1
|
Park SI, Hwang S, Lee Y, Lee HY, Kim S, Hong J, Jo SH, Choi SY. Chlorpromazine directly inhibits Kv1.3 channels by facilitating the inactivation of channels. Mol Brain 2025; 18:41. [PMID: 40340862 PMCID: PMC12063219 DOI: 10.1186/s13041-025-01211-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/16/2025] [Indexed: 05/10/2025] Open
Abstract
Kv1.3 channels in microglia are pivotal in regulating neuroinflammation. The antipsychotic chlorpromazine (CPZ) demonstrates anti-inflammatory effects by decreasing Kv1.3 activity in mPFC microglia. However, the precise mechanism of CPZ's effect in the mPFC remains unclear, given that CPZ is known to inhibit dopamine receptors and the mPFC contains various cell types with dopamine receptors. In this study, we investigate how CPZ inhibits Kv1.3 channels using human Kv1.3 channel-expressing Xenopus laevis oocytes. CPZ directly inhibits Kv1.3 channel currents in a concentration-dependent manner. The CPZ-mediated Kv1.3 channel inhibition is not voltage-dependent, and CPZ accelerates Kv1.3 channel inactivation without significantly affecting its activation. Our findings suggest that CPZ directly blocks Kv1.3 channels without involving other ion channels or receptors, including dopamine receptors, thereby contributing to the understanding of its neuroinflammation-suppressing mechanism.
Collapse
Affiliation(s)
- Seo-In Park
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, Republic of Korea
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, Republic of Korea
| | - Soobeen Hwang
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, Republic of Korea
| | - Young Lee
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, Republic of Korea
| | - Hee-Yoon Lee
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, Republic of Korea
| | - Soohyun Kim
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, Republic of Korea
| | - Junseo Hong
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, Republic of Korea
| | - Su-Hyun Jo
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, Republic of Korea.
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, Republic of Korea.
| |
Collapse
|
2
|
Lee HY, Lee Y, Chung C, Park SI, Shin HJ, Joe EH, Lee SJ, Kim DW, Jo SH, Choi SY. The antipsychotic chlorpromazine reduces neuroinflammation by inhibiting microglial voltage-gated potassium channels. Glia 2025; 73:210-227. [PMID: 39435609 DOI: 10.1002/glia.24629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024]
Abstract
Neuroinflammation, the result of microglial activation, is associated with the pathogenesis of a wide range of psychiatric and neurological disorders. Recently, chlorpromazine (CPZ), a dopaminergic D2 receptor antagonist and schizophrenia therapy, was proposed to exert antiinflammatory effects in the central nervous system. Here, we report that the expression of Kv1.3 channel, which is abundant in T cells, is upregulated in microglia upon infection, and that CPZ specifically inhibits these channels to reduce neuroinflammation. In the mouse medial prefrontal cortex, we show that CPZ lessens Kv1.3 channel activity and reduces proinflammatory cytokine production. In mice treated with LPS, we found that CPZ was capable of alleviating both neuroinflammation and depression-like behavior. Our findings suggest that CPZ acts as a microglial Kv1.3 channel inhibitor and neuroinflammation modulator, thereby exerting therapeutic effects in neuroinflammatory psychiatric/neurological disorders.
Collapse
Affiliation(s)
- Hee-Yoon Lee
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Young Lee
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Chaelin Chung
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Seo-In Park
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Hyo Jung Shin
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Eun-Hye Joe
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sung Joong Lee
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Dong Woon Kim
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
- Department of Oral Anatomy & Developmental Biology, Kyung Hee University College of Dentistry, Seoul, Republic of Korea
| | - Su-Hyun Jo
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| |
Collapse
|
3
|
Wong BHS, Shim H, Goay SSM, Ong ST, Muhammad Taib NAB, Chai KXY, Lim K, Huang D, Ong CK, Vaiyapuri TS, Cheah YC, Wang Y, Wulff H, Webster RD, Shelat VG, Verma NK. The novel quinoline derivative SKA-346 as a K Ca3.1 channel selective activator. RSC Adv 2024; 14:38364-38377. [PMID: 39635364 PMCID: PMC11615718 DOI: 10.1039/d4ra07330d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/17/2024] [Indexed: 12/07/2024] Open
Abstract
The calcium-activated KCa3.1 channel plays a crucial role in T-cell immune response. Genetic manipulation of T-cells to upregulate the expression of K+ channels has been shown to boost T-cell cytotoxicity in cancer. Here, we aimed to identify and characterize an activator that would augment KCa3.1 currents without affecting other channels. We synthesized five quinoline derivatives and used electrophysiology to screen them on KCa3.1 and a panel of 14 other ion channels. One quinoline derivative, SKA-346, activated KCa3.1 with an EC50 of 1.9 μM and showed selectivity against the other channels. In silico analysis using RosettaLigand and GLIDE demonstrated a well-converged pose of SKA-346 in a binding pocket at the interface between the calmodulin N-lobe and the S45A helix in the S4-S5 linker of the KCa3.1 channel. SKA-346 (30 mg kg-1), tolerated by mice after intra-peritoneal administration, exhibited a peak plasma concentration of 6.29 μg mL-1 (29.2 μM) at 15 min and a circulating half-life (t 1/2) of 2.8 h. SKA-346 could serve as a template for the development of more potent KCa3.1 activators to enhance T-cell cytotoxicity in cancer.
Collapse
Affiliation(s)
- Brandon Han Siang Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- Interdisciplinary Graduate Programme, NTU Institute for Health Technologies (HealthTech NTU), Nanyang Technological University Singapore Singapore
| | - Heesung Shim
- Physical and Life Sciences, Lawrence Livermore National Laboratory Livermore CA USA
| | - Stephanie Shee Min Goay
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- LKCMedicine-ICE Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Seow Theng Ong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- LKCMedicine-ICE Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Nur Ayuni Binte Muhammad Taib
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
| | - Kelila Xin Ye Chai
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
| | - Kerry Lim
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
| | - Dachuan Huang
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
- Duke-NUS Medical School Singapore
| | - Choon Kiat Ong
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
- Duke-NUS Medical School Singapore
| | | | - Yeong Cheng Cheah
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Yulan Wang
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Heike Wulff
- Department of Pharmacology, University of California Davis CA USA
| | - Richard D Webster
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University Singapore Singapore
| | - Vishalkumar G Shelat
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- Department of General Surgery, Tan Tock Seng Hospital Singapore
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| |
Collapse
|
4
|
Sharma P, Guo A, Poudel S, Boada-Romero E, Verbist KC, Palacios G, Immadisetty K, Chen MJ, Haydar D, Mishra A, Peng J, Babu MM, Krenciute G, Glazer ES, Green DR. An early, novel arginine methylation of KCa3.1 attenuates subsequent T cell exhaustion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593421. [PMID: 38798680 PMCID: PMC11118966 DOI: 10.1101/2024.05.09.593421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
T cell receptor (TCR) engagement initiates the activation process, and this signaling event is regulated in multifaceted ways. Nutrient availability in the immediate niche is one such mode of regulation 1-3 . Here, we investigated how the availability of an essential amino acid methionine (Met) and TCR signaling might interplay in the earliest events of T cell activation to affect subsequent T cell fate and function. We found that limiting Met during only the initial 30 minutes of CD8 + T cell activation increased Ca 2+ influx, Ca 2+ -mediated NFAT1 ( Nfatc2 ) activation, NFAT1 promoter occupancy, and T cell exhaustion. We identified changes in the protein arginine methylome during the initial 30 min of TCR engagement and discovered a novel arginine methylation of a Ca 2+ -activated potassium transporter, KCa3.1, which regulates Ca 2+ -mediated NFAT1 signaling to ensure optimal activation. Ablation of arginine methylation in KCa3.1 led to increased NFAT1 activation, rendering T cells dysfunctional in murine tumour and infection models. Furthermore, acute Met supplementation at early stages reduced nuclear NFAT1 in tumour-infiltrating T cells and augmented their anti-tumour activity. Our findings identify a metabolic event occurring early after T cell activation that influences the subsequent fate of the cell.
Collapse
|
5
|
Tobar CGR, Urmendiz YDMM, Vallejo MA, Manquillo DF, Castaño VEN, Caicedo AIO, Tobar LLM, Vargas JAG, Cuellar RAD. Immunomodulatory effect of Tityus sp. in mononuclear cells extracted from the blood of rheumatoid arthritis patients. J Venom Anim Toxins Incl Trop Dis 2024; 30:e20230064. [PMID: 39445068 PMCID: PMC11498904 DOI: 10.1590/1678-9199-jvatitd-2023-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 06/10/2024] [Indexed: 10/25/2024] Open
Abstract
Background Pathophysiological mechanisms of rheumatoid arthritis arise because of a proinflammatory environment, generated by the interaction of autoreactive lymphocytes and proinflammatory mediators. Current strategies to mitigate the progression of the disease produce adverse effects, so there is a need for new therapeutic strategies and molecular targets to treat this disease. In this context, evidence suggests that scorpion venoms could modulate the immune response and some important cellular mechanisms of pharmacological interest. To evaluate the immunomodulatory effect of the venom of Tityus sp. (a possible new species close to Tityus metuendus) peripheral blood mononuclear cells of women diagnosed with RA were compared to cells of a control group. Methods A case-control study was conducted with a sample of 10 women with a confirmed diagnosis of RA and controls matched by sex and age. The cytotoxicity of the venom was evaluated to find sublethal concentrations of the venom, and subsequently, their immunomodulatory capacity in terms of percentage of proliferation, cell activation, and cytokines production. Results the venom of Tityus sp. produced a decrease in the percentage of proliferation in the CD3+, CD3+CD4+, and CD3+CD8+ cell subpopulations of RA patients and healthy controls, at concentrations of 252 and 126 µg/mL. However, the venom did not induce significant differences in the percentage of cell activation markers. The venom caused a decrease in IL-10 at a concentration of 252 µg/mL compared to untreated cells from patients and controls. The remaining cytokines did not show significant differences. Conclusion the venom of Tityus sp. is a potential source of molecules with immunomodulatory ability in CD4 and CD8 T lymphocytes. This result directs venom characterization studies to identify pharmacological targets with immunomodulatory capacity in T lymphocytes to enhance research in the treatment of autoimmune disorders such as RA.
