1
|
Harrell CR, Volarevic V. Ion Channels as Potential Drug Targets in Dry Eye Disease and Their Clinical Relevance: A Review. Cells 2024; 13:2017. [PMID: 39682765 PMCID: PMC11639998 DOI: 10.3390/cells13232017] [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: 07/18/2024] [Revised: 10/17/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Dry eye disease (DED) is a common multifactorial disorder characterized by a deficiency in the quality and/or quantity of tear fluid. Tear hyperosmolarity, the dysfunction of ion channel proteins, and eye inflammation are primarily responsible for the development and progression of DED. Alterations in the structure and/or function of ion channel receptors (transient receptor potential ankyrin 1 (TRPA1), transient receptor potential melastatin 8 (TRPM8), transient receptor potential vanilloid 1 and 4 (TRPV1 and TRPV4)), and consequent hyperosmolarity of the tears represent the initial step in the development and progression of DED. Hyperosmolarity triggers the activation of ion channel-dependent signaling pathways in corneal epithelial cells and eye-infiltrated immune cells, leading to the activation of transcriptional factors that enhance the expression of genes regulating inflammatory cytokine production, resulting in a potent inflammatory response in the eyes of DED patients. A persistent and untreated detrimental immune response further modifies the structure and function of ion channel proteins, perpetuating tear hyperosmolarity and exacerbating DED symptoms. Accordingly, suppressing immune cell-driven eye inflammation and alleviating tear hyperosmolarity through the modulation of ion channels in DED patients holds promise for developing new therapeutic strategies. Here, we summarize current knowledge about the molecular mechanisms responsible for the inflammation-induced modification of ion channels leading to tear hyperosmolarity and immune cell dysfunction in DED patients. We also emphasize the therapeutic potential of the newly designed immunomodulatory and hypo-osmotic solution d-MAPPS™ Hypo-Osmotic Ophthalmic Solution, which can activate TRPV4 in corneal epithelial cells, stabilize the tear film, enhance natural cytokine communication, and suppress detrimental immune responses, an important novel approach for DED treatment.
Collapse
Affiliation(s)
| | - Vladislav Volarevic
- Center for Research on Harmful Effects of Biological and Chemical Hazards, Departments of Genetics, Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia;
- Faculty of Pharmacy Novi Sad, Heroja Pinkija 4, 21000 Novi Sad, Serbia
| |
Collapse
|
2
|
Wang W, Meng Y, Yin X, Zhao P, Wang M, Ren J, Zhang J, Zhang L, Cui Y, Xia X. Novel heterologously expressed protein, AjPSPLP-3, derived from Apostichopus japonicus exhibits cell proliferation and migration activities. Protein Expr Purif 2024; 224:106577. [PMID: 39153562 DOI: 10.1016/j.pep.2024.106577] [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: 06/27/2024] [Revised: 08/04/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
Developing more effective bioactive ingredients of natural origin is imperative for promoting wound healing. Sea cucumbers have long enjoyed a good reputation as both food delicacies and traditional medicines. In this study, we heterogeneously expressed a Apostichopus japonicus derived novel protein AjPSPLP-3, which exhibits a theoretical molecular weight of 13.034 kDa, through fusion with maltose binding protein (MBP). AjPSPLP-3 contains a strict CXXCXC motif, nine extremely conserved cysteine residues and two highly conserved cysteine residues. The predicted structure of AjPSPLP-3 consists of random coil and nine β-sheets, Cys30-Cys67, Cys38-Cys58, Cys53-Cys90, Cys56-Cys66, and Cys81-Cys102 participating in the formation of five pairs of disulfide bonds. In vitro experiments conducted on HaCaT cells proved that AjPSPLP-3 and MBP-fused AjPSPLP-3 significantly contribute to HaCaT cells proliferation and migration without exhibiting hemolytic activity on murine erythrocytes. Specifically, treatment with 10 μmol/L MBP-fused AjPSPLP-3 protein increased the viability of HaCaT cells by 12.28 % (p < 0.001), while treatment with 10 μmol/L AjPSPLP-3 protein increased viability of HaCaT cells by 6.01 % (p < 0.01). Furthermore, wound closure of MBP-fused AjPSPLP-3 and AjPSPLP-3 were 22.51 % (p < 0.01) and 7.32 % (p < 0.05) higher than that of the control groups in HaCaT cells following 24 h of incubation.
Collapse
Affiliation(s)
- Weitao Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250013, China
| | - Yiwei Meng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250013, China
| | - Xin Yin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250013, China
| | - Peipei Zhao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250013, China
| | - Mengmeng Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250013, China
| | - Jingli Ren
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250013, China
| | - Jiyuan Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250013, China
| | - Lixin Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250013, China; State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Yunqian Cui
- School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| | - Xuekui Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250013, China.
| |
Collapse
|
3
|
Gao YJ, Wang YC, Zhao DL, Wen Q, Shi HX, Wang SR. A review of electroacupuncture in bone repair: Mechanisms and clinical implications. Medicine (Baltimore) 2024; 103:e40725. [PMID: 39809159 PMCID: PMC11596701 DOI: 10.1097/md.0000000000040725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 11/08/2024] [Indexed: 01/16/2025] Open
Abstract
The journey of bone repair is a lengthy process. Traditionally, oral or topical medications have been employed to facilitate healing, approaches that are not only costly but may also lead to adverse effects such as gastrointestinal damage. With advancements in electrophysiology, the significance of bioelectric activity in tissue repair has become increasingly prominent, thereby enhancing the focus on research into electroacupuncture (EA) for bone repair. EA, a synthesis of traditional acupuncture and electrical stimulation, can regulate pain by inhibiting the transmission of electrical signals, reducing the expression of ion channel proteins, and promoting the release of neurotransmitters at targeted sites. Moreover, EA has the capability to influence macrophage polarization and modulate inflammatory cytokines, aiding in bone repair. Additionally, EA has the potential to regulate cytokines such as Ephrin type-B receptor 4 (EphB4), Vascular Endothelial Growth Factor (VEGF), Erythropoietin (EPO), and Bone Morphogenetic Protein 2 (BMP-2), thus promoting angiogenesis and fracture healing.This paper explores the mechanisms by which EA facilitates bone healing and assesses its advantages and limitations in clinical applications. It offers a theoretical foundation for the safe, effective, and rational use of EA, presenting a novel approach for enhancing bone regeneration.
Collapse
Affiliation(s)
- Yu-Jun Gao
- Graduate School, Heilongjiang University of Chinese Medicine, Haerbin, China
| | - Yin-cang Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Haerbin, China
| | - De-lai Zhao
- The Fifth Hospital of Harbin City, Haerbin, China
| | - Qing Wen
- Graduate School, Heilongjiang University of Chinese Medicine, Haerbin, China
| | - Hao-Xin Shi
- Graduate School, Heilongjiang University of Chinese Medicine, Haerbin, China
| | - Shu-Ren Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Haerbin, China
| |
Collapse
|
4
|
Tian X, Zhang Y, Gong H, Bai M, Sun C, Jia Y, Duan C, Wang X. Knockdown of ANO1 decreases TGF-β- and IL-6-induced adhesion and migration of cardiac fibroblasts by inhibiting the expression of integrin and focal adhesion kinase. Exp Cell Res 2024; 443:114321. [PMID: 39505094 DOI: 10.1016/j.yexcr.2024.114321] [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: 07/20/2024] [Revised: 11/02/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
Ischemic cardiac injury triggers a significant inflammatory response, activating and mobilizing cardiac fibroblasts (CFs), which ultimately contributes to myocardial fibrosis. In this study, we investigated the role of ANO1, a calcium-activated chloride channel (CaCC) protein, in regulating CFs migration and adhesion under inflammatory conditions. Our results demonstrated that ANO1 knockdown significantly attenuated TGF-β- and IL-6-induced adhesion and migration of CFs. This inhibitory effect was mediated through the downregulation of integrin expression and reduced activation of focal adhesion kinase (FAK), key components in cellular adhesion and motility pathways. This study provides new insights into the mechanisms underlying CFs migration and adhesion, highlighting the potential of ANO1 as a therapeutic target for mitigating adverse fibrotic remodeling following myocardial infarction.
Collapse
Affiliation(s)
- Xiangqin Tian
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Yajing Zhang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Hezhe Gong
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Mengru Bai
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Changye Sun
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Yangyang Jia
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Changen Duan
- Cardiovascular Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
5
|
Hu P, Zhang Z, Yu X, Wang Y. 5-Hydroxymethylfurfural Ameliorates Allergic Inflammation in HMC-1 Cells by Inactivating NF-κB and MAPK Signaling Pathways. Biochem Genet 2024; 62:1521-1538. [PMID: 37648883 DOI: 10.1007/s10528-023-10492-9] [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: 06/06/2023] [Accepted: 08/06/2023] [Indexed: 09/01/2023]
Abstract
Allergic inflammation is the foundation of multiple allergic disorders, such as allergic rhinitis and asthma. Mast cells are effector cells that initiate inflammatory response. 5-hydroxymethylfurfural (5-HMF), a furfural compound, is the heat-processed product of various fruit, foods, drinks, as well as some Chinese herbal medicines. 5-HMF was previously reported to inhibit mast cell activation. Our study aimed to explore the functions of 5-HMF in both phorbol 12-mystate 13-acetate (PMA) plus calcium ionophore (A23187)-induced allergic inflammation in human mast cell line HMC-1 and ovalbumin (OVA)-induced asthma mouse models. HMC-1 cells were pretreated with 5-HMF and then stimulated by PMA+A23187. The cytotoxicity of 5-HMF on HMC-1 cells was evaluated by MTT assay. Histamine content in cell supernatants was measured by the o-phthaldialdehyde spectrofluorometric procedure. Intracellular calcium was determined using the fluorescent dye Fura-2AM. The production and expression of pro-inflammatory cytokines were detected by ELISA and RT-qPCR. Caspase-1 colorimetric assay was employed to examine the enzymatic activity of caspase-1. Asthma mouse models were induced by OVA sensitization. The bronchoalveolar lavage fluid (BALF) and blood samples were collected for the detection of total and differential cell count as well as aspartate aminotransferase (AST), alanine aminotransferase (ALT), OVA-immunoglobulin E (OVA-IgE), OVA-immunoglobulin G1 (OVA-IgG1), and pro-inflammatory cytokine levels. The left lung of mouse was dissected for histopathological examination by hematoxylin and eosin (H&E) staining. The protein expression of pro-caspase-1 and the phosphorylation of NF-κB and MAPK pathway-associated molecules were assessed by Western blotting. Our findings revealed that 5-HMF efficiently suppressed the PMA+A23187-induced enhancement in histamine production and intracellular calcium in HMC-1 cells. Pro-inflammatory cytokine production and expression in HMC-1 cells were elevated after PMA plus A23187 stimulation, which, however, were inhibited by pretreatment of 5-HMF. Additionally, 5-HMF suppressed the activity of caspase-1 and the phosphorylation of NF-κB and MAPK-associated molecules including p65 NF-κB, p38 MAPK, ERK, and JNK in HMC-1 cells. In vivo experiments demonstrated that 5-HMF treatment reduced the lung/body weight index and total and differential (macrophages, neutrophils, lymphocytes, and eosinophils) cell counts in BALF of asthmatic mice, but exerted no influence on serum AST and ALT levels. Besides, 5-HMF reduced serum OVA-IgE and OVA-IgG1 levels, mitigated lung inflammation, and inhibited the NF-κB and MAPK signaling pathways in asthma mouse models. 5-HMF mitigates allergic inflammation in asthma by inactivating caspase-1 and NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Pan Hu
- Department of Emergency, Hubei Provincial Hospital of TCM, No. 4, Huayuan Mountain, Wuchang District, Wuhan, 430000, Hubei, China
| | - Zhuo Zhang
- Department of Emergency, Hubei Provincial Hospital of TCM, No. 4, Huayuan Mountain, Wuchang District, Wuhan, 430000, Hubei, China
| | - Xiaolin Yu
- Department of Pediatrics, Hubei Provincial Hospital of TCM, Wuhan, 430000, Hubei, China
| | - Yinglin Wang
- Department of Emergency, Hubei Provincial Hospital of TCM, No. 4, Huayuan Mountain, Wuchang District, Wuhan, 430000, Hubei, China.
