | - Tereza Cristina da Silva
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.
| | - Elke Decrock
- Department of Basic Medical Sciences, Physiology Group, Ghent University, Ghent, Belgium.
| | - Margitta Lebofsky
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, United States.
| | - Isabel Veloso Alves Pereira
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.
| | - Luc Leybaert
- Department of Basic Medical Sciences, Physiology Group, Ghent University, Ghent, Belgium.
| | - Anwar Farhood
- Department of Pathology, St. David's North Austin Medical Center, Austin, United States.
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, United States.
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
41
|
Forsberg D, Thonabulsombat C, Jäderstad J, Jäderstad LM, Olivius P, Herlenius E. Functional Stem Cell Integration into Neural Networks Assessed by Organotypic Slice Cultures. ACTA ACUST UNITED AC 2017; 42:2D.13.1-2D.13.30. [PMID: 28806855 DOI: 10.1002/cpsc.34] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Re-formation or preservation of functional, electrically active neural networks has been proffered as one of the goals of stem cell-mediated neural therapeutics. A primary issue for a cell therapy approach is the formation of functional contacts between the implanted cells and the host tissue. Therefore, it is of fundamental interest to establish protocols that allow us to delineate a detailed time course of grafted stem cell survival, migration, differentiation, integration, and functional interaction with the host. One option for in vitro studies is to examine the integration of exogenous stem cells into an existing active neural network in ex vivo organotypic cultures. Organotypic cultures leave the structural integrity essentially intact while still allowing the microenvironment to be carefully controlled. This allows detailed studies over time of cellular responses and cell-cell interactions, which are not readily performed in vivo. This unit describes procedures for using organotypic slice cultures as ex vivo model systems for studying neural stem cell and embryonic stem cell engraftment and communication with CNS host tissue. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- David Forsberg
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Charoensri Thonabulsombat
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Section of Otorhinolaryngology, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden.,Center for Hearing and Communication Research, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden.,Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Johan Jäderstad
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Linda Maria Jäderstad
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Petri Olivius
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Section of Otorhinolaryngology, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden.,Center for Hearing and Communication Research, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Eric Herlenius
- Department of Women's and Children's Health, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
42
|
Abstract
ATP, the energy exchange factor that connects anabolism and catabolism, is required for major reactions and processes that occur in living cells, such as muscle contraction, phosphorylation and active transport. ATP is also the key molecule in extracellular purinergic signaling mechanisms, with an established crucial role in inflammation and several additional disease conditions. Here, we describe detailed protocols to measure the ATP concentration in isolated living cells and animals using luminescence techniques based on targeted luciferase probes. In the presence of magnesium, oxygen and ATP, the protein luciferase catalyzes oxidation of the substrate luciferin, which is associated with light emission. Recombinantly expressed wild-type luciferase is exclusively cytosolic; however, adding specific targeting sequences can modify its cellular localization. Using this strategy, we have constructed luciferase chimeras targeted to the mitochondrial matrix and the outer surface of the plasma membrane. Here, we describe optimized protocols for monitoring ATP concentrations in the cytosol, mitochondrial matrix and pericellular space in living cells via an overall procedure that requires an average of 3 d. In addition, we present a detailed protocol for the in vivo detection of extracellular ATP in mice using luciferase-transfected reporter cells. This latter procedure may require up to 25 d to complete.
Collapse
|
43
|
Vicario N, Calabrese G, Zappalà A, Parenti C, Forte S, Graziano ACE, Vanella L, Pellitteri R, Cardile V, Parenti R. Inhibition of Cx43 mediates protective effects on hypoxic/reoxygenated human neuroblastoma cells. J Cell Mol Med 2017; 21:2563-2572. [PMID: 28488330 PMCID: PMC5618696 DOI: 10.1111/jcmm.13177] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/28/2017] [Indexed: 12/18/2022] Open
Abstract
Olfactory ensheathing cells (OECs), a special population of glial cells, are able to synthesise several trophic factors exerting a neuroprotective action and promoting growth and functional recovery in both in vitro and in vivo models. In the present work, we investigated the neuroprotective effects of OEC-conditioned medium (OEC-CM) on two different human neuron-like cell lines, SH-SY5Y and SK-N-SH (neuroblastoma cell lines), under normoxic and hypoxic conditions. In addition, we also focused our attention on the role of connexins (Cxs) in the neuroprotective processes. Our results confirmed OEC-CM mediated neuroprotection as shown by cell adherence, proliferation and cellular viability analyses. Reduced connexin 43 (Cx43) levels in OEC-CM compared to unconditioned cells in hypoxic conditions prompted us to investigate the role of Cx43-Gap junctions (GJs) and Cx43-hemichannels (HCs) in hypoxic/reoxygenation injury using carbenoxolone (non-selective GJ inhibitor), ioxynil octanoato (selective Cx43-GJ inhibitor) and Gap19 (selective Cx43-HC inhibitor). We found that Cx43-GJ and Cx43-HC inhibitors are able to protect SH-SY5Y and allow to these cultures to overcome the injury. Our findings support the hypothesis that both OEC-CM and the inhibition of Cx43-GJs and Cx43-HCs offer a neuroprotective effect by reducing Cx43-mediated cell-to-cell and cell-to-extracellular environment communications.