Collapse
Affiliation(s)
- Cindy Gabriela Rivera Tobar
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| | - Yisel del Mar Morales Urmendiz
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| | - Marcela Alejandra Vallejo
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| | - Diego Felipe Manquillo
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| | - Victoria Eugenia Niño Castaño
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| | - Ana Isabel Ospina Caicedo
- Grupo de Investigación en Salud, Programa de Medicina, Departamento
de Medicina Interna, Facultad de Ciencias de la Salud, Universidad del Cauca,
Popayán, Colombia
| | - Leydy Lorena Mendoza Tobar
- Grupo de Investigaciones Herpetológicas y Toxinológicas (GIHT),
Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la Educación,
Universidad del Cauca, Popayán, Colombia
| | - Jimmy Alexander Guerrero Vargas
- Grupo de Investigaciones Herpetológicas y Toxinológicas (GIHT),
Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la Educación,
Universidad del Cauca, Popayán, Colombia
- Grupo de Investigaciones Herpetológicas y Toxinológicas (GIHT),
Centro de Investigaciones Biomédicas - Bioterio (CIBUC-Bioterio), Museo de Historia
Natural, Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la
Educación, Universidad del Cauca, Popayán, Colombia
| | - Rosa Amalia Dueñas Cuellar
- Grupo de Investigación en Inmunología y Enfermedades Infecciosas,
Programa de Medicina, Departamento de Patología, Facultad de Ciencias de la Salud,
Universidad del Cauca, Popayán, Colombia
| |
Collapse
|
6
|
Lee JM, Kim J, Park SJ, Nam JH, Kim HJ, Kim WK. Regulation of T Lymphocyte Functions through Calcium Signaling Modulation by Nootkatone. Int J Mol Sci 2024; 25:5240. [PMID: 38791278 PMCID: PMC11121628 DOI: 10.3390/ijms25105240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Recent advancements in understanding the intricate molecular mechanisms underlying immunological responses have underscored the critical involvement of ion channels in regulating calcium influx, particularly in inflammation. Nootkatone, a natural sesquiterpenoid found in Alpinia oxyphylla and various citrus species, has gained attention for its diverse pharmacological properties, including anti-inflammatory effects. This study aimed to elucidate the potential of nootkatone in modulating ion channels associated with calcium signaling, particularly CRAC, KV1.3, and KCa3.1 channels, which play pivotal roles in immune cell activation and proliferation. Using electrophysiological techniques, we demonstrated the inhibitory effects of nootkatone on CRAC, KV1.3, and KCa3.1 channels in HEK293T cells overexpressing respective channel proteins. Nootkatone exhibited dose-dependent inhibition of channel currents, with IC50 values determined for each channel. Nootkatone treatment did not significantly affect cell viability, indicating its potential safety for therapeutic applications. Furthermore, we observed that nootkatone treatment attenuated calcium influx through activated CRAC channels and showed anti-proliferative effects, suggesting its role in regulating inflammatory T cell activation. These findings highlight the potential of nootkatone as a natural compound for modulating calcium signaling pathways by targeting related key ion channels and it holds promise as a novel therapeutic agent for inflammatory disorders.
Collapse
Affiliation(s)
- Ji Min Lee
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Gyeongsangbuk-do, Republic of Korea; (J.M.L.); (S.J.P.); (J.H.N.)
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
| | - Jintae Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
| | - Su Jin Park
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Gyeongsangbuk-do, Republic of Korea; (J.M.L.); (S.J.P.); (J.H.N.)
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Gyeongsangbuk-do, Republic of Korea; (J.M.L.); (S.J.P.); (J.H.N.)
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
| | - Hyun Jong Kim
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Gyeongsangbuk-do, Republic of Korea; (J.M.L.); (S.J.P.); (J.H.N.)
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
- Department of Internal Medicine, Graduate School of Medicine, Dongguk University, 27 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea
| |
Collapse
|
7
|
More NE, Mandlik R, Zine S, Gawali VS, Godad AP. Exploring the therapeutic opportunities of potassium channels for the treatment of rheumatoid arthritis. Front Pharmacol 2024; 15:1286069. [PMID: 38783950 PMCID: PMC11111972 DOI: 10.3389/fphar.2024.1286069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/18/2024] [Indexed: 05/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that affects the synovial joint, which leads to inflammation, loss of function, joint destruction, and disability. The disease biology of RA involves complex interactions between genetic and environmental factors and is strongly associated with various immune cells, and each of the cell types contributes differently to disease pathogenesis. Several immunomodulatory molecules, such as cytokines, are secreted from the immune cells and intervene in the pathogenesis of RA. In immune cells, membrane proteins such as ion channels and transporters mediate the transport of charged ions to regulate intracellular signaling pathways. Ion channels control the membrane potential and effector functions such as cytotoxic activity. Moreover, clinical studies investigating patients with mutations and alterations in ion channels and transporters revealed their importance in effective immune responses. Recent studies have shown that voltage-gated potassium channels and calcium-activated potassium channels and their subtypes are involved in the regulation of immune cells and RA. Due to the role of these channels in the pathogenesis of RA and from multiple pieces of clinical evidence, they can be considered therapeutic targets for the treatment of RA. Here, we describe the role of voltage-gated and calcium-activated potassium channels and their subtypes in RA and their pharmacological application as drug targets.
Collapse
Affiliation(s)
| | - Rahul Mandlik
- Medical Affairs, Shalina Healthcare DMCC, Dubai, United Arab Emirates
| | - Sandip Zine
- SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | | | - Angel Pavalu Godad
- SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| |
Collapse
|
8
|
Chen Y, Liu H, Yan Y, Chen H, Ye S, Qiu F, Liang CL, Zhang Q, Zheng F, Han L, Lu C, Dai Z. Methotrexate and electrostimulation cooperate to alleviate the relapse of psoriasiform skin inflammation by suppressing memory T cells. Biochem Pharmacol 2024; 219:115979. [PMID: 38081367 DOI: 10.1016/j.bcp.2023.115979] [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: 09/07/2023] [Revised: 11/24/2023] [Accepted: 12/08/2023] [Indexed: 12/26/2023]
Abstract
Methotrexate (MTX) is an immunosuppressant used to treat autoimmune diseases, including psoriasis. However, like other immunosuppressants, MTX alone does not prevent their recurrence. Electrostimulation (ES) has been utilized to treat some inflammatory disorders without any major side-effect. But it remains unknown if ES alone, or together with MTX, ameliorates autoimmune disease relapse: a sticky medical problem. In particular, the mechanisms underlying ES action remain unclear. The objective of this study was to determine an impact of ES and/or MTX on psoriasis relapse and their potential cooperation. We found that regional ES, but not MTX, ameliorated psoriasiform skin inflammation recurrence. Interestingly, treatment with both MTX and ES further prevented psoriasis recurrence compared to ES alone. Moreover, ES downregulated potassium channel Kv1.3 on T-cells and reduced CD4+/CD8+ effector memory (TEM) and CD8+ skin-resident memory T (TRM) cells, while ES plus MTX further decreased CD8+ TEM/TRM cells compared to ES alone. However, ES failed to further attenuate psoriasis recurrence or suppress T cell memory in Kv1.3-deficient mice, whereas lack of Kv1.3 itself ameliorated psoriasis relapse by shrinking T cell memory pool. Importantly, ES moderately inhibited T-cell proliferation in vitro. ES also reduced human CD8+ TRM cells and attenuated human skin lesions in humanized mice grafted with lesional skin from patients with recurrent psoriasis, with an enhanced efficacy in mice treated with both ES and MTX. Thus, ES and MTX cooperated to prevent psoriasis relapse by reducing T-cell memory via targeting potassium channel Kv1.3. Our studies may be implicated for treating human psoriasis.
Collapse
Affiliation(s)
- Yuchao Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Huazhen Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Yuhong Yan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Haiming Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Shuyan Ye
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Feifei Qiu
- Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Chun-Ling Liang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Qunfang Zhang
- Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Fang Zheng
- Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Ling Han
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Chuanjian Lu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China.
| | - Zhenhua Dai
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
9
|
Yuan X, Han S, Manyande A, Gao F, Wang J, Zhang W, Tian X. Spinal voltage-gated potassium channel Kv1.3 contributes to neuropathic pain via the promotion of microglial M1 polarization and activation of the NLRP3 inflammasome. Eur J Pain 2023; 27:289-302. [PMID: 36440534 DOI: 10.1002/ejp.2059] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/06/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUD Studies have shown that the activation of microglia is the main mechanism of neuropathic pain. Kv1.3 channel is a novel therapeutic target for treating neuroinflammatory disorders due to its crucial role in subsets of microglial cells. As such, it may be involved in the processes of neuropathic pain, however, whether Kv1.3 plays a role in neuroinflammation following peripheral nerve injury is unclear. METHOD The spared nerve injury model (SNI) was used to establish neuropathic pain. Western blot and immunofluorescence were used to examine the effect of Kv1.3 in the SNI rats. PAP-1, a Kv1.3 specific blocker was administered to alleviate neuropathic pain in the SNI rats. RESULTS Neuropathic pain and allodynia occurred after SNI, the levels of M1 (CD68, iNos) and M2 (CD206, Arg-1) phenotypes were up-regulated in the spinal cord, and the protein levels of NLRP3, caspase-1 and IL-1β were also increased. Pharmacological blocking of Kv1.3 with PAP-1 alleviated hyperpathia induced by SNI. Meanwhile, intrathecal injection of PAP-1 reduced M1 polarization and decreased NLRP3, caspase-1 and IL-1β expressions of protein levels. CONCLUSION Our research indicates that the Kv1.3 channel in the spinal cord contributes to neuropathic pain by promoting microglial M1 polarization and activating the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Xiaoman Yuan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Siyi Han
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, UK
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Wen Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Xuebi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| |
Collapse
|
10
|
Bioactive peptides from scorpion venoms: therapeutic scaffolds and pharmacological tools. Chin J Nat Med 2023; 21:19-35. [PMID: 36641229 DOI: 10.1016/s1875-5364(23)60382-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Indexed: 01/14/2023]
Abstract
Evolution and natural selection have endowed animal venoms, including scorpion venoms, with a wide range of pharmacological properties. Consequently, scorpions, their venoms, and/or their body parts have been used since time immemorial in traditional medicines, especially in Africa and Asia. With respect to their pharmacological potential, bioactive peptides from scorpion venoms have become an important source of scientific research. With the rapid increase in the characterization of various components from scorpion venoms, a large number of peptides are identified with an aim of combating a myriad of emerging global health problems. Moreover, some scorpion venom-derived peptides have been established as potential scaffolds helpful for drug development. In this review, we summarize the promising scorpion venoms-derived peptides as drug candidates. Accordingly, we highlight the data and knowledge needed for continuous characterization and development of additional natural peptides from scorpion venoms, as potential drugs that can treat related diseases.