| |
Collapse
|
6
|
Pio-Lopez L, Levin M. Aging as a loss of morphostatic information: A developmental bioelectricity perspective. Ageing Res Rev 2024; 97:102310. [PMID: 38636560 DOI: 10.1016/j.arr.2024.102310] [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: 11/05/2023] [Revised: 02/21/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Maintaining order at the tissue level is crucial throughout the lifespan, as failure can lead to cancer and an accumulation of molecular and cellular disorders. Perhaps, the most consistent and pervasive result of these failures is aging, which is characterized by the progressive loss of function and decline in the ability to maintain anatomical homeostasis and reproduce. This leads to organ malfunction, diseases, and ultimately death. The traditional understanding of aging is that it is caused by the accumulation of molecular and cellular damage. In this article, we propose a complementary view of aging from the perspective of endogenous bioelectricity which has not yet been integrated into aging research. We propose a view of aging as a morphostasis defect, a loss of biophysical prepattern information, encoding anatomical setpoints used for dynamic tissue and organ homeostasis. We hypothesize that this is specifically driven by abrogation of the endogenous bioelectric signaling that normally harnesses individual cell behaviors toward the creation and upkeep of complex multicellular structures in vivo. Herein, we first describe bioelectricity as the physiological software of life, and then identify and discuss the links between bioelectricity and life extension strategies and age-related diseases. We develop a bridge between aging and regeneration via bioelectric signaling that suggests a research program for healthful longevity via morphoceuticals. Finally, we discuss the broader implications of the homologies between development, aging, cancer and regeneration and how morphoceuticals can be developed for aging.
Collapse
Affiliation(s)
- Léo Pio-Lopez
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Ribeiro Liberato H, Bezerra Maciel J, Wlisses Da Silva A, Eduarda Uchoa Bezerra M, San De Oliveira Brito L, Silva J, Kuerislene Amâncio Ferreira M, Machado Marinho M, Marinho GS, Deusdênia Loiola Pessoa O, Guedes MIF, Goberlânio De Barros Silva P, Ferreira de Castro Gomes A, Silva Alencar De Menezes JE, Silva Dos Santos H. Neuromodulation of Acid-Sensitive Ion Channels (ASICs) and Anti-Inflammatory Potential by Lichenxanthone in Adult Zebrafish (Danio rerio): Experimental and Docking Studies. Chem Biodivers 2024; 21:e202400063. [PMID: 38329295 DOI: 10.1002/cbdv.202400063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/09/2024]
Abstract
The xanthone lichenxanthone did not show toxic effects (LC50>1.0 mg/mL). lichenxanthone prevented nociceptive behavior induced by acidic saline, and its analgesic effect was blocked by amiloride, highlighting the involvement of neuromodulation of acid-sensitive ion channels (ASICs). In the analysis of anti-inflammatory activity, concentrations of 0.1 and 0.5 mg/mL of lichenxanthone reduced the edema induced by k-carrageenan 3.5 %, observed from the fourth hour of analysis. This effect was similar to that observed with ibuprofen (positive control). No leukocyte infiltrates were observed in lichenxanthone, suggesting that the compound acts in the acute inflammatory response. The results of the molecular docking study revealed that lichenxanthone exhibited better affinity energy when compared to the ibuprofen control against the two targets evaluated.
Collapse
Affiliation(s)
- Hortência Ribeiro Liberato
- Programa de Pós-graduação em Ciências Naturais, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | - Jéssica Bezerra Maciel
- Programa de Pós-graduação em Ciências Naturais, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | | | | | - Luana San De Oliveira Brito
- Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, Campus do Pici s/n, Fortaleza, Ceará, Brazil
| | - Jacilene Silva
- Programa de Pós-graduação em Ciências Naturais, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | | | - Marcia Machado Marinho
- Programa de Pós-graduação em Ciências Naturais, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | - Gabrielle S Marinho
- Programa de Pós-graduação em Ciências Naturais, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | - Otília Deusdênia Loiola Pessoa
- Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, Campus do Pici s/n, Fortaleza, Ceará, Brazil
| | - Maria Izabel F Guedes
- Centro de Ciências da Saúde, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | | | | | | | - Hélcio Silva Dos Santos
- Programa de Pós-graduação em Ciências Naturais, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
- Universidade Estadual do Vale do Acaraú, Centro de Ciências Exatas e Tecnologia, Sobral, Ceará, Brasil
| |
Collapse
|
8
|
Zhang Y, Ye F, Fu X, Li S, Wang L, Chen Y, Li H, Hao S, Zhao K, Feng Q, Li P. Mitochondrial Regulation of Macrophages in Innate Immunity and Diverse Roles of Macrophages During Cochlear Inflammation. Neurosci Bull 2024; 40:255-267. [PMID: 37391607 PMCID: PMC10838870 DOI: 10.1007/s12264-023-01085-y] [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: 02/14/2023] [Accepted: 05/05/2023] [Indexed: 07/02/2023] Open
Abstract
Macrophages are essential components of the innate immune system and constitute a non-specific first line of host defense against pathogens and inflammation. Mitochondria regulate macrophage activation and innate immune responses in various inflammatory diseases, including cochlear inflammation. The distribution, number, and morphological characteristics of cochlear macrophages change significantly across different inner ear regions under various pathological conditions, including noise exposure, ototoxicity, and age-related degeneration. However, the exact mechanism underlying the role of mitochondria in macrophages in auditory function remains unclear. Here, we summarize the major factors and mitochondrial signaling pathways (e.g., metabolism, mitochondrial reactive oxygen species, mitochondrial DNA, and the inflammasome) that influence macrophage activation in the innate immune response. In particular, we focus on the properties of cochlear macrophages, activated signaling pathways, and the secretion of inflammatory cytokines after acoustic injury. We hope this review will provide new perspectives and a basis for future research on cochlear inflammation.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fanglei Ye
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaolong Fu
- Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250000, China
| | - Shen Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Le Wang
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yutian Chen
- The Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hongmin Li
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Shaojuan Hao
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Kun Zhao
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qi Feng
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Province Research Center of Kidney Disease, Zhengzhou, 450052, China.
| | - Peipei Li
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Province Research Center of Kidney Disease, Zhengzhou, 450052, China.
| |
Collapse
|
9
|
Mittli D. Inflammatory processes in the prefrontal cortex induced by systemic immune challenge: Focusing on neurons. Brain Behav Immun Health 2023; 34:100703. [PMID: 38033612 PMCID: PMC10682838 DOI: 10.1016/j.bbih.2023.100703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/30/2023] [Accepted: 11/04/2023] [Indexed: 12/02/2023] Open
Abstract
Peripheral immune challenge induces neurobiological alterations in the brain and related neuropsychiatric symptoms both in humans and other mammals. One of the best known physiological effects of systemic inflammation is sickness behavior. However, in addition to this depression-like state, there are other cognitive outcomes of peripherally induced neuroinflammation that can be linked to the dysfunction of higher-order cortical areas, such as the prefrontal cortex (PFC). As the physiological activity of the PFC is largely based on the balanced interplay of excitatory pyramidal cells and inhibitory interneurons, it may be hypothesized that neuroinflammatory processes result in a shift of excitatory/inhibitory balance, which is a common hallmark of several neuropsychiatric conditions. Indeed, many data suggest that peripherally induced neuroinflammation is strongly associated with molecular and functional changes in PFC neurons leading to disturbances in their synaptic networks. Different experimental approaches may cause some incongruence in the reviewed data. However, it is commonly agreed that acute systemic inflammation leads to changes in the excitatory/inhibitory balance in the PFC by proinflammatory signaling at the brain borders and in the brain parenchyma. These cellular changes result in altered local and brain-wide network activity inducing disturbances in the top-down control of goal-directed behavior and cognition regulated by the PFC. Lipopolysaccharide (LPS)-treated rodents are the most widely used experimental models of peripherally induced neuroinflammation, so the majority of the reviewed data come from studies utilizing the LPS model. This may limit their general interpretation regarding the neuronal effects of peripheral immune activation. In addition, several biological variables (e.g., sex, age) can influence the PFC effects of systemic immune challenge, not only the nature and severity of immune activation. Therefore, it would be desirable to investigate inflammation-related neuronal changes in the PFC using other models of systemic inflammation as well, and to focus on the targeted fine-tuning of the affected cell types via common molecular mechanisms of the immune and nervous systems.
Collapse
Affiliation(s)
- Dániel Mittli
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Physiology and Neurobiology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- InnoScience Ltd., Mátranovák, Hungary
| |
Collapse
|
10
|
Gauthier MM, Hayoz S, Banek CT. Neuroimmune interplay in kidney health and disease: Role of renal nerves. Auton Neurosci 2023; 250:103133. [PMID: 38061177 PMCID: PMC10748436 DOI: 10.1016/j.autneu.2023.103133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023]
Abstract
Renal nerves and their role in physiology and disease have been a topic of increasing interest in the past few decades. Renal inflammation contributes to many cardiorenal disease conditions, including hypertension, chronic kidney disease, and polycystic kidney disease. Much is known about the role of renal sympathetic nerves in physiology - they contribute to the regulation of sodium reabsorption, renin release, and renal vascular resistance. In contrast, far less is known about afferent, or "sensory," renal nerves, which convey signals from the kidney to the brain. While much remains unknown about these nerves in the context of normal physiology, even less is known about their contribution to disease states. Furthermore, it has become apparent that the crosstalk between renal nerves and the immune system may augment or modulate disease. Research from other fields, especially pain research, has provided critical insight into neuroimmune crosstalk. Sympathetic renal nerve activity may increase immune cell recruitment, but far less work has been done investigating the interplay between afferent renal nerves and the immune system. Evidence from other fields suggests that inflammation may augment afferent renal nerve activity. Furthermore, these nerves may exacerbate renal inflammation through the release of afferent-specific neurotransmitters.