Collapse
Affiliation(s)
- Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania, Italy
| | - Giovanna Calabrese
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania, Italy
| | - Agata Zappalà
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania, Italy
| | - Carmela Parenti
- Department of Drug Sciences, University of Catania, Catania, Italy
| | | | | | - Luca Vanella
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Rosalia Pellitteri
- Institute Neurological Sciences, National Research Council, Catania, Italy
| | - Venera Cardile
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, Catania, Italy
| |
Collapse
|
44
|
White JPM, Cibelli M, Urban L, Nilius B, McGeown JG, Nagy I. TRPV4: Molecular Conductor of a Diverse Orchestra. Physiol Rev 2017; 96:911-73. [PMID: 27252279 DOI: 10.1152/physrev.00016.2015] [Citation(s) in RCA: 277] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Transient receptor potential vanilloid type 4 (TRPV4) is a calcium-permeable nonselective cation channel, originally described in 2000 by research teams led by Schultz (Nat Cell Biol 2: 695-702, 2000) and Liedtke (Cell 103: 525-535, 2000). TRPV4 is now recognized as being a polymodal ionotropic receptor that is activated by a disparate array of stimuli, ranging from hypotonicity to heat and acidic pH. Importantly, this ion channel is constitutively expressed and capable of spontaneous activity in the absence of agonist stimulation, which suggests that it serves important physiological functions, as does its widespread dissemination throughout the body and its capacity to interact with other proteins. Not surprisingly, therefore, it has emerged more recently that TRPV4 fulfills a great number of important physiological roles and that various disease states are attributable to the absence, or abnormal functioning, of this ion channel. Here, we review the known characteristics of this ion channel's structure, localization and function, including its activators, and examine its functional importance in health and disease.
Collapse
Affiliation(s)
- John P M White
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Mario Cibelli
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Laszlo Urban
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Bernd Nilius
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - J Graham McGeown
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Istvan Nagy
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, London, United Kingdom; Department of Anaesthetics, The Queen Elizabeth Hospital, Birmingham, United Kingdom; Academic Department of Anaesthesia and Intensive Care Medicine, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, United Kingdom; Preclinical Secondary Pharmacology, Preclinical Safety, Novartis Institute for Biomedical Research, Cambridge, Massachusetts; Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg, Leuven, Belgium; and School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
45
|
Sword J, Croom D, Wang PL, Thompson RJ, Kirov SA. Neuronal pannexin-1 channels are not molecular routes of water influx during spreading depolarization-induced dendritic beading. J Cereb Blood Flow Metab 2017; 37:1626-1633. [PMID: 26994044 PMCID: PMC5435276 DOI: 10.1177/0271678x16639328] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Spreading depolarization-induced focal dendritic swelling (beading) is an early hallmark of neuronal cytotoxic edema. Pyramidal neurons lack membrane-bound aquaporins posing a question of how water enters neurons during spreading depolarization. Recently, we have identified chloride-coupled transport mechanisms that can, at least in part, participate in dendritic beading. Yet transporter-mediated ion and water fluxes could be paralleled by water entry through additional pathways such as large-pore pannexin-1 channels opened by spreading depolarization. Using real-time in vivo two-photon imaging in mice with pharmacological inhibition or conditional genetic deletion of pannexin-1, we showed that pannexin-1 channels are not required for spreading depolarization-induced focal dendritic swelling.