Collapse
|
11
|
Stopschinski BE, Weideman RA, McMahan D, Jacob DA, Little BB, Chiang HS, Saez Calveras N, Stuve O. Microglia as a cellular target of diclofenac therapy in Alzheimer's disease. Ther Adv Neurol Disord 2023; 16:17562864231156674. [PMID: 36875711 PMCID: PMC9974624 DOI: 10.1177/17562864231156674] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/26/2023] [Indexed: 03/07/2023] Open
Abstract
Alzheimer's disease (AD) is an untreatable cause of dementia, and new therapeutic approaches are urgently needed. AD pathology is defined by extracellular amyloid plaques and intracellular neurofibrillary tangles. Research of the past decades has suggested that neuroinflammation plays a critical role in the pathophysiology of AD. This has led to the idea that anti-inflammatory treatments might be beneficial. Early studies investigated non-steroidal anti-inflammatory drugs (NSAIDS) such as indomethacin, celecoxib, ibuprofen, and naproxen, which had no benefit. More recently, protective effects of diclofenac and NSAIDs in the fenamate group have been reported. Diclofenac decreased the frequency of AD significantly compared to other NSAIDs in a large retrospective cohort study. Diclofenac and fenamates share similar chemical structures, and evidence from cell and mouse models suggests that they inhibit the release of pro-inflammatory mediators from microglia with leads to the reduction of AD pathology. Here, we review the potential role of diclofenac and NSAIDs in the fenamate group for targeting AD pathology with a focus on its potential effects on microglia.
Collapse
Affiliation(s)
- Barbara E Stopschinski
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Danni McMahan
- Pharmacy Service, Dallas VA Medical Center, Dallas, TX, USA
| | - David A Jacob
- Veterans Integrated Service Network 17, Arlington, TX, USA
| | - Bertis B Little
- School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Hsueh-Sheng Chiang
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nil Saez Calveras
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Neurology Section, Dallas VA Medical Center, 4500 South Lancaster Road, Dallas, TX 75216, USA
| |
Collapse
|
12
|
Development and Challenges of Diclofenac-Based Novel Therapeutics: Targeting Cancer and Complex Diseases. Cancers (Basel) 2022; 14:cancers14184385. [PMID: 36139546 PMCID: PMC9496891 DOI: 10.3390/cancers14184385] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Diclofenac is a widely used drug for its anti-inflammatory and pain alleviating properties. This review summarizes the current understanding about the drug diclofenac. The potential applications of diclofenac beyond its well-known anti-inflammatory properties for other diseases such as cancer are discussed, along with existing limitations. Abstract Diclofenac is a highly prescribed non-steroidal anti-inflammatory drug (NSAID) that relieves inflammation, pain, fever, and aches, used at different doses depending on clinical conditions. This drug inhibits cyclooxygenase-1 and cyclooxygenase-2 enzymes, which are responsible for the generation of prostaglandin synthesis. To improve current diclofenac-based therapies, we require new molecular systematic therapeutic approaches to reduce complex multifactorial effects. However, the critical challenge that appears with diclofenac and other drugs of the same class is their side effects, such as signs of stomach injuries, kidney problems, cardiovascular issues, hepatic issues, and diarrhea. In this article, we discuss why defining diclofenac-based mechanisms, pharmacological features, and its medicinal properties are needed to direct future drug development against neurodegeneration and imperfect ageing and to improve cancer therapy. In addition, we describe various advance molecular mechanisms and fundamental aspects linked with diclofenac which can strengthen and enable the better designing of new derivatives of diclofenac to overcome critical challenges and improve their applications.
Collapse
|
13
|
Laska MJ, Møller JB, Graversen JH, Strøbæk D, Blomster L, Christophersen P, Bahrami S. Retroviral glycoprotein-mediated immune suppression via the potassium channel KCa3.1 - A new strategy for amelioration of inflammatory bowel diseases. Clin Immunol 2022; 242:109081. [PMID: 35905828 DOI: 10.1016/j.clim.2022.109081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/01/2022] [Accepted: 07/20/2022] [Indexed: 11/03/2022]
Abstract
Peptides derived from retroviral envelope proteins have been shown to possess a wide range of immunosuppressive and anti-inflammatory activities. We have previously reported identification of such a peptide derived from the envelope protein coded by a human endogenous retrovirus (HERV). In this study, we identified that in vitro the peptide inhibits the KCa3.1 potassium channel, a potential target for therapy of immune diseases. We describe in vitro ENV59-GP3 effects with respect to potency of inhibition on KCa3.1 channels and calcium influx. Furthermore, we asses in vivo the effect of blocking KCa3.1 with ENV59-GP3 peptide or KCa3.1-blocker NS6180 on protection against DSS-induced acute colitis. ENV59-GP3 peptide treatment showed reduction of the disease score in the DSS-induced acute colitis mice model, which was comparable to effects of the KCa3.1 channel blocker NS6180. Analysis of cytokine production from DSS-mice model treated animals revealed equipotent inhibitory effects of the ENV59-GP3 and NS6180 compounds on the production of IL-6, TNF-α, IL-1β. These findings altogether suggest that ENV59-GP3 functions as a KCa3.1 channel inhibitor and underline the implications of using virus derived channel blockers for treatment of autoimmune diseases. Additionally, they open the possibilities whether KCa3.1 inhibition is efficacious in patients with inflammatory bowel diseases.
Collapse
Affiliation(s)
- Magdalena J Laska
- Department of Molecular Biology and Genetics, Aarhus University, Denmark.
| | - Jesper Bonnet Møller
- Department of Cancer and Inflammation Research, University of Southern Denmark, Denmark
| | | | | | | | | | | |
Collapse
|
14
|
How the Potassium Channel Response of T Lymphocytes to the Tumor Microenvironment Shapes Antitumor Immunity. Cancers (Basel) 2022; 14:cancers14153564. [PMID: 35892822 PMCID: PMC9330401 DOI: 10.3390/cancers14153564] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/10/2022] Open
Abstract
Competent antitumor immune cells are fundamental for tumor surveillance and combating active cancers. Once established, tumors generate a tumor microenvironment (TME) consisting of complex cellular and metabolic elements that serve to suppress the function of antitumor immune cells. T lymphocytes are key cellular elements of the TME. In this review, we explore the role of ion channels, particularly K+ channels, in mediating the suppressive effects of the TME on T cells. First, we will review the complex network of ion channels that mediate Ca2+ influx and control effector functions in T cells. Then, we will discuss how multiple features of the TME influence the antitumor capabilities of T cells via ion channels. We will focus on hypoxia, adenosine, and ionic imbalances in the TME, as well as overexpression of programmed cell death ligand 1 by cancer cells that either suppress K+ channels in T cells and/or benefit from regulating these channels’ activity, ultimately shaping the immune response. Finally, we will review some of the cancer treatment implications related to ion channels. A better understanding of the effects of the TME on ion channels in T lymphocytes could promote the development of more effective immunotherapies, especially for resistant solid malignancies.
Collapse
|
15
|
Naseem MU, Carcamo-Noriega E, Beltrán-Vidal J, Borrego J, Szanto TG, Zamudio FZ, Delgado-Prudencio G, Possani LD, Panyi G. Cm28, a scorpion toxin having a unique primary structure, inhibits KV1.2 and KV1.3 with high affinity. J Gen Physiol 2022; 154:213282. [PMID: 35699659 PMCID: PMC9202693 DOI: 10.1085/jgp.202213146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/23/2022] [Indexed: 02/03/2023] Open
Abstract
The Cm28 in the venom of Centruroides margaritatus is a short peptide consisting of 27 amino acid residues with a mol wt of 2,820 D. Cm28 has <40% similarity with other known α-KTx from scorpions and lacks the typical functional dyad (lysine-tyrosine) required to block KV channels. However, its unique sequence contains the three disulfide-bond traits of the α-KTx scorpion toxin family. We propose that Cm28 is the first example of a new subfamily of α-KTxs, registered with the systematic number α-KTx32.1. Cm28 inhibited voltage-gated K+ channels KV1.2 and KV1.3 with Kd values of 0.96 and 1.3 nM, respectively. There was no significant shift in the conductance-voltage (G-V) relationship for any of the channels in the presence of toxin. Toxin binding kinetics showed that the association and dissociation rates are consistent with a bimolecular interaction between the peptide and the channel. Based on these, we conclude that Cm28 is not a gating modifier but rather a pore blocker. In a selectivity assay, Cm28 at 150 nM concentration (>100× Kd value for KV1.3) did not inhibit KV1.5, KV11.1, KCa1.1, and KCa3.1 K+ channels; NaV1.5 and NaV1.4 Na+ channels; or the hHV1 H+ channel but blocked ∼27% of the KV1.1 current. In a biological functional assay, Cm28 strongly inhibited the expression of the activation markers interleukin-2 receptor and CD40 ligand in anti-CD3-activated human CD4+ effector memory T lymphocytes. Cm28, due to its unique structure, may serve as a template for the generation of novel peptides targeting KV1.3 in autoimmune diseases.
Collapse
Affiliation(s)
- Muhammad Umair Naseem
- Department of Biophysics and Cell Biology, Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Edson Carcamo-Noriega
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnologia, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - José Beltrán-Vidal
- Grupo de Investigaciones Herpetológicas y Toxinológicas, Centro de Investigaciones Biomédicas, Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la Educación, Universidad del Cauca, Popayán, Colombia
| | - Jesus Borrego
- Department of Biophysics and Cell Biology, Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Tibor G. Szanto
- Department of Biophysics and Cell Biology, Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Fernando Z. Zamudio
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnologia, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Gustavo Delgado-Prudencio
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnologia, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Lourival D. Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnologia, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary,Correspondence to Gyorgy Panyi:
| |
Collapse
|
16
|
Imaging Kv1.3 Expressing Memory T Cells as a Marker of Immunotherapy Response. Cancers (Basel) 2022; 14:cancers14051217. [PMID: 35267526 PMCID: PMC8909107 DOI: 10.3390/cancers14051217] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 01/25/2023] Open
Abstract
Immune checkpoint inhibitors have shown great promise, emerging as a new pillar of treatment for cancer; however, only a relatively small proportion of recipients show a durable response to treatment. Strategies that reliably differentiate durably-responding tumours from non-responsive tumours are a critical unmet need. Persistent and durable immunological responses are associated with the generation of memory T cells. Effector memory T cells associated with tumour response to immune therapies are characterized by substantial upregulation of the potassium channel Kv1.3 after repeated antigen stimulation. We have developed a new Kv1.3 targeting radiopharmaceutical, [18F]AlF-NOTA-KCNA3P, and evaluated whether it can reliably differentiate tumours successfully responding to immune checkpoint inhibitor (ICI) therapy targeting PD-1 alone or combined with CLTA4. In a syngeneic colon cancer model, we compared tumour retention of [18F]AlF-NOTA-KCNA3P with changes in the tumour immune microenvironment determined by flow cytometry. Imaging with [18F]AlF-NOTA-KCNA3P reliably differentiated tumours responding to ICI therapy from non-responding tumours and was associated with substantial tumour infiltration of T cells, especially Kv1.3-expressing CD8+ effector memory T cells.