Collapse
Affiliation(s)
- Madeline M Gauthier
- Department of Physiology, University of Arizona Health Sciences Center, Tucson, AZ, USA
| | - Sebastien Hayoz
- Department of Physiology, University of Arizona Health Sciences Center, Tucson, AZ, USA
| | - Christopher T Banek
- Department of Physiology, University of Arizona Health Sciences Center, Tucson, AZ, USA.
| |
Collapse
|
11
|
Gladkikh IN, Klimovich AA, Kalina RS, Kozhevnikova YV, Khasanov TA, Osmakov DI, Koshelev SG, Monastyrnaya MM, Andreev YA, Leychenko EV, Kozlov SA. Anxiolytic, Analgesic and Anti-Inflammatory Effects of Peptides Hmg 1b-2 and Hmg 1b-4 from the Sea Anemone Heteractis magnifica. Toxins (Basel) 2023; 15:toxins15050341. [PMID: 37235375 DOI: 10.3390/toxins15050341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Acid-sensing ion channels (ASICs) have been known as sensors of a local pH change within both physiological and pathological conditions. ASIC-targeting peptide toxins could be potent molecular tools for ASIC-manipulating in vitro, and for pathology treatment in animal test studies. Two sea anemone toxins, native Hmg 1b-2 and recombinant Hmg 1b-4, both related to APETx-like peptides, inhibited the transient current component of human ASIC3-Δ20 expressed in Xenopus laevis oocytes, but only Hmg 1b-2 inhibited the rat ASIC3 transient current. The Hmg 1b-4 action on rASIC3 as a potentiator was confirmed once again. Both peptides are non-toxic molecules for rodents. In open field and elevated plus maze tests, Hmg 1b-2 had more of an excitatory effect and Hmg 1b-4 had more of an anxiolytic effect on mouse behavior. The analgesic activity of peptides was similar and comparable to diclofenac activity in an acid-induced muscle pain model. In models of acute local inflammation induced by λ-carrageenan or complete Freund's adjuvant, Hmg 1b-4 had more pronounced and statistically significant anti-inflammatory effects than Hmg 1b-2. It exceeded the effect of diclofenac and, at a dose of 0.1 mg/kg, reduced the volume of the paw almost to the initial volume. Our data highlight the importance of a comprehensive study of novel ASIC-targeting ligands, and in particular, peptide toxins, and present the slightly different biological activity of the two similar toxins.
Collapse
Affiliation(s)
- Irina N Gladkikh
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia
| | - Anna A Klimovich
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia
| | - Rimma S Kalina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia
| | - Yulia V Kozhevnikova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia
| | - Timur A Khasanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Dmitry I Osmakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Sergey G Koshelev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Margarita M Monastyrnaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia
| | - Yaroslav A Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Elena V Leychenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 690022 Vladivostok, Russia
| | - Sergey A Kozlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
12
|
Rychlik N, Hundehege P, Budde T. Influence of inflammatory processes on thalamocortical activity. Biol Chem 2023; 404:303-310. [PMID: 36453998 DOI: 10.1515/hsz-2022-0215] [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: 06/29/2022] [Accepted: 11/12/2022] [Indexed: 12/03/2022]
Abstract
It is known that the thalamus plays an important role in pathological brain conditions involved in demyelinating, inflammatory and neurodegenerative diseases such as Multiple Sclerosis (MS). Beside immune cells and cytokines, ion channels were found to be key players in neuroinflammation. MS is a prototypical example of an autoimmune disease of the central nervous system that is classified as a channelopathy where abnormal ion channel function leads to symptoms and clinical signs. Here we review the influence of the cytokine-ion channel interaction in the thalamocortical system in demyelination and inflammation.
Collapse
Affiliation(s)
- Nicole Rychlik
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany
| | - Petra Hundehege
- Department of Neurology with Institute of Translational Neurology, D-48149 Münster, Germany
| | - Thomas Budde
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany
| |
Collapse
|
13
|
Kim YY, Lee S, Kim MJ, Rho MC, Jang YH, Kim SH. Oleanolic Acid Acetate Inhibits Mast Cell Activation in Ovalbumin-Induced Allergic Airway Inflammation. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2023; 15:214-230. [PMID: 37021507 PMCID: PMC10079514 DOI: 10.4168/aair.2023.15.2.214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/24/2022] [Accepted: 10/02/2022] [Indexed: 04/07/2023]
Abstract
PURPOSE Asthma is a complex, heterogeneous chronic inflammatory airway disease with multiple phenotypes. There has been a great progress in managing asthma, but there are still unmet needs for developing uncontrolled asthma treatments. The present study aimed to determine the effectiveness of oleanolic acid acetate (OAA) from Vigna angularis against allergic airway inflammation and the underlying mechanism of action with a focus on mast cells. METHODS To investigate the effect of OAA in allergic airway inflammation, we used the ovalbumin (OVA)-sensitized and challenged mice. To examine allergic airway inflammation associated with immune responses of mast cell activation in vitro, various types of mast cells were used. Systemic and cutaneous anaphylaxis models were used for mast cell-mediated hyper-responsiveness in vivo. RESULTS OAA reduced OVA-induced airway inflammatory responses such as bronchospasm, increase of immune cell infiltration and serum immunoglobulin E and G1 levels. Especially, OAA decreased the mast cell infiltration, and β-hexosaminidase release as a mast cell activation marker in the bronchoalveolar lavage fluid. OAA inhibited mast cell degranulation in mast cell line (RBL-2H3) and primary cells (rat peritoneal mast cell and mouse bone marrow-derived mast cell). Mechanistically, OAA suppressed intracellular signaling pathways including the phosphorylation of phospholipase Cγ and nuclear factor-κB, resulting from the suppression of intracellular calcium influx and pro-inflammatory cytokine expression. Further, oral administration of OAA attenuated mast cell-mediated systemic and cutaneous anaphylaxis. CONCLUSIONS Our study showed that OAA can inhibit mast cell-mediated allergic reaction. Consequently, the application of OAA to mast cells for the allergic airway inflammation facilitate a new direction of treating allergic asthma.
Collapse
Affiliation(s)
- Yeon-Yong Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Korea
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Korea
| | - Soyoung Lee
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Korea
| | - Min-Jong Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Mun-Chual Rho
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Korea
| | - Yong Hyun Jang
- Department of Dermatology, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea.
| | - Sang-Hyun Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Korea.
| |
Collapse
|
14
|
First Anti-Inflammatory Peptide AnmTX Sco 9a-1 from the Swimming Sea Anemone Stomphia coccinea. Biomolecules 2022; 12:biom12111705. [DOI: 10.3390/biom12111705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
A novel peptide AnmTX Sco 9a-1 with the β-hairpin fold was isolated from the swimming sea anemone Stomphia coccinea (Actinostolidae family). The peptide consists of 28 amino acid residues, including modified hydroxyproline residue, and its measured molecular mass is 2960 Da. The peptide was not toxic on mice; however, it stimulated their exploratory motivation and active search behavior, and demonstrated an anti-anxiety effect. AnmTX Sco 9a-1 at doses of 0.1 and 1 mg/kg reduced the volume of edema during 24 h better than the nonsteroidal anti-inflammatory drug, Diclofenac, at dose of 1 mg/kg in a model of acute local λ-carrageenan-induced inflammation. ELISA analysis of the animal’s blood showed that peptide at a dose of 1 mg/kg reduced the content of tumor necrosis factor-α (TNF-α), a pro-inflammatory mediator responsible in the edema development, up to the level of TNF-α in the intact group. Besides, AnmTX Sco 9a-1 demonstrated a significant analgesic effect on acute pain sensitivity in the carrageenan-induced thermal hyperalgesia model at doses of 0.1 and 1 mg/kg. Activity of AnmTX Sco 9a-1 was shown not to be associated with modulation of nociceptive ASIC channels.
Collapse
|
15
|
Kelkar S, Nailwal N, Bhatia NY, Doshi G, Sathaye S, Godad AP. An Update On Proficiency of Voltage-gated Ion Channel Blockers in the Treatment of Inflammation-associated Diseases. Curr Drug Targets 2022; 23:1290-1303. [PMID: 35996239 DOI: 10.2174/1389450123666220819141827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/24/2022] [Accepted: 06/21/2022] [Indexed: 01/25/2023]
Abstract
Inflammation is the body's mechanism to trigger the immune system, thereby preventing bacteria and viruses from manifesting their toxic effect. Inflammation plays a vital role in regulating inflammatory mediator levels to initiate the wound healing process depending on the nature of the stimuli. This process occurs due to chemical release from white blood cells by elevating blood flow to the site of action, leading to redness and increased body temperature. Currently, there are numerous Non-steroidal anti-inflammatory drugs (NSAIDs) available, but these drugs are reported with adverse effects such as gastric bleeding, progressive kidney damage, and increased risk of heart attacks when prolonged use. For such instances, alternative options need to be adopted. The introduction of voltage-gated ion channel blockers can be a substantial alternative to mask the side effects of these currently available drugs. Chronic inflammatory disorders such as rheumatoid and osteoarthritis, cancer and migraine, etc., can cause dreadful pain, which is often debilitating for the patient. The underlying mechanism for both acute and chronic inflammation involves various complex receptors, different types of cells, receptors, and proteins. The working of voltage-gated sodium and calcium channels is closely linked to both inflammatory and neuropathic pain. Certain drugs such as carbamazepine and gabapentin, which are ion channel blockers, have greater pharmacotherapeutic activity for sodium and calcium channel blockers for the treatment of chronic inflammatory pain states. This review intends to provide brief information on the mechanism of action, latest clinical trials, and applications of these blockers in treating inflammatory conditions.
Collapse
Affiliation(s)
- Siddesh Kelkar
- MET Institute of Pharmacy, Bhujbal Knowledge City, Reclamation, Bandra West, Mumbai, Maharashtra 400050, India
| | - Namrata Nailwal
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra 400056, India
| | - Nirav Yogesh Bhatia
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra 400056, India
| | - Gaurav Doshi
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra 400056, India
| | - Sadhana Sathaye
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Angel Pavalu Godad
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra 400056, India.,Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| |
Collapse
|
16
|
Schmidt H, Gutjahr L, Sauter A, Zech F, Nchioua R, Stenger S, Frick M, Kirchhoff F, Dietl P, Wittekindt OH. Serially passaged, conditionally reprogrammed nasal epithelial cells as a model to study epithelial functions and SARS-CoV-2 infection. Am J Physiol Cell Physiol 2022; 322:C591-C604. [PMID: 35196166 PMCID: PMC8977148 DOI: 10.1152/ajpcell.00363.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Primary airway epithelial cells (pAECs) cultivated at air-liquid interface (ALI) conditions are widely used as surrogates for human in vivo epithelia. To extend the proliferative capacity and to enable serially passaging of pAECs, conditional reprogramming (cr) has been employed in recent years. However, ALI epithelia derived from cr cells often display functional changes with increasing passages. This highlights the need for thorough validation of the ALI cultures for the respective application. In our study, we evaluated the use of serially passaged cr nasal epithelial cells (crNECs) as a model to study SARS-CoV-2 infection and effects on ion and water transport. NECs were obtained from healthy individuals and cultivated as ALI epithelia derived from passages 1, 2, 3, and 5. We compared epithelial differentiation, ion and water transport, and infection with SARS-CoV-2 between passages. Our results show that epithelia maintained major differentiation characteristics and physiological ion and water transport properties through all passages. However, the frequency of ciliated cells, short circuit currents reflecting epithelial Na+ channel (ENaC) and cystic fibrosis transmembrane conductance regulator (CFTR) activity and expression of aquaporin 3 and 5 decreased gradually over passages. crNECs also expressed SARS-CoV-2 receptors angiotensin converting enzyme 2 (ACE2) and transmembrane serin2 protease 2 (TMPRSS2) across all passages and allowed SARS-CoV-2 replication in all passages. In summary, we provide evidence that passaged crNECs provide an appropriate model to study SARS-CoV-2 infection and also epithelial transport function when considering some limitations that we defined herein.