Collapse
Affiliation(s)
- Jeremy Sword
- 1 Brain and Behavior Discovery Institute, Medical College of Georgia, Augusta, GA, USA
| | - Deborah Croom
- 1 Brain and Behavior Discovery Institute, Medical College of Georgia, Augusta, GA, USA
| | - Phil L Wang
- 1 Brain and Behavior Discovery Institute, Medical College of Georgia, Augusta, GA, USA
| | - Roger J Thompson
- 2 Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - Sergei A Kirov
- 1 Brain and Behavior Discovery Institute, Medical College of Georgia, Augusta, GA, USA.,3 Department of Neurosurgery, Medical College of Georgia, Augusta, GA, USA
| |
Collapse
|
46
|
Cell culture: complications due to mechanical release of ATP and activation of purinoceptors. Cell Tissue Res 2017; 370:1-11. [PMID: 28434079 PMCID: PMC5610203 DOI: 10.1007/s00441-017-2618-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/21/2017] [Indexed: 12/11/2022]
Abstract
There is abundant evidence that ATP (adenosine 5′-triphosphate) is released from a variety of cultured cells in response to mechanical stimulation. The release mechanism involved appears to be a combination of vesicular exocytosis and connexin and pannexin hemichannels. Purinergic receptors on cultured cells mediate both short-term purinergic signalling of secretion and long-term (trophic) signalling such as proliferation, migration, differentiation and apoptosis. We aim in this review to bring to the attention of non-purinergic researchers using tissue culture that the release of ATP in response to mechanical stress evoked by the unavoidable movement of the cells acting on functional purinergic receptors on the culture cells is likely to complicate the interpretation of their data.
Collapse
|
47
|
Nielsen BS, Hansen DB, Ransom BR, Nielsen MS, MacAulay N. Connexin Hemichannels in Astrocytes: An Assessment of Controversies Regarding Their Functional Characteristics. Neurochem Res 2017; 42:2537-2550. [DOI: 10.1007/s11064-017-2243-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/17/2017] [Accepted: 03/17/2017] [Indexed: 12/19/2022]
|
48
|
Meda P. Gap junction proteins are key drivers of endocrine function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:124-140. [PMID: 28284720 DOI: 10.1016/j.bbamem.2017.03.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 01/07/2023]
Abstract
It has long been known that the main secretory cells of exocrine and endocrine glands are connected by gap junctions, made by a variety of connexin species that ensure their electrical and metabolic coupling. Experiments in culture systems and animal models have since provided increasing evidence that connexin signaling contributes to control the biosynthesis and release of secretory products, as well as to the life and death of secretory cells. More recently, genetic studies have further provided the first lines of evidence that connexins also control the function of human glands, which are central to the pathogenesis of major endocrine diseases. Here, we summarize the recent information gathered on connexin signaling in these systems, since the last reviews on the topic, with particular regard to the pancreatic beta cells which produce insulin, and the renal cells which produce renin. These cells are keys to the development of various forms of diabetes and hypertension, respectively, and combine to account for the exploding, worldwide prevalence of the metabolic syndrome. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Switzerland.
| |
Collapse
|
49
|
Abstract
Being critical mediators of liver homeostasis, connexins and their channels are frequently involved in liver toxicity. In the current paper, specific attention is paid to actions of hepatotoxic drugs on these communicative structures. In a first part, an overview is provided on the structural, regulatory and functional properties of connexin-based channels in the liver. In the second part, documented effects of acetaminophen, hypolipidemic drugs, phenobarbital and methapyriline on connexin signaling are discussed. Furthermore, the relevance of this subject for the fields of clinical and in vitro toxicology is demonstrated. Relevance for patients: The role of connexin signaling in drug-induced hepatotoxicity may be of high clinical relevance, as it offers perspectives for the therapeutic treatment of such insults by interfering with connexin channel opening.
Collapse
Affiliation(s)
- Michaël Maes
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
50
|
Johnson RG, Le HC, Evenson K, Loberg SW, Myslajek TM, Prabhu A, Manley AM, O’Shea C, Grunenwald H, Haddican M, Fitzgerald PM, Robinson T, Cisterna BA, Sáez JC, Liu TF, Laird DW, Sheridan JD. Connexin Hemichannels: Methods for Dye Uptake and Leakage. J Membr Biol 2016; 249:713-741. [DOI: 10.1007/s00232-016-9925-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/22/2016] [Indexed: 01/18/2023]
|