Collapse
|
17
|
Xiang ZN, Tong QL, Su JC, Hu ZF, Zhao N, Xia RF, Wu JL, Chen C, Chen JC, Wan LS. Diterpenoids with Rearranged 9(10→11)- abeo-10,12-Cyclojatrophane Skeleton and the First (15 S)-Jatrophane from Euphorbia helioscopia: Structural Elucidation, Biomimetic Conversion, and Their Immunosuppressive Effects. Org Lett 2022; 24:697-701. [PMID: 34965138 DOI: 10.1021/acs.orglett.1c04145] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Two novel diterpenoids, one with a rearranged trans,trans-fused tricyclo[10.3.0.04,6]pentadecane framework (1) and the other with an unprecedented 15S configuration (2), were isolated from Euphorbia helioscopia. Their structures were elucidated by extensive analysis of HR-ESI-MS, NMR, quantum-chemical calculation, and X-ray crystallographic data. Biosynthetically, 1 has a unique "cyclopropane-shift-like" biogenesis involving an oxa-di-π-methane (ODPM) rearrangement, which inspired us to accomplish the biomimetic conversion of 3 to 1. Moreover, compound 1 displayed a potent immunosuppressive effect by inhibiting Kv1.3 voltage-gated channels.
Collapse
Affiliation(s)
- Zhi-Nan Xiang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Pharmacy Department of Tongji Medical School, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Qi-Lin Tong
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Pharmacy Department of Tongji Medical School, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Jun-Cheng Su
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, People's Republic of China
| | - Zhuo-Fan Hu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Pharmacy Department of Tongji Medical School, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Ning Zhao
- Research Center of Ion Channelopathy, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Ru-Feng Xia
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Pharmacy Department of Tongji Medical School, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Jia-Le Wu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Pharmacy Department of Tongji Medical School, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Chen Chen
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Pharmacy Department of Tongji Medical School, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Jia-Chun Chen
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Pharmacy Department of Tongji Medical School, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Luo-Sheng Wan
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Pharmacy Department of Tongji Medical School, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| |
Collapse
|
18
|
Hwang S, Kim JH, Jo SH. Inhibitory effect of the selective serotonin reuptake inhibitor paroxetine on human Kv1.3 channels. Eur J Pharmacol 2021; 912:174567. [PMID: 34662565 DOI: 10.1016/j.ejphar.2021.174567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/30/2021] [Accepted: 10/11/2021] [Indexed: 01/12/2023]
Abstract
Paroxetine is one of the most effective selective serotonin reuptake inhibitors used to treat depressive and panic disorders that reduce the viability of human T lymphocytes, in which Kv1.3 channels are highly expressed. We examined whether paroxetine could modulate human Kv1.3 channels acutely and directly with the aim of understanding the biophysical effects and the underlying mechanisms of the drug. Kv1.3 channel proteins were expressed in Xenopus oocytes. Paroxetine rapidly inhibited the steady-state current and peak current of these channels within 6 min in a concentration-dependent manner; IC50s were 26.3 μM and 53.9 μM, respectively, and these effects were partially reversed by washout, which excluded the possibility of genomic regulation. At the same test voltage, paroxetine blockade of the steady-state currents was higher than that of the peak currents, and the inhibition of the steady-state current increased relative to the degree of depolarization. Paroxetine decreased the inactivation time constant in a concentration-dependent manner, but it did not affect the activation time constant, which resulted in the acceleration of intrinsic inactivation without changing ultrarapid activation. Blockade of Kv1.3 channels by paroxetine exhibited more rapid inhibition at higher activation frequencies showing the use-dependency of the blockade. Overall, these results show that paroxetine directly suppresses human Kv1.3 channels in an open state and accelerates the process of steady-state inactivation; thus, we have revealed a biophysical mechanism for possible acute immunosuppressive effects of paroxetine.
Collapse
Affiliation(s)
- Soobeen Hwang
- Department of Physiology, Institute of Bioscience and Biotechnology, Interdisciplinary Graduate Program in BIT Medical Convergence, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Jong-Hui Kim
- Department of Physiology, Institute of Bioscience and Biotechnology, Interdisciplinary Graduate Program in BIT Medical Convergence, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Su-Hyun Jo
- Department of Physiology, Institute of Bioscience and Biotechnology, Interdisciplinary Graduate Program in BIT Medical Convergence, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea.
| |
Collapse
|
19
|
Tajti G, Szanto TG, Csoti A, Racz G, Evaristo C, Hajdu P, Panyi G. Immunomagnetic separation is a suitable method for electrophysiology and ion channel pharmacology studies on T cells. Channels (Austin) 2021; 15:53-66. [PMID: 33356811 PMCID: PMC7781520 DOI: 10.1080/19336950.2020.1859753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/01/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Ion channels play pivotal role in the physiological and pathological function of immune cells. As immune cells represent a functionally diverse population, subtype-specific functional studies, such as single-cell electrophysiology require proper subset identification and separation. Magnetic-activated cell sorting (MACS) techniques provide an alternative to fluorescence-activated cell sorting (FACS), however, the potential impact of MACS-related beads on the biophysical and pharmacological properties of the ion channels were not studied yet. We studied the aforementioned properties of the voltage-gated Kv1.3 K+ channel in activated CD4+ T-cells as well as the membrane capacitance using whole-cell patch-clamp following immunomagnetic positive separation, using the REAlease® kit. This kit allows three experimental configurations: bead-bound configuration, bead-free configuration following the removal of magnetic beads, and the label-free configuration following removal of CD4 recognizing antibody fragments. As controls, we used FACS separation as well as immunomagnetic negative selection. The membrane capacitance and of the biophysical parameters of Kv1.3 gating, voltage-dependence of steady-state activation and inactivation kinetics of the current were not affected by the presence of MACS-related compounds on the cell surface. We found subtle differences in the activation kinetics of the Kv1.3 current that could not be explained by the presence of MACS-related compounds. Neither the equilibrium block of Kv1.3 by TEA+ or charybdotoxin (ChTx) nor the kinetics of ChTx block are affected by the presence of the magnetics beads on the cell surface. Based on our results MACS is a suitable method to separate cells for studying ion channels in non-excitable cells, such as T-lymphocytes.
Collapse
Affiliation(s)
- Gabor Tajti
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tibor Gabor Szanto
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Agota Csoti
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Greta Racz
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - César Evaristo
- R&D Reagents Chemical Biology, Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Peter Hajdu
- Department of Biophysics and Cell Biology, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
20
|
Nicolazzo JA, Pan Y, Di Stefano I, Choy KHC, Reddiar SB, Low YL, Wai DCC, Norton RS, Jin L. Blockade of Microglial Kv1.3 Potassium Channels by the Peptide HsTX1[R14A] Attenuates Lipopolysaccharide-mediated Neuroinflammation. J Pharm Sci 2021; 111:638-647. [PMID: 34767826 DOI: 10.1016/j.xphs.2021.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/03/2021] [Accepted: 11/03/2021] [Indexed: 12/19/2022]
Abstract
The expression of voltage-gated potassium Kv1.3 channels is increased in activated microglia, with non-selective blockade reported to attenuate microglial-mediated neuroinflammation. In this study, we evaluated the impact of a potent and selective peptidic blocker of Kv1.3 channels, HsTX1[R14A], on microglial-mediated neuroinflammation in vitro and in vivo. Treatment with both 0.1 and 1 µg/mL lipopolysaccharide (LPS) significantly (p < 0.05) increased Kv1.3 abundance on the surface of BV-2 microglia in association with increased levels of mRNA for tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6). The increased transcription of TNF-α and IL-6 was significantly attenuated (by 24.9 and 20.2%, respectively) by HsTX1[R14A] (100 nM). The concomitant increase in TNF-α and IL-6 release from BV-2 microglia was significantly attenuated by HsTX1[R14A] by 10.7 and 12.6%, respectively. In LPS-treated primary mouse microglia, the levels of TNF-α and nitric oxide were also attenuated by HsTX1[R14A] (26.1 and 20.4%, respectively). In an LPS-induced mouse model of neuroinflammation, both an immediate and delayed subcutaneous dose of HsTX1[R14A] (2 mg/kg) significantly reduced plasma and brain levels of the pro-inflammatory mediators TNF-α, IL-1β and IL-6, with no impact on the anti-inflammatory IL-10. These results demonstrate that HsTX1[R14A] is a promising therapeutic candidate for the treatment of diseases with a neuroinflammatory component.
Collapse
Affiliation(s)
- Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Yijun Pan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Ilenia Di Stefano
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Kwok H C Choy
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Sanjeevini Babu Reddiar
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Yi Ling Low
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Dorothy C C Wai
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia; ARC Centre for Fragment-Based Design, Monash University, Parkville, Victoria 3052, Australia
| | - Liang Jin
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
21
|
Naseem MU, Tajti G, Gaspar A, Szanto TG, Borrego J, Panyi G. Optimization of Pichia pastoris Expression System for High-Level Production of Margatoxin. Front Pharmacol 2021; 12:733610. [PMID: 34658872 PMCID: PMC8511391 DOI: 10.3389/fphar.2021.733610] [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] [Accepted: 08/30/2021] [Indexed: 11/16/2022] Open
Abstract
Margatoxin (MgTx) is a high-affinity blocker of voltage-gated potassium (Kv) channels. It inhibits Kv1.1–Kv1.3 ion channels in picomolar concentrations. This toxin is widely used to study physiological function of Kv ion channels in various cell types, including immune cells. Isolation of native MgTx in large quantities from scorpion venom is not affordable. Chemical synthesis and recombinant production in Escherichia coli need in vitro oxidative refolding for proper disulfide bond formation, resulting in a very low yield of peptide production. The Pichia pastoris expression system offers an economical approach to overcome all these limitations and gives a higher yield of correctly refolded recombinant peptides. In this study, improved heterologous expression of recombinant MgTx (rMgTx) in P. pastoris was obtained by using preferential codons, selecting the hyper-resistant clone against Zeocin, and optimizing the culturing conditions. About 36 ± 4 mg/L of >98% pure His-tagged rMgTx (TrMgTx) was produced, which is a threefold higher yield than has been previously reported. Proteolytic digestion of TrMgTx with factor Xa generated untagged rMgTx (UrMgTx). Both TrMgTx and UrMgTx blocked the Kv1.2 and Kv1.3 currents (patch-clamp) (Kd for Kv1.2 were 64 and 14 pM, and for Kv1.3, 86 and 50 pM, respectively) with comparable potency to the native MgTx. The analysis of the binding kinetics showed that TrMgTx had a lower association rate than UrMgTx for both Kv1.2 and Kv1.3. The dissociation rate of both the analogues was the same for Kv1.3. However, in the case of Kv1.2, TrMgTx showed a much higher dissociation rate with full recovery of the block than UrMgTx. Moreover, in a biological functional assay, both peptides significantly downregulated the expression of early activation markers IL2R and CD40L in activated CD4+ TEM lymphocytes whose activation was Kv1.3 dependent. In conclusion, the authors report that the Pichia expression system is a powerful method to produce disulfide-rich peptides, the overexpression of which could be enhanced noticeably through optimization strategies, making it more cost-effective. Since the presence of the His-tag on rMgTx only mildly altered the block equilibrium and binding kinetics, recombinant toxins could be used in ion channel research without removing the tag and could thus reduce the cost and time demand for toxin production.