Collapse
Affiliation(s)
- Hanna Schmidt
- Department of Pediatric and Adolescent Medicine, Ulm University Medical Cente, Ulm, Germany.,Institute of General Physiology, Ulm University, Ulm, Germany
| | - Lara Gutjahr
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Rayhane Nchioua
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Steffen Stenger
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Paul Dietl
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | |
Collapse
|
17
|
Bazard P, Pineros J, Frisina RD, Bauer MA, Acosta AA, Paganella LR, Borakiewicz D, Thivierge M, Mannering FL, Zhu X, Ding B. Cochlear Inflammaging in Relation to Ion Channels and Mitochondrial Functions. Cells 2021; 10:2761. [PMID: 34685743 PMCID: PMC8534887 DOI: 10.3390/cells10102761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 12/20/2022] Open
Abstract
The slow accumulation of inflammatory biomarker levels in the body-also known as inflammaging-has been linked to a myriad of age-related diseases. Some of these include neurodegenerative conditions such as Parkinson's disease, obesity, type II diabetes, cardiovascular disease, and many others. Though a direct correlation has not been established, research connecting age-related hearing loss (ARHL)-the number one communication disorder and one of the most prevalent neurodegenerative diseases of our aged population-and inflammaging has gained interest. Research, thus far, has found that inflammatory markers, such as IL-6 and white blood cells, are associated with ARHL in humans and animals. Moreover, studies investigating ion channels and mitochondrial involvement have shown promising relationships between their functions and inflammaging in the cochlea. In this review, we summarize key findings in inflammaging within the auditory system, the involvement of ion channels and mitochondrial functions, and lastly discuss potential treatment options focusing on controlling inflammation as we age.
Collapse
Affiliation(s)
- Parveen Bazard
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Jennifer Pineros
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Robert D. Frisina
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
- Department Communication Sciences and Disorders, College of Behavioral & Communication Sciences, Tampa, FL 33620, USA
- Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Mark A. Bauer
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Alejandro A. Acosta
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Lauren R. Paganella
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Dominika Borakiewicz
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Mark Thivierge
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Freyda L. Mannering
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
- Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Xiaoxia Zhu
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| | - Bo Ding
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (J.P.); (M.A.B.); (A.A.A.); (L.R.P.); (D.B.); (M.T.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA;
| |
Collapse
|
18
|
Regulation of Store-Operated Ca 2+ Entry by SARAF. Cells 2021; 10:cells10081887. [PMID: 34440656 PMCID: PMC8391525 DOI: 10.3390/cells10081887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022] Open
Abstract
Calcium (Ca2+) signaling plays a dichotomous role in cellular biology, controlling cell survival and proliferation on the one hand and cellular toxicity and cell death on the other. Store-operated Ca2+ entry (SOCE) by CRAC channels represents a major pathway for Ca2+ entry in non-excitable cells. The CRAC channel has two key components, the endoplasmic reticulum Ca2+ sensor stromal interaction molecule (STIM) and the plasma-membrane Ca2+ channel Orai. Physical coupling between STIM and Orai opens the CRAC channel and the resulting Ca2+ flux is regulated by a negative feedback mechanism of slow Ca2+ dependent inactivation (SCDI). The identification of the SOCE-associated regulatory factor (SARAF) and investigations of its role in SCDI have led to new functional and molecular insights into how SOCE is controlled. In this review, we provide an overview of the functional and molecular mechanisms underlying SCDI and discuss how the interaction between SARAF, STIM1, and Orai1 shapes Ca2+ signaling in cells.
Collapse
|
19
|
Eisenhut M. The significance of a lack of rhinorrhea in severe coronavirus 19 lung disease. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1194-L1195. [PMID: 34156889 PMCID: PMC8238443 DOI: 10.1152/ajplung.00066.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Michael Eisenhut
- Paediatric Department, Luton & Dunstable University Hospital, Luton, United Kingdom
| |
Collapse
|
20
|
Zhou J, Zhou Z, Liu X, Yin HY, Tang Y, Cao X. P2X7 Receptor-Mediated Inflammation in Cardiovascular Disease. Front Pharmacol 2021; 12:654425. [PMID: 33995071 PMCID: PMC8117356 DOI: 10.3389/fphar.2021.654425] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/09/2021] [Indexed: 02/05/2023] Open
Abstract
Purinergic P2X7 receptor, a nonselective cation channel, is highly expressed in immune cells as well as cardiac smooth muscle cells and endothelial cells. Its activation exhibits to mediate nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) inflammasome activation, resulting in the release of interleukin-1 beta (IL-1β) and interleukin-18 (IL-18), and pyroptosis, thus triggering inflammatory response. These pathological mechanisms lead to the deterioration of various cardiovascular diseases, including atherosclerosis, arrhythmia, myocardial infarction, pulmonary vascular remodeling, and cardiac fibrosis. All these worsening cardiac phenotypes are proven to be attenuated after the P2X7 receptor inhibition in experimental studies. The present review aimed to summarize key aspects of P2X7 receptor-mediated inflammation and pyroptosis in cardiovascular diseases. The main focus is on the evidence addressing the involvement of the P2X7 receptor in the inflammatory responses to the occurrence and development of cardiovascular disease and therapeutic interventions.
Collapse
Affiliation(s)
- Junteng Zhou
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Xiaojing Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hai-Yan Yin
- School of Acupuncture and Tuina and International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Yong Tang
- School of Acupuncture and Tuina and International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Xin Cao
- School of Acupuncture and Tuina and International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
21
|
Fouda MA, Ruben PC. Protein Kinases Mediate Anti-Inflammatory Effects of Cannabidiol and Estradiol Against High Glucose in Cardiac Sodium Channels. Front Pharmacol 2021; 12:668657. [PMID: 33995099 PMCID: PMC8115126 DOI: 10.3389/fphar.2021.668657] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/16/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Cardiovascular anomalies are predisposing factors for diabetes-induced morbidity and mortality. Recently, we showed that high glucose induces changes in the biophysical properties of the cardiac voltage-gated sodium channel (Nav1.5) that could be strongly correlated to diabetes-induced arrhythmia. However, the mechanisms underlying hyperglycemia-induced inflammation, and how inflammation provokes cardiac arrhythmia, are not well understood. We hypothesized that inflammation could mediate the high glucose-induced biophyscial changes on Nav1.5 through protein phosphorylation by protein kinases A and C. We also hypothesized that this signaling pathway is, at least partly, involved in the cardiprotective effects of cannabidiol (CBD) and 17β-estradiol (E2). Methods and Results: To test these ideas, we used Chinese hamster ovarian (CHO) cells transiently co-transfected with cDNA encoding human Nav1.5 α-subunit under control, a cocktail of inflammatory mediators or 100 mM glucose conditions (for 24 h). We used electrophysiological experiments and action potential modeling. Inflammatory mediators, similar to 100 mM glucose, right shifted the voltage dependence of conductance and steady-state fast inactivation and increased persistent current leading to computational prolongation of action potential (hyperexcitability) which could result in long QT3 arrhythmia. We also used human iCell cardiomyocytes derived from inducible pluripotent stem cells (iPSC-CMs) as a physiologically relevant system, and they replicated the effects produced by inflammatory mediators observed in CHO cells. In addition, activators of PK-A or PK-C replicated the inflammation-induced gating changes of Nav1.5. Inhibitors of PK-A or PK-C, CBD or E2 mitigated all the potentially deleterious effects provoked by high glucose/inflammation. Conclusion: These findings suggest that PK-A and PK-C may mediate the anti-inflammatory effects of CBD and E2 against high glucose-induced arrhythmia. CBD, via Nav1.5, may be a cardioprotective therapeutic approach in diabetic postmenopausal population.
Collapse
Affiliation(s)
- Mohamed A Fouda
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada.,Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
| | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
22
|
Glaser N, Chu S, Hung B, Fernandez L, Wulff H, Tancredi D, ODonnell ME. Acute and chronic neuroinflammation is triggered by diabetic ketoacidosis in a rat model. BMJ Open Diabetes Res Care 2020; 8:e001793. [PMID: 33318070 PMCID: PMC7737057 DOI: 10.1136/bmjdrc-2020-001793] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/21/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Cognitive decline is common in patients with type 1 diabetes and has been attributed to the effects of chronic hyperglycemia and severe hypoglycemia. Diabetic ketoacidosis (DKA) has only recently been suspected to be involved in causing cognitive decline. We hypothesized that DKA triggers both acute and chronic neuroinflammation, contributing to brain injury. RESEARCH METHODS AND DESIGN We measured concentrations of cytokines, chemokines and matrix metalloproteinases (MMP) in serum and brain tissue lysates in juvenile rats during and after DKA (during acute DKA, 24 hours and 7 days after DKA), and compared these to healthy controls and hyperglycemic controls. We also measured cytokine, chemokine and MMP concentrations in serum and brain tissue of adult rats (70 days) that had experienced DKA as juveniles and compared these measurements to those of adult diabetic rats without exposure to DKA. RESULTS During acute DKA in the juvenile rats, serum concentrations of CCL3, tumor necrosis factor (TNF)-α, interleukin (IL)-1ß and MMP-9 were significantly increased. Serum concentrations of IL-2 and IL-17A increased 7 days after DKA recovery. In brain tissue lysates, concentrations of CCL3, CCL5, interferon (IFN)-γ and MMP-9 were significantly elevated during acute DKA. In adult rats that had DKA as juveniles (28 days previously), serum concentrations of IL-1ß and brain concentrations of IL-10 and IL-12p70 were elevated in comparison to diabetic rats without prior DKA. Composite scores for highly correlated cytokines and chemokines (mean z-scores for IL-10, IL-1ß, TNF-α, IL-17A, IFN-γ, CXCL-1 and CCL5) were also significantly elevated in adult rats with prior DKA. CONCLUSIONS These data confirm that DKA causes acute systemic inflammation and neuroinflammation in a rat model. Importantly, the neuroinflammatory response triggered by DKA is long-lasting, suggesting the possibility that DKA-induced chronic neuroinflammation could contribute to long-term cognitive decline in individuals with diabetes.
Collapse
Affiliation(s)
- Nicole Glaser
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, USA
| | - Steven Chu
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, USA
| | - Benjamin Hung
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Sacramento, California, USA
| | - Luis Fernandez
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, USA
| | - Heike Wulff
- Department of Pharmacology, University of California Davis School of Medicine, Sacramento, California, USA
| | - Daniel Tancredi
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, USA
| | - Martha E ODonnell
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Sacramento, California, USA
| |
Collapse
|
23
|
Eisenhut M, Shin JI. Pathways in the Pathophysiology of Coronavirus 19 Lung Disease Accessible to Prevention and Treatment. Front Physiol 2020; 11:872. [PMID: 32922301 PMCID: PMC7457053 DOI: 10.3389/fphys.2020.00872] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Background: In COVID 19 related lung disease, which is a leading cause of death from this disease, cytokines like tumor necrosis factor-alpha (TNF alpha) may be pivotal in the pathogenesis. TNF alpha reduces fluid absorption due to impairment of sodium and chloride transport required for building an osmotic gradient across epithelial cells, which in the airways maintains airway surface liquid helping to keep airways open and enabling bacterial clearance and aids water absorption from the alveolar spaces. TNF alpha can, through Rho-kinase, disintegrate the endothelial and epithelial cytoskeleton, and thus break up intercellular tight junctional proteins, breaching the intercellular barrier, which prevents flooding of the interstitial and alveolar spaces with fluid. Hypotheses: (1) Preservation and restoration of airway and alveolar epithelial sodium and chloride transport and the cytoskeleton dependent integrity of the cell barriers within the lung can prevent and treat COVID 19 lung disease. (2) TNF alpha is the key mediator of pulmonary edema in COVID 19 lung disease. Confirmation of hypothesis and implications: The role of a reduction in the function of epithelial sodium and chloride transport could with regards to chloride transport be tested by analysis of chloride levels in exhaled breath condensate and levels correlated with TNF alpha concentrations. Reduced levels would indicate a reduction of the function of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel and a correlation with TNF alpha levels indicative of its involvement. Anti-TNF alpha treatment with antibodies is already available and needs to be tested in randomized controlled trials of COVID 19 lung disease. TNF alpha levels could also be reduced by statins, aspirin, and curcumin. Chloride transport could be facilitated by CFTR activators, including curcumin and phosphodiesterase-5 inhibitors. Sodium and chloride transport could be further regulated to prevent accumulation of alveolar fluid by use of Na(+)/K(+)/2Cl(-) cotransporter type 1 inhibitors, which have been associated with improved outcome in adults ventilated for acute respiratory distress syndrome (ARDS) in randomized controlled trials. Primary prevention of coronavirus infection and TNF alpha release in response to it could be improved by induction of antimicrobial peptides LL-37 and human beta defensin-2 and reduction of TNF alpha production by vitamin D prophylaxis for the population as a whole.
Collapse
Affiliation(s)
- Michael Eisenhut
- Children's & Adolescent Services, Luton & Dunstable University Hospital NHS Foundation Trust, Luton, United Kingdom
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
24
|
Ahuja M, Chung WY, Lin WY, McNally BA, Muallem S. Ca 2+ Signaling in Exocrine Cells. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035279. [PMID: 31636079 DOI: 10.1101/cshperspect.a035279] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Calcium (Ca2+) and cyclic AMP (cAMP) signaling cross talk and synergize to stimulate the cardinal functions of exocrine cells, regulated exocytosis, and fluid and electrolyte secretion. This physiological process requires the organization of the two signaling pathways into complexes at defined cellular domains and close placement. Such domains are formed by membrane contact sites (MCS). This review discusses the basic properties of Ca2+ signaling in exocrine cells, the role of MCS in the organization of cell signaling and in cross talk and synergism between the Ca2+ and cAMP signaling pathways and, finally, the mechanism by which the Ca2+ and cAMP pathways synergize to stimulate epithelial fluid and electrolyte secretion.
Collapse
Affiliation(s)
- Malini Ahuja
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, Maryland 20892
| | - Woo Young Chung
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, Maryland 20892
| | - Wei-Yin Lin
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, Maryland 20892
| | - Beth A McNally
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, Maryland 20892
| | - Shmuel Muallem
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, Maryland 20892
| |
Collapse
|
25
|
Role of macrophage TRPV4 in inflammation. J Transl Med 2020; 100:178-185. [PMID: 31645630 PMCID: PMC7261496 DOI: 10.1038/s41374-019-0334-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/19/2019] [Accepted: 08/30/2019] [Indexed: 01/05/2023] Open
Abstract
Transient receptor ion channels have emerged as immensely important channels/receptors in diverse physiological and pathological responses. Of particular interest is the transient receptor potential channel subfamily V member 4 (TRPV4), which is a polymodal, nonselective, calcium-permeant cation channel, and is activated by both endogenous and exogenous stimuli. Both neuronal and nonneuronal cells express functional TRPV4, which is responsive to a variety of biochemical and biomechanical stimuli. Emerging discoveries have advanced our understanding of the role of macrophage TRPV4 in numerous inflammatory diseases. In lung injury, TRPV4 mediates macrophage phagocytosis, secretion of pro-resolution cytokines, and generation of reactive oxygen species. TRPV4 regulates lipid-laden macrophage foam cell formation, the hallmark of atheroinflammatory conditions, in response to matrix stiffness and lipopolysaccharide stimulation. Accumulating data also point to a role of macrophage TRPV4 in the pathogenesis of the foreign body response, a chronic inflammatory condition, through the formation of foreign body giant cells. Deletion of TRPV4 in macrophages suppresses the allergic and nonallergic itch in a mouse model, suggesting a role of TRPV4 in skin disease. Here, we discuss the current understanding of the role of macrophage TRPV4 in various inflammatory conditions.
Collapse
|
26
|
Aller MA, Arias N, Blanco-Rivero J, Arias J. Metabolism in Acute-On-Chronic Liver Failure: The Solution More than the Problem. Arch Med Res 2019; 50:271-284. [PMID: 31593852 DOI: 10.1016/j.arcmed.2019.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022]
Abstract
Chronic inflammatory liver disease with an acute deterioration of liver function is named acute-on-chronic inflammation and could be regulated by the metabolic impairments related to the liver dysfunction. In this way, the experimental cholestasis model is excellent for studying metabolism in both types of inflammatory responses. Along the evolution of this model, the rats develop biliary fibrosis and an acute-on-chronic decompensation. The acute decompensation of the liver disease is associated with encephalopathy, ascites, acute renal failure, an acute phase response and a splanchnic increase of pro- and anti-inflammatory cytokines. This multiorgan inflammatory dysfunction is mainly associated with a splanchnic and systemic metabolic switch with dedifferentiation of the epithelial, endothelial and mesothelial splanchnic barriers. Furthermore, a splanchnic infiltration by mast cells occurs, which suggests that these cells could carry out a compensatory metabolic role, especially through the modulation of hepatic and extrahepatic mitochondrial-peroxisome crosstalk. For this reason, we propose the hypothesis that mastocytosis in the acute-on-chronic hepatic insufficiency could represent the development of a survival metabolic mechanisms that mitigates the noxious effect of the hepatic functional deficit. A better understanding the pathophysiological response of the mast cells in liver insufficiency and portal hypertension would help to find new pathways for decreasing the high morbidity and mortality rate of these patients.
Collapse
Affiliation(s)
- Maria-Angeles Aller
- Department of Surgery, School of Medicine, Complutense University of Madrid, Madrid, Spain.
| | - Natalia Arias
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; INEUROPA (Instituto de Neurociencias del Principado de Asturias), Oviedo, Spain
| | - Javier Blanco-Rivero
- Department of Physiology, School of Medicine, Autonoma University of Madrid, Madrid, Spain, Instituto de Investigación Biomédica La Paz (IdIPAZ), Madrid, España; Centro de Investigación Biomédica en Red (Ciber) de Enfermedades Cardiovasculares, Madrid, España
| | - Jaime Arias
- Department of Surgery, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
27
|
DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol 2019; 20:95-112. [PMID: 31558839 DOI: 10.1038/s41577-019-0215-7] [Citation(s) in RCA: 1036] [Impact Index Per Article: 172.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2019] [Indexed: 12/11/2022]
Abstract
The innate immune system has the capacity to detect 'non-self' molecules derived from pathogens, known as pathogen-associated molecular patterns, via pattern recognition receptors. In addition, an increasing number of endogenous host-derived molecules, termed damage-associated molecular patterns (DAMPs), have been found to be sensed by various innate immune receptors. The recognition of DAMPs, which are produced or released by damaged and dying cells, promotes sterile inflammation, which is important for tissue repair and regeneration, but can also lead to the development of numerous inflammatory diseases, such as metabolic disorders, neurodegenerative diseases, autoimmune diseases and cancer. Here we examine recent discoveries concerning the roles of DAMP-sensing receptors in sterile inflammation and in diseases resulting from dysregulated sterile inflammation, and then discuss insights into the cross-regulation of these receptors and their ligands.
Collapse
|
28
|
Hsu HC, Hsieh CL, Wu SY, Lin YW. Toll-like receptor 2 plays an essential role in electroacupuncture analgesia in a mouse model of inflammatory pain. Acupunct Med 2019; 37:356-364. [PMID: 31517506 DOI: 10.1136/acupmed-2017-011469] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Inflammatory pain occurs when local tissue injury activates macrophages and neutrophils, hence increasing pro-inflammatory cytokine and chemokine levels. Toll-like receptor 2 (TLR2) antagonism reportedly suppresses neuropathic and inflammatory pain. AIMS In the present study, we investigated the effect of electroacupuncture (EA) on TLR2 and related signalling molecules in a complete Freund's adjuvant (CFA)-induced mouse model of inflammatory pain to determine whether EA can attenuate inflammatory pain via the TLR2 signalling pathway. METHODS EA significantly reduced mechanical and thermal hyperalgesia in the animal model. A similar effect was produced by TLR2 antagonism induced by CU-CPT22 injection. RESULTS TLR2 expression in the dorsal root ganglia, spinal cord and thalamus increased following induction of inflammation. Expression levels of downstream molecules such as pPI3K, pAkt and pmTOR also increased, as did those of MAPK subfamily members such as pERK, pp38 and pJNK. Transcription factors (pCREB and pNFκB) and nociceptive ion channels (Nav1.7 and Nav1.8) were also involved. CONCLUSION Increased expression of the above molecules was attenuated by both EA and TLR2 antagonism. Our results show that EA attenuates inflammatory pain via TLR2 signalling.
Collapse
Affiliation(s)
- Hsin-Cheng Hsu
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Ching-Liang Hsieh
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Shu-Yih Wu
- Department of Rehabilitation Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yi-Wen Lin
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Research Center for Chinese Medicine & Acupuncture, China Medical University, Taichung, Taiwan
| |
Collapse
|
29
|
Hyponatremia in infants with community-acquired infections on hospital admission. PLoS One 2019; 14:e0219299. [PMID: 31276475 PMCID: PMC6611618 DOI: 10.1371/journal.pone.0219299] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 06/20/2019] [Indexed: 12/17/2022] Open
Abstract
Acute moderate to severe gastroenteritis is traditionally associated with hypernatremia but recent observations suggest that hypernatremia is currently less common than hyponatremia. The latter has sometimes been documented also in children with acute community-acquired diseases, such as bronchiolitis and pyelonephritis. We investigated the prevalence of dysnatremia in children with acute moderate severe gastroenteritis, bronchiolitis and pyelonephritis. This prospective observational study included 400 consecutive previously healthy infants ≥4 weeks to ≤24 months of age (232 males and 168 females): 160 with gastroenteritis and relevant dehydration, 160 with moderate-severe bronchiolitis and 80 with pyelonephritis admitted to our emergency department between 2009 and 2017. Circulating sodium was determined by means of direct potentiometry. For analysis, the Kruskal-Wallis test and the Fisher’s exact test were used. Hyponatremia was found in 214 of the 400 patients. It was common in gastroenteritis (43%) and significantly more frequent in bronchiolitis (57%) and pyelonephritis (68%). Patients with hyponatremia were significantly younger than those without hyponatremia (3.9 [1.6–13] versus 7.5 [3.4–14] months). The gender ratio was similar in children with and without hyponatremia. Hyponatremia was associated with further metabolic abnormalities (hypokalemia, hyperkalemia, metabolic acidosis or metabolic alkalosis) in gastroenteritis (71%) and pyelonephritis (54%), and always isolated in bronchiolitis. In conclusion, hyponatremia is common at presentation among previously healthy infants with gastroenteritis, bronchiolitis or pyelonephritis. These data have relevant consequences for the nutrition and rehydration management in these conditions.