Collapse
Affiliation(s)
- Muhammad Umair Naseem
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gabor Tajti
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Gaspar
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Technology, Institute of Chemistry, University of Debrecen, Debrecen, Hungary
| | - Tibor G Szanto
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Jesús Borrego
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
22
|
Bohmwald K, Gálvez NMS, Andrade CA, Mora VP, Muñoz JT, González PA, Riedel CA, Kalergis AM. Modulation of Adaptive Immunity and Viral Infections by Ion Channels. Front Physiol 2021; 12:736681. [PMID: 34690811 PMCID: PMC8531258 DOI: 10.3389/fphys.2021.736681] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/10/2021] [Indexed: 12/15/2022] Open
Abstract
Most cellular functions require of ion homeostasis and ion movement. Among others, ion channels play a crucial role in controlling the homeostasis of anions and cations concentration between the extracellular and intracellular compartments. Calcium (Ca2+) is one of the most relevant ions involved in regulating critical functions of immune cells, allowing the appropriate development of immune cell responses against pathogens and tumor cells. Due to the importance of Ca2+ in inducing the immune response, some viruses have evolved mechanisms to modulate intracellular Ca2+ concentrations and the mobilization of this cation through Ca2+ channels to increase their infectivity and to evade the immune system using different mechanisms. For instance, some viral infections require the influx of Ca2+ through ionic channels as a first step to enter the cell, as well as their replication and budding. Moreover, through the expression of viral proteins on the surface of infected cells, Ca2+ channels function can be altered, enhancing the pathogen evasion of the adaptive immune response. In this article, we review those ion channels and ion transporters that are essential for the function of immune cells. Specifically, cation channels and Ca2+ channels in the context of viral infections and their contribution to the modulation of adaptive immune responses.
Collapse
Affiliation(s)
- Karen Bohmwald
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M. S. Gálvez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A. Andrade
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina P. Mora
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José T. Muñoz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Millennium Institute on Immunology and Immunotherapy, Universidad Andres Bello, Santiago, Chile
| | - Alexis M. Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
23
|
Phan HTL, Kim HJ, Jo S, Kim WK, Namkung W, Nam JH. Anti-Inflammatory Effect of Licochalcone A via Regulation of ORAI1 and K + Channels in T-Lymphocytes. Int J Mol Sci 2021; 22:ijms221910847. [PMID: 34639190 PMCID: PMC8509259 DOI: 10.3390/ijms221910847] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023] Open
Abstract
Calcium signaling plays a vital role in the regulation of various cellular processes, including activation, proliferation, and differentiation of T-lymphocytes, which is mediated by ORAI1 and potassium (K+) channels. These channels have also been identified as highly attractive therapeutic targets for immune-related diseases. Licochalcone A is a licorice-derived chalconoid known for its multifaceted beneficial effects in pharmacological treatments, including its anti-inflammatory, anti-asthmatic, antioxidant, antimicrobial, and antitumorigenic properties. However, its anti-inflammatory effects involving ion channels in lymphocytes remain unclear. Thus, the present study aimed to investigate whether licochalcone A inhibits ORAI1 and K+ channels in T-lymphocytes. Our results indicated that licochalcone A suppressed all three channels (ORAI1, Kv1.3, and KCa3.1) in a concentration-dependent matter, with IC50 values of 2.97 ± 1.217 µM, 0.83 ± 1.222 µM, and 11.21 ± 1.07 µM, respectively. Of note, licochalcone A exerted its suppressive effects on the IL-2 secretion and proliferation in CD3 and CD28 antibody-induced T-cells. These results indicate that the use of licochalcone A may provide an effective treatment strategy for inflammation-related immune diseases.
Collapse
Affiliation(s)
- Hong T. L. Phan
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Goyang 10326, Korea; (H.J.K.); (W.K.K.)
| | - Hyun J. Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Goyang 10326, Korea; (H.J.K.); (W.K.K.)
| | - Sungwoo Jo
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Incheon 21983, Korea;
| | - Woo K. Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Goyang 10326, Korea; (H.J.K.); (W.K.K.)
- Department of Internal Medicine Graduate School of Medicine, Dongguk University, 27 Dongguk-ro, Goyang 10326, Korea
| | - Wan Namkung
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Incheon 21983, Korea;
- Correspondence: (W.N.); (J.H.N.)
| | - Joo H. Nam
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Goyang 10326, Korea; (H.J.K.); (W.K.K.)
- Correspondence: (W.N.); (J.H.N.)
| |
Collapse
|
24
|
Comparison of the Effect of Oral Versus Intravenous Bisphosphonate Administration on Osteoclastogenesis in Advanced-Stage Medication-Related Osteonecrosis of the Jaw Patients. J Clin Med 2021; 10:jcm10132988. [PMID: 34279472 PMCID: PMC8268194 DOI: 10.3390/jcm10132988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022] Open
Abstract
It is yet unknown whether the intravenous administration route alone can fully account for the exacerbation of medication-related osteonecrosis of the jaw (MRONJ). The purpose of this retrospective study was to identify the potential role of the bisphosphonate (BP) administration route as an independent prognostic factor for non-cancerous, stage III MRONJ patients. Bone samples were retrospectively obtained from two groups of osteoporosis patients who underwent surgery for the treatment of stage III MRONJ. Among the subjects, 10 had a history of only oral BP consumption and 10 of intravenous (IV) BP administration. The samples were assessed for osteoclast morphology and immunohistochemical expression of the receptor activator of NF-κB ligand (RANKL), osteoprotegerin (OPG), and potassium calcium-activated channel subfamily N member 4 (Kcnn4). Although the osteoclasts derived from both groups exhibited no significant differences in the mean quantity, diameter, and nuclearity, significantly attenuated tartrate-resistant acid phosphatase activity was noted among the IV BP-induced MRONJ bones compared to those of the oral BP group. Significant suppression of the RANKL/OPG ratio and Kcnn4 expression among the retrieved bones of IV BP group patients was also noted. Our results indicate the potential of the BP administration route as an independent prognostic factor for advanced-stage MRONJ, regardless of the dosage or indication for which the BP was prescribed.
Collapse
|
25
|
Kim HJ, Park S, Shin HY, Nam YR, Lam Hong PT, Chin YW, Nam JH, Kim WK. Inhibitory effects of α-Mangostin on T cell cytokine secretion via ORAI1 calcium channel and K + channels inhibition. PeerJ 2021; 9:e10973. [PMID: 33717700 PMCID: PMC7936567 DOI: 10.7717/peerj.10973] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/29/2021] [Indexed: 12/16/2022] Open
Abstract
Background As one of the main components of mangosteen (Garcinia mangostana), a tropical fruit, α-mangostin has been reported to have numerous pharmacological benefits such as anti-cancer, anti-inflammatory, and anti-allergic effects through various mechanisms of action. The effects of α-mangostin on intracellular signaling proteins is well studied, but the effects of α-mangostin on ion channels and its physiological effects in immune cells are unknown. Generation of intracellular calcium signaling is a fundamental step for T cell receptor stimulation. This signaling is mediated not only by the ORAI1 calcium channel, but also by potassium ion channels, which provide the electrical driving forces for generating sufficient calcium ion influx. This study investigated whether α-mangosteen suppress T cell stimulation by inhibiting ORAI1 and two kinds of potassium channels (Kv1.3 and KCa3.1), which are normally expressed in human T cells. Methods This study analyzed the inhibitory effect of α-mangostin on immune cell activity via inhibition of calcium and potassium ion channels expressed in immune cells. Results α-mangostin inhibited ORAI1 in a concentration-dependent manner, and the IC50 value was 1.27 ± 1.144 µM. Kv1.3 was suppressed by 41.38 ± 6.191% at 3 µM, and KCa3.1 was suppressed by 51.16 ± 5.385% at 3 µM. To measure the inhibition of cytokine secretion by immune cells, Jurkat T cells were stimulated to induce IL-2 secretion, and α-mangostin was found to inhibit it. This study demonstrated the anti-inflammatory effect of α-mangostin, the main component of mangosteen, through the regulation of calcium signals.
Collapse
Affiliation(s)
- Hyun Jong Kim
- Department of Physiology, Dongguk University College of Medicine, Gyeong-ju, Gyeongsangbuk-do, Republic of Korea.,Channelopathy Research Center (CRC), College of Medicine, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea
| | - Seorin Park
- Department of Physiology, Dongguk University College of Medicine, Gyeong-ju, Gyeongsangbuk-do, Republic of Korea
| | - Hui Young Shin
- Department of Physiology, Dongguk University College of Medicine, Gyeong-ju, Gyeongsangbuk-do, Republic of Korea
| | - Yu Ran Nam
- Channelopathy Research Center (CRC), College of Medicine, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea
| | - Phan Thi Lam Hong
- Department of Physiology, Dongguk University College of Medicine, Gyeong-ju, Gyeongsangbuk-do, Republic of Korea.,Channelopathy Research Center (CRC), College of Medicine, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea
| | - Young-Won Chin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeong-ju, Gyeongsangbuk-do, Republic of Korea.,Channelopathy Research Center (CRC), College of Medicine, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), College of Medicine, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea.,Department of Internal Medicine Graduate School of Medicine, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea
| |
Collapse
|
26
|
Novel Therapeutic Approaches of Ion Channels and Transporters in Cancer. Rev Physiol Biochem Pharmacol 2020; 183:45-101. [PMID: 32715321 DOI: 10.1007/112_2020_28] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The expression and function of many ion channels and transporters in cancer cells display major differences in comparison to those from healthy cells. These differences provide the cancer cells with advantages for tumor development. Accordingly, targeting ion channels and transporters have beneficial anticancer effects including inhibition of cancer cell proliferation, migration, invasion, metastasis, tumor vascularization, and chemotherapy resistance, as well as promoting apoptosis. Some of the molecular mechanisms associating ion channels and transporters with cancer include the participation of oxidative stress, immune response, metabolic pathways, drug synergism, as well as noncanonical functions of ion channels. This diversity of mechanisms offers an exciting possibility to suggest novel and more effective therapeutic approaches to fight cancer. Here, we review and discuss most of the current knowledge suggesting novel therapeutic approaches for cancer therapy targeting ion channels and transporters. The role and regulation of ion channels and transporters in cancer provide a plethora of exceptional opportunities in drug design, as well as novel and promising therapeutic approaches that may be used for the benefit of cancer patients.