Collapse
|
30
|
Strickland M, Yacoubi-Loueslati B, Bouhaouala-Zahar B, Pender SLF, Larbi A. Relationships Between Ion Channels, Mitochondrial Functions and Inflammation in Human Aging. Front Physiol 2019; 10:158. [PMID: 30881309 PMCID: PMC6405477 DOI: 10.3389/fphys.2019.00158] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/08/2019] [Indexed: 12/19/2022] Open
Abstract
Aging is often associated with a loss of function. We believe aging to be more an adaptation to the various, and often continuous, stressors encountered during life in order to maintain overall functionality of the systems. The maladaptation of a system during aging may increase the susceptibility to diseases. There are basic cellular functions that may influence and/or are influenced by aging. Mitochondrial function is amongst these. Their presence in almost all cell types makes of these valuable targets for interventions to slow down or even reserve signs of aging. In this review, the role of mitochondria and essential physiological regulators of mitochondria and cellular functions, ion channels, will be discussed in the context of human aging. The origins of inflamm-aging, associated with poor clinical outcomes, will be linked to mitochondria and ion channel biology.
Collapse
Affiliation(s)
- Marie Strickland
- Singapore Immunology Network, Agency for Science Technology and Research, Singapore, Singapore
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Besma Yacoubi-Loueslati
- Laboratory of Mycology, Pathologies and Biomarkers, Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis, Tunisia
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Venoms and Therapeutic Molecules, Institut Pasteur de Tunis, University Tunis El Manar, Tunis, Tunisia
- Medical School of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Sylvia L. F. Pender
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Chinese University of Hong Kong – University of Southampton Joint Lab for Stem Cell and Regenerative Medicine, Hong Kong, China
| | - Anis Larbi
- Singapore Immunology Network, Agency for Science Technology and Research, Singapore, Singapore
- Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis, Tunisia
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Geriatrics Division, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
31
|
Carcinogenesis: the cancer cell–mast cell connection. Inflamm Res 2018; 68:103-116. [DOI: 10.1007/s00011-018-1201-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 12/20/2022] Open
|
32
|
Kang BC, Kim MJ, Lee S, Choi YA, Park PH, Shin TY, Kwon TK, Khang D, Kim SH. Nothofagin suppresses mast cell-mediated allergic inflammation. Chem Biol Interact 2018; 298:1-7. [PMID: 30392763 DOI: 10.1016/j.cbi.2018.10.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/16/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022]
Abstract
Mast cells play a major role in immunoglobulin E-mediated allergic inflammation, which is involved in asthma, atopic dermatitis, and allergic rhinitis. Nothofagin has been shown to ameliorate various inflammatory responses such as the septic response and vascular inflammation. In this study, we assessed the inhibitory effect of nothofagin on allergic inflammation using cultured/isolated mast cells and an anaphylaxis mouse model. Nothofagin treatment prevented histamine and β-hexosaminidase release by reducing the influx of calcium into the cytosol in a concentration-dependent manner. Nothofagin also inhibited the gene expression and secretion of pro-inflammatory cytokines such as tumor necrosis factor-α and interleukin-4 by downregulating the phosphorylation of Lyn, Syk, Akt and nuclear translocation of nuclear factor-κB. To confirm these effects of nothofagin in vivo, we used a passive cutaneous anaphylaxis mouse model. Topical administration of nothofagin suppressed local pigmentation and ear thickness. Taken together, these results suggest nothofagin as a potential candidate for the treatment of mast cell-involved allergic inflammatory diseases.
Collapse
Affiliation(s)
- Byeong-Cheol Kang
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Min-Jong Kim
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Soyoung Lee
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Young-Ae Choi
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Tae-Yong Shin
- College of Pharmacy, Woosuk University, Jeonju, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Dongwoo Khang
- Department of Physiology, School of Medicine, Gachon University, Incheon, Republic of Korea.
| | - Sang-Hyun Kim
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
33
|
In diabetic ketoacidosis brain injury including cerebral oedema and infarction is caused by interleukin-1. Med Hypotheses 2018; 121:44-46. [PMID: 30396487 DOI: 10.1016/j.mehy.2018.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Brain injury in diabetic ketoacidosis (DKA) is common but under recognized and affects up to 54% of patients with this complication. It's manifestations include cerebral oedema (CE) and cerebral infarction (CI). The etiology of CE in DKA has up to the present time been uncertain. Practical management had been guided by the assumption that rapid osmotic shifts due to rapid correction of hypovolemia and reduction of plasma glucose could cause a shift of water into the cells. The osmotic effect of glucose can cause inflammation by activation of inflammasomes. Recently it has been recognized that the body is in a pro-inflammatory state during DKA involving interleukin-1 production by inflammasomes. Interleukin-1 has been involved in the pathogenesis of cerebral oedema and CI. HYPOTHESIS In diabetic ketoacidosis brain injury including cerebral oedema and infarction is caused by interleukin-1. CONFIRMATION OF HYPOTHESIS AND IMPLICATIONS Inflammasome activity could be quantified in peripheral blood mononuclear cells and in patients with and without clinical and/or subclinical CE and/or stroke or features of cerebral ischemia on MRI. Surrogates of brain injury in peripheral blood like neuron specific enolase could be measured and correlated with inflammasome activity. Interleukin-1 receptor antagonists and inflammasome inhibitors including telmisartan could be assessed in their effect on MRI changes consistent with CE or CI in patients with DKA in randomised placebo-controlled trials.
Collapse
|
34
|
Chen X, Sun X, Wang Z, Zhou X, Xu L, Li F, Zhang X, Pan J, Qi L, Qian H, Mao Z. Involvement of acid-sensing ion channel 1a in gastric carcinoma cell migration and invasion. Acta Biochim Biophys Sin (Shanghai) 2018; 50:440-446. [PMID: 29584803 DOI: 10.1093/abbs/gmy026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 02/22/2018] [Indexed: 12/18/2022] Open
Abstract
Acidic microenvironment, particularly acid-sensing ion channel 1a (ASIC1a), has been reported to promote carcinoma cell proliferation as well as migration. In this study, we explored the effect of ASIC1a on migration and invasion of gastric carcinoma (GC). ASIC1a expression levels were examined in paired GC and adjacent normal tissues from 16 patients by immunohistochemistry. Reverse transcription real-time PCR and immunoblotting were conducted to assess the ASIC1a expression levels in the GC cell line AGS after transfection with ASIC1a small hairpin RNA (shRNA). Wound healing and transwell invasion assays were utilized to detect metastasis and invasion following ASIC1a silencing. Tumor formation was used to detect the role of ASIC1a in tumorigenicity in vivo. It was found that ASIC1a expression level was significantly higher in GC tissues showing postoperative metastasis compared with non-metastasis and non-tumor tissues. Moreover, silencing of ASIC1a with shRNA significantly down-regulated ASIC1a expression and reduced GC cell migration and invasion. A moderately acidic extracellular environment inhibited GC cell viability. Furthermore, ASIC1a shRNA caused inhibition of tumorigenicity in vivo. Our study is the first report of attenuating the malignant phenotype of GC in vitro and in vivo by suppressing ASIC1a, and suggests a novel approach to study the relationship between ASICs and GC cell migration and invasion.
Collapse
Affiliation(s)
- Xin Chen
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xue Sun
- Department of Intensive Care Unit, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Zhe Wang
- Department of Infectious Disease, Children's Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiaojun Zhou
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lu Xu
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Feng'e Li
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xingding Zhang
- School of Medicine (Shenzhen), Sun Yat-Sen University, Guangzhou 510006, China
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Hematology & Oncology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Ji'an Pan
- School of Medicine (Shenzhen), Sun Yat-Sen University, Guangzhou 510006, China
| | - Lin Qi
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Hematology & Oncology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Haixin Qian
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Zhongqi Mao
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
35
|
Wang Y, Gao Y, Tian Q, Deng Q, Wang Y, Zhou T, Liu Q, Mei K, Wang Y, Liu H, Ma R, Ding Y, Rong W, Cheng J, Yao J, Xu TL, Zhu MX, Li Y. TRPV1 SUMOylation regulates nociceptive signaling in models of inflammatory pain. Nat Commun 2018; 9:1529. [PMID: 29670121 PMCID: PMC5906468 DOI: 10.1038/s41467-018-03974-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 03/26/2018] [Indexed: 01/08/2023] Open
Abstract
Although TRPV1 channels represent a key player of noxious heat sensation, the precise mechanisms for thermal hyperalgesia remain unknown. We report here that conditional knockout of deSUMOylation enzyme, SENP1, in mouse dorsal root ganglion (DRG) neurons exacerbated thermal hyperalgesia in both carrageenan- and Complete Freund’s adjuvant-induced inflammation models. TRPV1 is SUMOylated at a C-terminal Lys residue (K822), which specifically enhances the channel sensitivity to stimulation by heat, but not capsaicin, protons or voltage. TRPV1 SUMOylation is decreased by SENP1 but upregulated upon peripheral inflammation. More importantly, the reduced ability of TRPV1 knockout mice to develop inflammatory thermal hyperalgesia was rescued by viral infection of lumbar 3/4 DRG neurons of wild-type TRPV1, but not its SUMOylation-deficient mutant, K822R. These data suggest that TRPV1 SUMOylation is essential for the development of inflammatory thermal hyperalgesia, through a mechanism that involves sensitization of the channel response specifically to thermal stimulation. SUMOylation is a post translational modification. Here the authors show that TRPV1, which conveys thermal nociception, is SUMOylated in DRGs in inflammatory conditions and contributes to pain behavior in mice.
Collapse
Affiliation(s)
- Yan Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingwei Gao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Quan Tian
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Qi Deng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yangbo Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tian Zhou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qiang Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Kaidi Mei
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Yingping Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Huiqing Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruining Ma
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuqiang Ding
- Department of Anatomy and Neurobiology, Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, 200092, Shanghai, China
| | - Weifang Rong
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jinke Cheng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing Yao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, China.
| | - Tian-Le Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
36
|
Noureldein M, Mardare R, Pickard J, Shing HL, Eisenhut M. Cerebrospinal fluid protein and glucose levels in neonates with a systemic inflammatory response without meningitis. Fluids Barriers CNS 2018. [PMID: 29540199 PMCID: PMC5853144 DOI: 10.1186/s12987-018-0095-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND It has been estimated that paediatric meningitis without elevated CSF white cell count (pleocytosis) accounts for 0.5-12% of all cases of bacterial meningitis. CSF protein and glucose measurements are therefore essential in management but may be neglected in clinical practice. In order to improve recognition of bacterial meningitis in neonates and to enable adequate management and audit, we investigated whether a systemic inflammatory response in the absence of meningitis is associated with elevated CSF protein and reduced CSF glucose levels. A further aim was to determine whether abnormal levels of these parameters were associated with increased incidence of neurological damage. METHODS As part of an audit into management of abnormal CSF findings in neonates, we conducted a retrospective analysis of neonates without meningitis as evident from normal CSF white blood cell counts and negative CSF culture. We compared data from neonates with fever (temperature > 38.0 °C) and/or elevated C-reactive protein (CRP) levels (> 5 mg/l) (possible sepsis) with data from neonates without fever or CRP elevation. RESULTS We analysed results from a total of 244 neonates. CSF protein levels were 0.89 g/l (SD 0.37) in neonates without fever or elevated CRP (n = 26) and not significantly different from neonates with possible sepsis (n = 218) with 0.92 g/l (SD 0.40). CSF glucose levels in infants with possible sepsis were 2.71 (SD 0.83) mmol/l and not significantly different from infants without sepsis with 2.55 mmol/l (SD 0.34). CONCLUSIONS CSF protein and glucose levels are not affected by a systemic inflammatory response syndrome if there is no meningitis.