Collapse
|
27
|
Tajti G, Wai DCC, Panyi G, Norton RS. The voltage-gated potassium channel K V1.3 as a therapeutic target for venom-derived peptides. Biochem Pharmacol 2020; 181:114146. [PMID: 32653588 DOI: 10.1016/j.bcp.2020.114146] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/05/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023]
Abstract
The voltage-gated potassium channel KV1.3 is a well-established therapeutic target for a range of autoimmune diseases, in addition to being the site of action of many venom-derived peptides. Numerous studies have documented the efficacy of venom peptides that target KV1.3, in particular from sea anemones and scorpions, in animal models of autoimmune diseases such as rheumatoid arthritis, psoriasis and multiple sclerosis. Moreover, an analogue of the sea anemone peptide ShK (known as dalazatide) has successfully completed Phase 1 clinical trials in mild-to-moderate plaque psoriasis. In this article we consider other potential therapeutic applications of inhibitors of KV1.3, including in inflammatory bowel disease and neuroinflammatory conditions such as Alzheimer's and Parkinson's diseases, as well as fibrotic diseases. We also summarise strategies for facilitating the entry of peptides to the central nervous system, given that this will be a pre-requisite for the treatment of most neuroinflammatory diseases. Venom-derived peptides that have been reported recently to target KV1.3 are also described. The increasing number of autoimmune and other conditions in which KV1.3 is upregulated and is therefore a potential therapeutic target, combined with the fact that many venom-derived peptides are potent inhibitors of KV1.3, suggests that venoms are likely to continue to serve as a rich source of new pharmacological tools and therapeutic leads targeting this channel.
Collapse
Affiliation(s)
- Gabor Tajti
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Dorothy C C Wai
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia; ARC Centre for Fragment-Based Design, Monash University, Parkville, VIC 3052, Australia.
| |
Collapse
|
28
|
Ahmadi S, Knerr JM, Argemi L, Bordon KCF, Pucca MB, Cerni FA, Arantes EC, Çalışkan F, Laustsen AH. Scorpion Venom: Detriments and Benefits. Biomedicines 2020; 8:biomedicines8050118. [PMID: 32408604 PMCID: PMC7277529 DOI: 10.3390/biomedicines8050118] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/01/2020] [Accepted: 05/07/2020] [Indexed: 12/17/2022] Open
Abstract
Scorpion venom may cause severe medical complications and untimely death if injected into the human body. Neurotoxins are the main components of scorpion venom that are known to be responsible for the pathological manifestations of envenoming. Besides neurotoxins, a wide range of other bioactive molecules can be found in scorpion venoms. Advances in separation, characterization, and biotechnological approaches have enabled not only the development of more effective treatments against scorpion envenomings, but have also led to the discovery of several scorpion venom peptides with interesting therapeutic properties. Thus, scorpion venom may not only be a medical threat to human health, but could prove to be a valuable source of bioactive molecules that may serve as leads for the development of new therapies against current and emerging diseases. This review presents both the detrimental and beneficial properties of scorpion venom toxins and discusses the newest advances within the development of novel therapies against scorpion envenoming and the therapeutic perspectives for scorpion toxins in drug discovery.
Collapse
Affiliation(s)
- Shirin Ahmadi
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark; (J.M.K.); (L.A.); (M.B.P.); (F.A.C.)
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Sciences, Eşkisehir Osmangazi University, TR-26040 Eşkisehir, Turkey;
- Correspondence: (S.A.); (A.H.L.); Tel.: +45-7164-6042 (S.A.); +45-2988-1134 (A.H.L.)
| | - Julius M. Knerr
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark; (J.M.K.); (L.A.); (M.B.P.); (F.A.C.)
| | - Lídia Argemi
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark; (J.M.K.); (L.A.); (M.B.P.); (F.A.C.)
| | - Karla C. F. Bordon
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto—São Paulo 14040-903, Brazil; (K.C.F.B.); (E.C.A.)
| | - Manuela B. Pucca
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark; (J.M.K.); (L.A.); (M.B.P.); (F.A.C.)
- Medical School, Federal University of Roraima, Boa Vista, Roraima 69310-000, Brazil
| | - Felipe A. Cerni
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark; (J.M.K.); (L.A.); (M.B.P.); (F.A.C.)
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto—São Paulo 14040-903, Brazil; (K.C.F.B.); (E.C.A.)
| | - Eliane C. Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto—São Paulo 14040-903, Brazil; (K.C.F.B.); (E.C.A.)
| | - Figen Çalışkan
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Sciences, Eşkisehir Osmangazi University, TR-26040 Eşkisehir, Turkey;
- Department of Biology, Faculty of Science and Letters, Eskisehir Osmangazi University, TR-26040 Eskisehir, Turkey
| | - Andreas H. Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark; (J.M.K.); (L.A.); (M.B.P.); (F.A.C.)
- Correspondence: (S.A.); (A.H.L.); Tel.: +45-7164-6042 (S.A.); +45-2988-1134 (A.H.L.)
| |
Collapse
|
29
|
Zheng Q, Na R, Yang L, Yu H, Zhao X, Huang X. The binding process of BmKTX and BmKTX-D33H toward to Kv1.3 channel: a molecular dynamics simulation study. J Biomol Struct Dyn 2020; 39:2788-2797. [PMID: 32329410 DOI: 10.1080/07391102.2020.1760135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The potassium channel Kv1.3 is an important pharmacological target and the Kaliotoxin-type toxins (α-KTX-3 family) are its specific blockers. Here, we study the binding process of two kinds of Kaliotoxin-type toxins:BmKTX and its mutant (BmKTX-D33H) toward to Kv1.3 channel using MD simulation and umbrella sampling simulation, respectively. The calculated binding free energies are -27 kcal/mol and -34 kcal/mol for BmKTX and BmKTX-D33H, respectively, which are consistent with experimental results. The further analysis indicate that the characteristic of electrostatic potential of the α-KTX-3 have important effect on their binding modes with Kv1.3 channel; the residue 33 in BmKTX or BmKTX-D33H plays a key role in determine their binding orientations toward to Kv1.3 channel; when residue 33 (or 34) has negative electrostatic potential, the anti-parallel β-sheet domain of α-KTX-3 toxin peptide will keep away from the filter region of Kv1.3 channel, as BmKTX; when residue 33(or 34) has positive electrostatic potential, the anti-parallel β-sheet domain of α-KTX-3 toxin peptide will interact with the filter region of Kv1.3 channel, as BmKTX-D33H. Above all, electrostatic potential differences on toxin surfaces and correlations motions within the toxins will determine the toxin-potassium channel interaction model. In addition, the hydrogen bond interaction is the pivotal factor for the Kv1.3-Kaliotoxin association. Understanding the binding mechanism of toxin-potassium channel will facilitate the rational development of new toxin analogue.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Qiancheng Zheng
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University, Changchun, China
| | - Risong Na
- College of Plant Protection, Henan Agricultural University, Zhengzhou, P.R China
| | - Lianjuan Yang
- Department of Mycology, Shanghai Dermatology Hospital, Shanghai, China
| | - Hui Yu
- College of Science, Beihua Univesrity, Jilin, China
| | - Xi Zhao
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University, Changchun, China
| | - Xuri Huang
- Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University, Changchun, China
| |
Collapse
|
30
|
Yang PC, Jafri MS. Ca 2+ signaling in T lymphocytes: the interplay of the endoplasmic reticulum, mitochondria, membrane potential, and CRAC channels on transcription factor activation. Heliyon 2020; 6:e03526. [PMID: 32181396 PMCID: PMC7063158 DOI: 10.1016/j.heliyon.2020.e03526] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/12/2018] [Accepted: 02/28/2020] [Indexed: 11/16/2022] Open
Abstract
T cell receptor stimulation initiates a cascade of reactions that cause an increase in intracellular calcium (Ca2+) concentration mediated through inositol 1,4,5-trisphosphate (IP3). To understand the basic mechanisms by which the immune response in T cells is activated, it is useful to understand the signaling pathways that contain important targets for drugs in a quantitative fashion. A computational model helps us to understand how the selected elements in the pathways interact with each other, and which component plays the crucial role in systems. We have developed a mathematical model to explore the mechanism for controlling transcription factor activity, which regulates gene expression, by the modulation of calcium signaling triggered during T cell activation. The model simulates the activation and modulation of Ca2+ release-activated Ca2+ (CRAC) channels by mitochondrial dynamics and depletion of endoplasmic reticulum (ER) store, and also includes membrane potential in T-cells. The model simulates the experimental finding that increases in Ca2+ current enhances the activation of transcription factors and the Ca2+ influx through CRAC is also essential for the NFAT and NFκB activation. The model also suggests that plasma membrane Ca2+-ATPase (PMCA) controls a majority of the extrusion of Ca2+ and modulates the activation of CRAC channels. Furthermore, the model simulations explain how the complex interaction of the endoplasmic reticulum, membrane potential, mitochondria, and ion channels such as CRAC channels control T cell activation.
Collapse
Affiliation(s)
- Pei-Chi Yang
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, 95616, USA
- Krasnow Institute for Advanced Study and School of Systems Biology, George Mason University, Fairfax, VA, 22030, USA
| | - M. Saleet Jafri
- Krasnow Institute for Advanced Study and School of Systems Biology, George Mason University, Fairfax, VA, 22030, USA
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 20201, USA
| |
Collapse
|
31
|
Zhu YR, Jiang XX, Zhang DM. Critical regulation of atherosclerosis by the KCa3.1 channel and the retargeting of this therapeutic target in in-stent neoatherosclerosis. J Mol Med (Berl) 2019; 97:1219-1229. [DOI: 10.1007/s00109-019-01814-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 05/07/2019] [Accepted: 06/18/2019] [Indexed: 01/09/2023]
|
32
|
Anti-tumoral effect of scorpion peptides: Emerging new cellular targets and signaling pathways. Cell Calcium 2019; 80:160-174. [DOI: 10.1016/j.ceca.2019.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/10/2019] [Accepted: 05/10/2019] [Indexed: 12/31/2022]
|
33
|
Oliveira IS, Ferreira IG, Alexandre-Silva GM, Cerni FA, Cremonez CM, Arantes EC, Zottich U, Pucca MB. Scorpion toxins targeting Kv1.3 channels: insights into immunosuppression. J Venom Anim Toxins Incl Trop Dis 2019; 25:e148118. [PMID: 31131004 PMCID: PMC6483409 DOI: 10.1590/1678-9199-jvatitd-1481-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/17/2018] [Indexed: 01/26/2023] Open
Abstract
Scorpion venoms are natural sources of molecules that have, in addition to their
toxic function, potential therapeutic applications. In this source the
neurotoxins can be found especially those that act on potassium channels.