Collapse
Affiliation(s)
- Mona Noureldein
- Luton & Dunstable University Hospital NHS Foundation Trust, Lewsey Road, Luton, LU4ODZ, UK
| | - Roxana Mardare
- Luton & Dunstable University Hospital NHS Foundation Trust, Lewsey Road, Luton, LU4ODZ, UK
| | - Jack Pickard
- Luton & Dunstable University Hospital NHS Foundation Trust, Lewsey Road, Luton, LU4ODZ, UK
| | - Hoi Lun Shing
- Luton & Dunstable University Hospital NHS Foundation Trust, Lewsey Road, Luton, LU4ODZ, UK
| | - Michael Eisenhut
- Luton & Dunstable University Hospital NHS Foundation Trust, Lewsey Road, Luton, LU4ODZ, UK.
| |
Collapse
|
37
|
Clinical Presentation, Management, and Prognostic Factors of Idiopathic Systemic Capillary Leak Syndrome: A Systematic Review. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2018; 6:609-618. [DOI: 10.1016/j.jaip.2017.07.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/14/2017] [Accepted: 07/25/2017] [Indexed: 12/28/2022]
|
38
|
Hamacher J, Hadizamani Y, Borgmann M, Mohaupt M, Männel DN, Moehrlen U, Lucas R, Stammberger U. Cytokine-Ion Channel Interactions in Pulmonary Inflammation. Front Immunol 2018; 8:1644. [PMID: 29354115 PMCID: PMC5758508 DOI: 10.3389/fimmu.2017.01644] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022] Open
Abstract
The lungs conceptually represent a sponge that is interposed in series in the bodies’ systemic circulation to take up oxygen and eliminate carbon dioxide. As such, it matches the huge surface areas of the alveolar epithelium to the pulmonary blood capillaries. The lung’s constant exposure to the exterior necessitates a competent immune system, as evidenced by the association of clinical immunodeficiencies with pulmonary infections. From the in utero to the postnatal and adult situation, there is an inherent vital need to manage alveolar fluid reabsorption, be it postnatally, or in case of hydrostatic or permeability edema. Whereas a wealth of literature exists on the physiological basis of fluid and solute reabsorption by ion channels and water pores, only sparse knowledge is available so far on pathological situations, such as in microbial infection, acute lung injury or acute respiratory distress syndrome, and in the pulmonary reimplantation response in transplanted lungs. The aim of this review is to discuss alveolar liquid clearance in a selection of lung injury models, thereby especially focusing on cytokines and mediators that modulate ion channels. Inflammation is characterized by complex and probably time-dependent co-signaling, interactions between the involved cell types, as well as by cell demise and barrier dysfunction, which may not uniquely determine a clinical picture. This review, therefore, aims to give integrative thoughts and wants to foster the unraveling of unmet needs in future research.
Collapse
Affiliation(s)
- Jürg Hamacher
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Internal Medicine V - Pneumology, Allergology, Respiratory and Environmental Medicine, Faculty of Medicine, Saarland University, Saarbrücken, Germany.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Yalda Hadizamani
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Michèle Borgmann
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Markus Mohaupt
- Internal Medicine, Sonnenhofspital Bern, Bern, Switzerland
| | | | - Ueli Moehrlen
- Paediatric Visceral Surgery, Universitäts-Kinderspital Zürich, Zürich, Switzerland
| | - Rudolf Lucas
- Department of Pharmacology and Toxicology, Vascular Biology Center, Medical College of Georgia, Augusta, GA, United States
| | - Uz Stammberger
- Lungen- und Atmungsstiftung Bern, Bern, Switzerland.,Novartis Institutes for Biomedical Research, Translational Clinical Oncology, Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
39
|
Hołyńska-Iwan I, Dziembowska I, Smyk P, Lampka M, Olszewska-Słonina D. Capsaicin Used on Skin Influences Ion Transport Pathways: An in vitro Study. Skin Pharmacol Physiol 2017; 31:19-27. [PMID: 29131139 DOI: 10.1159/000481689] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 09/15/2017] [Indexed: 01/07/2023]
Abstract
Acute, adverse skin effects to capsaicin can be activated by inhibition of sodium transport not only in nociceptive neurons, but also in keratinocytes. The aim of the current study was to describe and compare immediate (15 s) and prolonged (30 min) effects of capsaicin on epidermal (not neural) sodium transport using a rabbit skin model. Skin fragments (n = 169) were incubated in 4 conditions: undisturbed ion transport (U; n = 48); inhibited sodium transport (INa; n = 34) with amiloride used as sodium transport blocker; long-term irritation by capsaicin with undisturbed ion transport (CAPSA-U; n = 43) and with inhibited sodium transport (CAPSA-INa; n = 35). After 30 min of incubation, a solution of capsaicin was applied directly to the skin fragments. The study demonstrated that sodium transport inhibition eliminated the effects of both immediate and prolonged capsaicin application. The results could be the basis for future research considering selective sodium transport inhibitors for human skin to reduce the side effects of capsaicin, related to activation of sodium channels in keratinocytes.
Collapse
Affiliation(s)
- Iga Hołyńska-Iwan
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | | | | | | | | |
Collapse
|
40
|
Kim SH, Lee KM, Lee GS, Seong JW, Kang TJ. Rifampicin Alleviates Atopic Dermatitis-Like Response in vivo and in vitro. Biomol Ther (Seoul) 2017; 25:634-640. [PMID: 29081091 PMCID: PMC5685433 DOI: 10.4062/biomolther.2017.147] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/11/2017] [Accepted: 08/11/2017] [Indexed: 11/05/2022] Open
Abstract
Atopic dermatitis (AD) is a common inflammatory skin disorder mediated by inflammatory cells, such as macrophages and mast cells. Rifampicin is mainly used for the treatment of tuberculosis. Recently, it was reported that rifampicin has anti-inflammatory and immune-suppressive activities. In this study, we investigated the effect of rifampicin on atopic dermatitis in vivo and in vitro. AD was induced by treatment with 2, 4-dinitrochlorobenzene (DNCB) in NC/Nga mice. A subset of mice was then treated with rifampicin by oral administration. The severity score and scratching behavior were alleviated in the rifampicin-treated group. Serum immunoglobulin E (IgE) and interleukin-4 (IL-4) levels were also ameliorated in mice treated with rifampicin. We next examined whether rifampicin has anti-atopic activity via suppression of mast cell activation. Rifampicin suppressed the release of β-hexosaminidase and histamine from human mast cell (HMC)-1 cultures stimulated with compound 48/80. Treatment with rifampicin also inhibited secretion of inflammatory mediators, such tumor necrosis factor-α (TNF-α) and prostaglandin D₂ (PGD₂), in mast cells activated by compound 48/80. The mRNA expression of cyclooxygenase 2 (COX-2) was reduced in the cells treated with rifampicin in a concentration-dependent manner. These results suggest that rifampicin can be used to treat atopic dermatitis.
Collapse
Affiliation(s)
- Seung Hyun Kim
- Institute of Chronic Disease and College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Ki Man Lee
- Institute of Chronic Disease and College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Geum Seon Lee
- Institute of Chronic Disease and College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Ju-Won Seong
- Institute of Chronic Disease and College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Tae Jin Kang
- Institute of Chronic Disease and College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
41
|
Thurber AE, Nelson M, Frost CL, Levin M, Brackenbury WJ, Kaplan DL. IK channel activation increases tumor growth and induces differential behavioral responses in two breast epithelial cell lines. Oncotarget 2017; 8:42382-42397. [PMID: 28415575 PMCID: PMC5522074 DOI: 10.18632/oncotarget.16389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 03/08/2017] [Indexed: 12/26/2022] Open
Abstract
Many potassium channel families are over-expressed in cancer, but their mechanistic role in disease progression is poorly understood. Potassium channels modulate membrane potential (Vmem) and thereby influence calcium ion dynamics and other voltage-sensitive signaling mechanisms, potentially acting as transcriptional regulators. This study investigated the differential response to over-expression and activation of a cancer-associated potassium channel, the intermediate conductance calcium-activated potassium channel (IK), on aggressive behaviors in mammary epithelial and breast cancer cell lines. IK was over-expressed in the highly metastatic breast cancer cell line MDA-MB-231 and the spontaneously immortalized breast epithelial cell line MCF-10A, and the effect on cancer-associated behaviors was assessed. IK over-expression increased primary tumor growth and metastasis of MDA-MB-231 in orthotopic xenografts, demonstrating for the first time in any cancer type that increased IK is sufficient to promote cancer aggression. The primary tumors had similar vascularization as determined by CD31 staining and similar histological characteristics. Interestingly, despite the increased in vivo growth and metastasis, neither IK over-expression nor activation with agonist had a significant effect on MDA-MB-231 proliferation, invasion, or migration in vitro. In contrast, IK decreased MCF-10A proliferation and invasion through Matrigel but had no effect on migration in a scratch-wound assay. We conclude that IK activity is sufficient to promote cell aggression in vivo. Our data provide novel evidence supporting IK and downstream signaling networks as potential targets for cancer therapies.
Collapse
Affiliation(s)
- Amy E. Thurber
- Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Michaela Nelson
- Department of Biology, University of York, Heslington, York, UK
| | | | - Michael Levin
- Biology Department, and Tufts Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts, USA
| | | | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
42
|
Scheraga RG, Southern BD, Grove LM, Olman MA. The Role of Transient Receptor Potential Vanilloid 4 in Pulmonary Inflammatory Diseases. Front Immunol 2017; 8:503. [PMID: 28523001 PMCID: PMC5415870 DOI: 10.3389/fimmu.2017.00503] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/12/2017] [Indexed: 01/01/2023] Open
Abstract
Ion channels/pumps are essential regulators of organ homeostasis and disease. In the present review, we discuss the role of the mechanosensitive cation channel, transient receptor potential vanilloid 4 (TRPV4), in cytokine secretion and pulmonary inflammatory diseases such as asthma, cystic fibrosis (CF), and acute lung injury/acute respiratory distress syndrome (ARDS). TRPV4 has been shown to play a role in lung diseases associated with lung parenchymal stretch or stiffness. TRPV4 indirectly mediates hypotonicity-induced smooth muscle contraction and airway remodeling in asthma. Further, the literature suggests that in CF TRPV4 may improve ciliary beat frequency enhancing mucociliary clearance, while at the same time increasing pro-inflammatory cytokine secretion/lung tissue injury. Currently it is understood that the role of TRPV4 in immune cell function and associated lung tissue injury/ARDS may depend on the injury stimulus. Uncovering the downstream mechanisms of TRPV4 action in pulmonary inflammatory diseases is likely important to understanding disease pathogenesis and may lead to novel therapeutics.