Potassium channels are responsible for maintaining the membrane potential in the
excitable cells, especially the voltage-dependent potassium channels (Kv),
including Kv1.3 channels. These channels (Kv1.3) are expressed by various types
of tissues and cells, being part of several physiological processes. However,
the major studies of Kv1.3 are performed on T cells due its importance on
autoimmune diseases. Scorpion toxins capable of acting on potassium channels
(KTx), mainly on Kv1.3 channels, have gained a prominent role for their possible
ability to control inflammatory autoimmune diseases. Some of these toxins have
already left bench trials and are being evaluated in clinical trials, presenting
great therapeutic potential. Thus, scorpion toxins are important natural
molecules that should not be overlooked in the treatment of autoimmune and other
diseases.
Collapse
Affiliation(s)
- Isadora S Oliveira
- School of Pharmaceutical Sciences of Ribeirão Preto, Department of Physics and Chemistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Isabela G Ferreira
- School of Pharmaceutical Sciences of Ribeirão Preto, Department of Physics and Chemistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Felipe A Cerni
- Ribeirão Preto Medical School, Department of Biochemistry and Immunology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Caroline M Cremonez
- School of Pharmaceutical Sciences of Ribeirão Preto, Department of Physics and Chemistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Eliane C Arantes
- School of Pharmaceutical Sciences of Ribeirão Preto, Department of Physics and Chemistry, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Umberto Zottich
- Medical School, Federal University of Roraima, Boa Vista, RR, Brazil
| | - Manuela B Pucca
- Medical School, Federal University of Roraima, Boa Vista, RR, Brazil
| |
Collapse
|
34
|
Du C, Li J, Shao Z, Mwangi J, Xu R, Tian H, Mo G, Lai R, Yang S. Centipede KCNQ Inhibitor SsTx Also Targets K V1.3. Toxins (Basel) 2019; 11:toxins11020076. [PMID: 30717088 PMCID: PMC6409716 DOI: 10.3390/toxins11020076] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/25/2019] [Accepted: 01/27/2019] [Indexed: 12/19/2022] Open
Abstract
It was recently discovered that Ssm Spooky Toxin (SsTx) with 53 residues serves as a key killer factor in red-headed centipede’s venom arsenal, due to its potent blockage of the widely expressed KCNQ channels to simultaneously and efficiently disrupt cardiovascular, respiratory, muscular, and nervous systems, suggesting that SsTx is a basic compound for centipedes’ defense and predation. Here, we show that SsTx also inhibits KV1.3 channel, which would amplify the broad-spectrum disruptive effect of blocking KV7 channels. Interestingly, residue R12 in SsTx extends into the selectivity filter to block KV7.4, however, residue K11 in SsTx replaces this ploy when toxin binds on KV1.3. Both SsTx and its mutant SsTx_R12A inhibit cytokines production in T cells without affecting the level of KV1.3 expression. The results further suggest that SsTx is a key molecule for defense and predation in the centipedes’ venoms and it evolves efficient strategy to disturb multiple physiological targets.
Collapse
Affiliation(s)
- Canwei Du
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | - Jiameng Li
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | - Zicheng Shao
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | - James Mwangi
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Yunnan Province, Kunming Institute of Zoology, Kunming 650223, Yunnan, China.
- University of Chinese Academy of Sciences, Beijing 100009, China.
| | - Runjia Xu
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | - Huiwen Tian
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | - Guoxiang Mo
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | - Ren Lai
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Yunnan Province, Kunming Institute of Zoology, Kunming 650223, Yunnan, China.
- Sino-African Joint Research Center, Chinese Academy of Science, Wuhan 430074, Hubei, China.
| | - Shilong Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Yunnan Province, Kunming Institute of Zoology, Kunming 650223, Yunnan, China.
- Sino-African Joint Research Center, Chinese Academy of Science, Wuhan 430074, Hubei, China.
| |
Collapse
|
35
|
Cancer-Associated Intermediate Conductance Ca 2+-Activated K⁺ Channel K Ca3.1. Cancers (Basel) 2019; 11:cancers11010109. [PMID: 30658505 PMCID: PMC6357066 DOI: 10.3390/cancers11010109] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 12/14/2022] Open
Abstract
Several tumor entities have been reported to overexpress KCa3.1 potassium channels due to epigenetic, transcriptional, or post-translational modifications. By modulating membrane potential, cell volume, or Ca2+ signaling, KCa3.1 has been proposed to exert pivotal oncogenic functions in tumorigenesis, malignant progression, metastasis, and therapy resistance. Moreover, KCa3.1 is expressed by tumor-promoting stroma cells such as fibroblasts and the tumor vasculature suggesting a role of KCa3.1 in the adaptation of the tumor microenvironment. Combined, this features KCa3.1 as a candidate target for innovative anti-cancer therapy. However, immune cells also express KCa3.1 thereby contributing to T cell activation. Thus, any strategy targeting KCa3.1 in anti-cancer therapy may also modulate anti-tumor immune activity and/or immunosuppression. The present review article highlights the potential of KCa3.1 as an anti-tumor target providing an overview of the current knowledge on its function in tumor pathogenesis with emphasis on vasculo- and angiogenesis as well as anti-cancer immune responses.
Collapse
|
36
|
Hutchings CJ, Colussi P, Clark TG. Ion channels as therapeutic antibody targets. MAbs 2018; 11:265-296. [PMID: 30526315 PMCID: PMC6380435 DOI: 10.1080/19420862.2018.1548232] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/01/2018] [Accepted: 11/03/2018] [Indexed: 12/12/2022] Open
Abstract
It is now well established that antibodies have numerous potential benefits when developed as therapeutics. Here, we evaluate the technical challenges of raising antibodies to membrane-spanning proteins together with enabling technologies that may facilitate the discovery of antibody therapeutics to ion channels. Additionally, we discuss the potential targeting opportunities in the anti-ion channel antibody landscape, along with a number of case studies where functional antibodies that target ion channels have been reported. Antibodies currently in development and progressing towards the clinic are highlighted.
Collapse
Affiliation(s)
| | | | - Theodore G. Clark
- TetraGenetics Inc, Arlington Massachusetts, USA
- Department of Microbiology and Immunology, Cornell University, Ithaca New York, USA
| |
Collapse
|
37
|
Matsui M, Terasawa K, Kajikuri J, Kito H, Endo K, Jaikhan P, Suzuki T, Ohya S. Histone Deacetylases Enhance Ca 2+-Activated K⁺ Channel K Ca3.1 Expression in Murine Inflammatory CD4⁺ T Cells. Int J Mol Sci 2018; 19:ijms19102942. [PMID: 30262728 PMCID: PMC6213394 DOI: 10.3390/ijms19102942] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 12/11/2022] Open
Abstract
The up-regulated expression of the Ca2+-activated K+ channel KCa3.1 in inflammatory CD4+ T cells has been implicated in the pathogenesis of inflammatory bowel disease (IBD) through the enhanced production of inflammatory cytokines, such as interferon-γ (IFN-γ). However, the underlying mechanisms have not yet been elucidated. The objective of the present study is to clarify the involvement of histone deacetylases (HDACs) in the up-regulation of KCa3.1 in the CD4+ T cells of IBD model mice. The expression levels of KCa3.1 and its regulators, such as function-modifying molecules and transcription factors, were quantitated using a real-time polymerase chain reaction (PCR) assay, Western blotting, and depolarization responses, which were induced by the selective KCa3.1 blocker TRAM-34 (1 μM) and were measured using a voltage-sensitive fluorescent dye imaging system. The treatment with 1 μM vorinostat, a pan-HDAC inhibitor, for 24 h repressed the transcriptional expression of KCa3.1 in the splenic CD4+ T cells of IBD model mice. Accordingly, TRAM-34-induced depolarization responses were significantly reduced. HDAC2 and HDAC3 were significantly up-regulated in the CD4+ T cells of IBD model mice. The down-regulated expression of KCa3.1 was observed following treatments with the selective inhibitors of HDAC2 and HDAC3. The KCa3.1 K+ channel regulates inflammatory cytokine production in CD4+ T cells, mediating epigenetic modifications by HDAC2 and HDAC3.
Collapse
Affiliation(s)
- Miki Matsui
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Kyoko Terasawa
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| | - Junko Kajikuri
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Hiroaki Kito
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Kyoko Endo
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Pattaporn Jaikhan
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 403-8334, Japan.
| | - Takayoshi Suzuki
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 403-8334, Japan.
| | - Susumu Ohya
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| |
Collapse
|
38
|
Differential DNA methylation of potassium channel KCa3.1 and immune signalling pathways is associated with infant immune responses following BCG vaccination. Sci Rep 2018; 8:13086. [PMID: 30166570 PMCID: PMC6117309 DOI: 10.1038/s41598-018-31537-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 08/21/2018] [Indexed: 12/14/2022] Open
Abstract
Bacillus Calmette–Guérin (BCG) is the only licensed vaccine for tuberculosis (TB) and induces highly variable protection against pulmonary disease in different countries. We hypothesised that DNA methylation is one of the molecular mechanisms driving variability in BCG-induced immune responses. DNA methylation in peripheral blood mononuclear cells (PBMC) from BCG vaccinated infants was measured and comparisons made between low and high BCG-specific cytokine responders. We found 318 genes and 67 pathways with distinct patterns of DNA methylation, including immune pathways, e.g. for T cell activation, that are known to directly affect immune responses. We also highlight signalling pathways that could indirectly affect the BCG-induced immune response: potassium and calcium channel, muscarinic acetylcholine receptor, G Protein coupled receptor (GPCR), glutamate signalling and WNT pathways. This study suggests that in addition to immune pathways, cellular processes drive vaccine-induced immune responses. Our results highlight mechanisms that require consideration when designing new TB vaccines.