Collapse
Affiliation(s)
- Rachel G Scheraga
- Cleveland Clinic, Department of Pathobiology, Lerner Research Institute, Cleveland, OH, USA
| | - Brian D Southern
- Cleveland Clinic, Department of Pathobiology, Lerner Research Institute, Cleveland, OH, USA
| | - Lisa M Grove
- Cleveland Clinic, Department of Pathobiology, Lerner Research Institute, Cleveland, OH, USA
| | - Mitchell A Olman
- Cleveland Clinic, Department of Pathobiology, Lerner Research Institute, Cleveland, OH, USA
| |
Collapse
|
43
|
Kim YY, Je IG, Kim MJ, Kang BC, Choi YA, Baek MC, Lee B, Choi JK, Park HR, Shin TY, Lee S, Yoon SB, Lee SR, Khang D, Kim SH. 2-Hydroxy-3-methoxybenzoic acid attenuates mast cell-mediated allergic reaction in mice via modulation of the FcεRI signaling pathway. Acta Pharmacol Sin 2017; 38:90-99. [PMID: 27890918 DOI: 10.1038/aps.2016.112] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/23/2016] [Indexed: 12/18/2022]
Abstract
Mast cells are important effector cells in immunoglobulin (Ig) E-mediated allergic reactions such as asthma, atopic dermatitis and rhinitis. Vanillic acid, a natural product, has shown anti-oxidant and anti-inflammatory activities. In the present study, we investigated the anti-allergic inflammatory effects of ortho-vanillic acid (2-hydroxy-3-methoxybenzoic acid, o-VA) that was a derivative of vanillic acid isolated from Amomum xanthioides. In mouse anaphylaxis models, oral administration of o-VA (2, 10, 50 mg/kg) dose-dependently attenuated ovalbumin-induced active systemic anaphylaxis and IgE-mediated cutaneous allergic reactions such as hypothermia, histamine release, IgE production and vasodilation; administration of o-VA also suppressed the mast cell degranulator compound 48/80-induced anaphylaxis. In cultured mast cell line RBL-2H3 and isolated rat peritoneal mast cells in vitro, pretreatment with o-VA (1-100 μmol/L) dose-dependently inhibited DNP-HSA-induced degranulation of mast cells by decreasing the intracellular free calcium level, and suppressed the expression of pro-inflammatory cytokines TNF-α and IL-4. Pretreatment of RBL-2H3 cells with o-VA suppressed DNP-HSA-induced phosphorylation of Lyn, Syk, Akt, and the nuclear translocation of nuclear factor-κB. In conclusion, o-VA suppresses the mast cell-mediated allergic inflammatory response by blocking the signaling pathways downstream of high affinity IgE receptor (FcεRI) on the surface of mast cells.
Collapse
|
44
|
Gerdle B, Ernberg M, Mannerkorpi K, Larsson B, Kosek E, Christidis N, Ghafouri B. Increased Interstitial Concentrations of Glutamate and Pyruvate in Vastus Lateralis of Women with Fibromyalgia Syndrome Are Normalized after an Exercise Intervention - A Case-Control Study. PLoS One 2016; 11:e0162010. [PMID: 27695113 PMCID: PMC5047648 DOI: 10.1371/journal.pone.0162010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 08/12/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Fibromyalgia syndrome (FMS) is associated with central alterations, but controversies exist regarding the presence and role of peripheral factors. Microdialysis (MD) can be used in vivo to study muscle alterations in FMS. Furthermore for chronic pain conditions such as FMS, the mechanisms for the positive effects of exercise are unclear. This study investigates the interstitial concentrations of algesics and metabolites in the vastus lateralis muscle of 29 women with FMS and 28 healthy women before and after an exercise intervention. METHODS All the participants went through a clinical examination and completed a questionnaire. In addition, their pressure pain thresholds (PPTs) in their upper and lower extremities were determined. For both groups, MD was conducted in the vastus lateralis muscle before and after a 15-week exercise intervention of mainly resistance training of the lower limbs. Muscle blood flow and interstitial muscle concentrations of lactate, pyruvate, glutamate, glucose, and glycerol were determined. RESULTS FMS was associated with significantly increased interstitial concentrations of glutamate, pyruvate, and lactate. After the exercise intervention, the FMS group exhibited significant decreases in pain intensity and in mean interstitial concentrations of glutamate, pyruvate, and glucose. The decrease in pain intensity in FMS correlated significantly with the decreases in pyruvate and glucose. In addition, the FMS group increased their strength and endurance. CONCLUSION This study supports the suggestion that peripheral metabolic and algesic muscle alterations are present in FMS patients and that these alterations contribute to pain. After an exercise intervention, alterations normalized, pain intensity decreased (but not abolished), and strength and endurance improved, all findings that suggest the effects of exercise are partially peripheral.
Collapse
Affiliation(s)
- Björn Gerdle
- Pain and Rehabilitation Centre, and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Malin Ernberg
- Karolinska Institute, Department of Dental Medicine, Section of Orofacial Pain and Jaw Function and Scandinavian Centre for Orofacial Neuroscience (SCON), Stockholm, Sweden
| | - Kaisa Mannerkorpi
- Section of Physiotherapy, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- University of Gothenburg Centre for Person-Centred Care (GPCC), Sahlgrenska Academy, Gothenburg, Sweden
| | - Britt Larsson
- Pain and Rehabilitation Centre, and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Eva Kosek
- Department of Clinical Neuroscience and Osher Centre for Integrative Medicine, Karolinska Institute, Stockholm, Sweden
| | - Nikolaos Christidis
- Karolinska Institute, Department of Dental Medicine, Section of Orofacial Pain and Jaw Function and Scandinavian Centre for Orofacial Neuroscience (SCON), Stockholm, Sweden
| | - Bijar Ghafouri
- Pain and Rehabilitation Centre, and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
45
|
Epithelial Anion Transport as Modulator of Chemokine Signaling. Mediators Inflamm 2016; 2016:7596531. [PMID: 27382190 PMCID: PMC4921137 DOI: 10.1155/2016/7596531] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 05/03/2016] [Accepted: 05/12/2016] [Indexed: 12/16/2022] Open
Abstract
The pivotal role of epithelial cells is to secrete and absorb ions and water in order to allow the formation of a luminal fluid compartment that is fundamental for the epithelial function as a barrier against environmental factors. Importantly, epithelial cells also take part in the innate immune system. As a first line of defense they detect pathogens and react by secreting and responding to chemokines and cytokines, thus aggravating immune responses or resolving inflammatory states. Loss of epithelial anion transport is well documented in a variety of diseases including cystic fibrosis, chronic obstructive pulmonary disease, asthma, pancreatitis, and cholestatic liver disease. Here we review the effect of aberrant anion secretion with focus on the release of inflammatory mediators by epithelial cells and discuss putative mechanisms linking these transport defects to the augmented epithelial release of chemokines and cytokines. These mechanisms may contribute to the excessive and persistent inflammation in many respiratory and gastrointestinal diseases.
Collapse
|
46
|
Abstract
Activation of ion channels and pores are essential steps during regulated cell death. Channels and pores participate in execution of apoptosis, necroptosis and other forms of caspase-independent cell death. Within the program of regulated cell death, these channels are strategically located. Ion channels can shrink cells and drive them towards apoptosis, resulting in silent, i.e. immunologically unrecognized cell death. Alternatively, activation of channels can induce cell swelling, disintegration of the cell membrane, and highly immunogenic necrotic cell death. The underlying cell death pathways are not strictly separated as identical stimuli may induce cell shrinkage and apoptosis when applied at low strength, but may also cause cell swelling at pronounced stimulation, resulting in regulated necrosis. Nevertheless, the precise role of ion channels during regulated cell death is far from being understood, as identical channels may support regulated death in some cell types, but may cause cell proliferation, cancer development, and metastasis in others. Along this line, the phospholipid scramblase and Cl(-)/nonselective channel anoctamin 6 (ANO6) shows interesting features, as it participates in apoptotic cell death during lower levels of activation, thereby inducing cell shrinkage. At strong activation, e.g. by stimulation of purinergic P2Y7 receptors, it participates in pore formation, causes massive membrane blebbing, cell swelling, and membrane disintegration. The LRRC8 proteins deserve much attention as they were found to have a major role in volume regulation, apoptotic cell shrinkage and resistance towards anticancer drugs.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany.
| |
Collapse
|
47
|
Metabolic Connection of Inflammatory Pain: Pivotal Role of a Pyruvate Dehydrogenase Kinase-Pyruvate Dehydrogenase-Lactic Acid Axis. J Neurosci 2016; 35:14353-69. [PMID: 26490872 DOI: 10.1523/jneurosci.1910-15.2015] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED Pyruvate dehydrogenase kinases (PDK1-4) are mitochondrial metabolic regulators that serve as decision makers via modulation of pyruvate dehydrogenase (PDH) activity to convert pyruvate either aerobically to acetyl-CoA or anaerobically to lactate. Metabolic dysregulation and inflammatory processes are two sides of the same coin in several pathophysiological conditions. The lactic acid surge associated with the metabolic shift has been implicated in diverse painful states. In this study, we investigated the role of PDK-PDH-lactic acid axis in the pathogenesis of chronic inflammatory pain. Deficiency of Pdk2 and/or Pdk4 in mice attenuated complete Freund's adjuvant (CFA)-induced pain hypersensitivities. Likewise, Pdk2/4 deficiency attenuated the localized lactic acid surge along with hallmarks of peripheral and central inflammation following intraplantar administration of CFA. In vitro studies supported the role of PDK2/4 as promoters of classical proinflammatory activation of macrophages. Moreover, the pharmacological inhibition of PDKs or lactic acid production diminished CFA-induced inflammation and pain hypersensitivities. Thus, a PDK-PDH-lactic acid axis seems to mediate inflammation-driven chronic pain, establishing a connection between metabolism and inflammatory pain. SIGNIFICANCE STATEMENT The mitochondrial pyruvate dehydrogenase (PDH) kinases (PDKs) and their substrate PDH orchestrate the conversion of pyruvate either aerobically to acetyl-CoA or anaerobically to lactate. Lactate, the predominant end product of glycolysis, has recently been identified as a signaling molecule for neuron-glia interactions and neuronal plasticity. Pathological metabolic shift and subsequent lactic acid production are thought to play an important role in diverse painful states; however, their contribution to inflammation-driven pain is still to be comprehended. Here, we report that the PDK-PDH-lactic acid axis constitutes a key component of inflammatory pain pathogenesis. Our findings establish an unanticipated link between metabolism and inflammatory pain. This study unlocks a previously ill-explored research avenue for the metabolic control of inflammatory pain pathogenesis.
Collapse
|
48
|
Son A, Park S, Shin DM, Muallem S. Orai1 and STIM1 in ER/PM junctions: roles in pancreatic cell function and dysfunction. Am J Physiol Cell Physiol 2016; 310:C414-22. [PMID: 26739495 DOI: 10.1152/ajpcell.00349.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Membrane contact sites (MCS) are critical junctions that form between the endoplasmic reticulum (ER) and membranes of various organelles, including the plasma membrane (PM). Signaling complexes, including mediators of Ca(2+) signaling, are assembled within MCS, such as the ER/PM junction. This is most evident in polarized epithelial cells, such as pancreatic cells. Core Ca(2+) signaling proteins cluster at the apical pole, the site of inositol 1,4,5-trisphosphate-mediated Ca(2+) release and Orai1/transient receptor potential canonical-mediated store-dependent Ca(2+) entry. Recent advances have characterized the proteins that tether the membranes at MCS and the role of these proteins in modulating physiological and pathological intracellular signaling. This review discusses recent advances in the characterization of Ca(2+) signaling at ER/PM junctions and the relation of these junctions to physiological and pathological Ca(2+) signaling in pancreatic acini.
Collapse
Affiliation(s)
- Aran Son
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Seonghee Park
- Department of Physiology, School of Medicine, Ewha Womans University, Seoul, Korea
| | - Dong Min Shin
- Department of Oral Biology, BK 21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Shmuel Muallem
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland;
| |
Collapse
|
49
|
How Hypoxia Can Influence Ion Transport. Pediatr Crit Care Med 2016; 17:90-2. [PMID: 26731324 DOI: 10.1097/pcc.0000000000000576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
50
|
Baron A, Lingueglia E. Pharmacology of acid-sensing ion channels – Physiological and therapeutical perspectives. Neuropharmacology 2015; 94:19-35. [DOI: 10.1016/j.neuropharm.2015.01.005] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 12/15/2014] [Accepted: 01/07/2015] [Indexed: 12/29/2022]
|