Collapse
|
39
|
Funakoshi Y, Azuma A, Ishikawa M, Itsuki S, Tamura Y, Kanemaru K, Hirai S, Oyama Y. Cytometrical analysis of the adverse effects of indican, indoxyl, indigo, and indirubin on rat thymic lymphocytes. Toxicol Res (Camb) 2018; 7:513-520. [PMID: 30090601 PMCID: PMC6062345 DOI: 10.1039/c7tx00244k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 03/20/2018] [Indexed: 12/31/2022] Open
Abstract
Many businesses thrive by producing health supplements from agricultural products, as exemplified by the production of functional (or health) foods using plants traditionally cultivated in rural areas. Dyes, such as indican, indigo, indoxyl, and indirubin, present in dye plants, possess antibacterial, antifungal, and antiproliferative activities. However, these effects may also lead to cytotoxicity. Thus, studies on normal mammalian cells are necessary to identify cytotoxicity and prevent adverse effects of functional foods that contain these dyes. In this study, the effects of indican, indigo, indoxyl, and indirubin were evaluated by flow cytometry using appropriate fluorescent probes in rat thymic lymphocytes. Among the dyes analyzed, indirubin exerted distinct cellular activities. Treatment with indirubin (10-30 μM) increased the population of shrunken dead cells. The side scatter, but not forward scatter, increased in indirubin-treated living cells. It increased the population of annexin V-bound living and dead cells and that of dead cells without annexin V. Indirubin elevated intracellular Ca2+, but not Zn2+ levels. The cellular content of superoxide anions increased and that of glutathione decreased. Indirubin depolarized the cellular plasma and mitochondrial membranes. It did not potentiate or attenuate the cytotoxicity of A23187 (Ca2+ overload) and H2O2 (oxidative stress). The results suggested that indirubin induces both apoptotic and non-apoptotic cell death. It may be difficult to predict and prevent the adverse effects of indirubin due to its diverse activities on normal mammalian cells. Therefore, indirubin should be removed from products that contain dye plant extracts.
Collapse
Affiliation(s)
- Yurie Funakoshi
- Faculty of Integrated Arts and Sciences , Tokushima University , Tokushima 770-8502 , Japan . ; Tel: +81-88-656-7256
| | - Ayako Azuma
- Graduate School of Integrated Arts and Sciences , Tokushima University , Tokushima 770-8502 , Japan
| | - Mizuki Ishikawa
- Graduate School of Integrated Arts and Sciences , Tokushima University , Tokushima 770-8502 , Japan
| | - Satoru Itsuki
- Graduate School of Integrated Arts and Sciences , Tokushima University , Tokushima 770-8502 , Japan
| | - Yasuaki Tamura
- Graduate School of Integrated Arts and Sciences , Tokushima University , Tokushima 770-8502 , Japan
| | - Kaori Kanemaru
- Faculty of Bioscience and Bioindustry , Tokushima University , Tokushima 770-8513 , Japan
| | - Shogo Hirai
- Faculty of Integrated Arts and Sciences , Tokushima University , Tokushima 770-8502 , Japan . ; Tel: +81-88-656-7256
- Graduate School of Integrated Arts and Sciences , Tokushima University , Tokushima 770-8502 , Japan
| | - Yasuo Oyama
- Faculty of Integrated Arts and Sciences , Tokushima University , Tokushima 770-8502 , Japan . ; Tel: +81-88-656-7256
- Graduate School of Integrated Arts and Sciences , Tokushima University , Tokushima 770-8502 , Japan
- Faculty of Bioscience and Bioindustry , Tokushima University , Tokushima 770-8513 , Japan
| |
Collapse
|
40
|
Bozic I, Tesovic K, Laketa D, Adzic M, Jakovljevic M, Bjelobaba I, Savic D, Nedeljkovic N, Pekovic S, Lavrnja I. Voltage Gated Potassium Channel Kv1.3 Is Upregulated on Activated Astrocytes in Experimental Autoimmune Encephalomyelitis. Neurochem Res 2018; 43:1020-1034. [PMID: 29574670 DOI: 10.1007/s11064-018-2509-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/13/2018] [Accepted: 03/17/2018] [Indexed: 12/13/2022]
Abstract
Kv1.3 is a voltage gated potassium channel that has been implicated in pathophysiology of multiple sclerosis (MS). In the present study we investigated temporal and cellular expression pattern of this channel in the lumbar part of spinal cords of animals with experimental autoimmune encephalomyelitis (EAE), animal model of MS. EAE was actively induced in female Dark Agouti rats. Expression of Kv1.3 was analyzed at different time points of disease progression, at the onset, peak and end of EAE. We here show that Kv1.3 increased by several folds at the peak of EAE at both gene and protein level. Double immunofluorescence analyses demonstrated localization of Kv1.3 on activated microglia, macrophages, and reactive astrocytes around inflammatory lesions. In vitro experiments showed that pharmacological block of Kv1.3 in activated astrocytes suppresses the expression of proinflammatory mediators, suggesting a role of this channel in inflammation. Our results support the hypothesis that Kv1.3 may be a therapeutic target of interest for MS and add astrocytes to the list of cells whose activation would be suppressed by inhibiting Kv1.3 in inflammatory conditions.
Collapse
Affiliation(s)
- Iva Bozic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia.
| | - Katarina Tesovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Danijela Laketa
- Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Marija Adzic
- Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Marija Jakovljevic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Ivana Bjelobaba
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Danijela Savic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Nadezda Nedeljkovic
- Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Sanja Pekovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Irena Lavrnja
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| |
Collapse
|
41
|
Nishimura-Danjobara Y, Oyama K, Yokoigawa K, Oyama Y. Hyperpolarization by N-(3-oxododecanoyl)-l-homoserine-lactone, a quorum sensing molecule, in rat thymic lymphocytes. Chem Biol Interact 2018; 283:91-96. [PMID: 29427588 DOI: 10.1016/j.cbi.2018.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/22/2018] [Accepted: 02/05/2018] [Indexed: 11/25/2022]
Abstract
To study the adverse effects of N-(3-oxododecanoyl)-l-homoserine-lactone (ODHL), a quorum sensing molecule, on mammalian host cells, its effect on membrane potential was examined in rat thymic lymphocytes using flow cytometric techniques with a voltage-sensitive fluorescent probe. As 3-300 μM ODHL elicited hyperpolarization, it is likely that it increases membrane K+ permeability because hyperpolarization is directly linked to changing K+ gradient across membranes, but not Na+ and Cl- gradients. ODHL did not increase intracellular Ca2+ concentration. ODHL also produced a response in the presence of an intracellular Zn2+ chelator. Thus, it is unlikely that intracellular Ca2+ and Zn2+ are attributed to the response. Quinine, a non-specific K+ channel blocker, greatly reduced hyperpolarization. However, because charybdotoxin, tetraethylammonium chloride, 4-aminopyridine, and glibenclamide did not affect it, it is pharmacologically hypothesized that Ca2+-activated K+ channels, voltage-gated K+ channels, and ATP-sensitive K+ channels are not involved in ODHL-induced hyperpolarization. Although the K+ channels responsible for ODHL-induced hyperpolarization have not been identified, it is suggested that ODHL can elicit hyperpolarization in mammalian host cells, disturbing cellular functions.
Collapse
Affiliation(s)
- Yumiko Nishimura-Danjobara
- Department of Food Science, Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Keisuke Oyama
- Department of Food Science, Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Kumio Yokoigawa
- Department of Food Science, Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Yasuo Oyama
- Department of Food Science, Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan.
| |
Collapse
|
42
|
Lowinus T, Bose T, Busse S, Busse M, Reinhold D, Schraven B, Bommhardt UHH. Immunomodulation by memantine in therapy of Alzheimer's disease is mediated through inhibition of Kv1.3 channels and T cell responsiveness. Oncotarget 2018; 7:53797-53807. [PMID: 27462773 PMCID: PMC5288222 DOI: 10.18632/oncotarget.10777] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 07/09/2016] [Indexed: 12/20/2022] Open
Abstract
Memantine is approved for the treatment of advanced Alzheimer's disease (AD) and reduces glutamate-mediated neuronal excitotoxicity by antagonism of N-methyl-D-aspartate receptors. In the pathophysiology of AD immune responses deviate and infectious side effects are observed during memantine therapy. However, the particular effects of memantine on human T lymphocytes are unresolved. Here, we provide evidence that memantine blocks Kv1.3 potassium channels, inhibits CD3-antibody- and alloantigen-induced proliferation and suppresses chemokine-induced migration of peripheral blood T cells of healthy donors. Concurrent with the in vitro data, CD4+ T cells from AD patients receiving therapeutic doses of memantine show a transient decline of Kv1.3 channel activity and a long-lasting reduced proliferative response to alloantigens in mixed lymphocyte reactions. Furthermore, memantine treatment provokes a profound depletion of peripheral blood memory CD45RO+ CD4+ T cells. Thus, standard doses of memantine profoundly reduce T cell responses in treated patients through blockade of Kv1.3 channels. This may normalize deviant immunopathology in AD and contribute to the beneficial effects of memantine, but may also account for the enhanced infection rate.
Collapse
Affiliation(s)
- Theresa Lowinus
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Tanima Bose
- Molecular Physiology, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Current address: Lee Kong Chian School of Medicine, Singapore
| | - Stefan Busse
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Mandy Busse
- Department of Pediatric Pulmonology & Allergology, Medical University of Hannover, Hannover, Germany
| | - Dirk Reinhold
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Department of Immune Control, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ursula H H Bommhardt
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| |
Collapse
|
43
|
Mathew John C, Khaddaj Mallat R, George G, Kim T, Mishra RC, Braun AP. Pharmacologic targeting of endothelial Ca 2+-activated K + channels: A strategy to improve cardiovascular function. Channels (Austin) 2018; 12:126-136. [PMID: 29577810 PMCID: PMC5972810 DOI: 10.1080/19336950.2018.1454814] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 03/15/2018] [Indexed: 12/17/2022] Open
Abstract
Endothelial small and intermediate-conductance, Ca2+-activated K+ channels (KCa2.3 and KCa3.1, respectively) play an important role in the regulation of vascular function and systemic blood pressure. Growing evidence indicates that they are intimately involved in agonist-evoked vasodilation of small resistance arteries throughout the circulation. Small molecule activators of KCa2.x and 3.1 channels, such as SKA-31, can acutely inhibit myogenic tone in isolated resistance arteries, induce effective vasodilation in intact vascular beds, such as the coronary circulation, and acutely decrease systemic blood pressure in vivo. The blood pressure-lowering effect of SKA-31, and early indications of improvement in endothelial dysfunction suggest that endothelial KCa channel activators could eventually be developed into a new class of endothelial targeted agents to combat hypertension or atherosclerosis. This review summarises recent insights into the activation of endothelial Ca2+ activated K+ channels in various vascular beds, and how tools, such as SKA-31, may be beneficial in disease-related conditions.
Collapse
Affiliation(s)
- Cini Mathew John
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rayan Khaddaj Mallat
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Grace George
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Taeyeob Kim
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ramesh C. Mishra
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrew P. Braun
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
44
|
Takeda M, Oyama K, Kamemura N, Kanemaru K, Yuasa K, Yokoigawa K, Oyama Y. Change in plasma membrane potential of rat thymocytes by tert-butylhydroquinone, a food additive: Possible risk on lymphocytes. Food Chem Toxicol 2017; 109:296-301. [PMID: 28899772 DOI: 10.1016/j.fct.2017.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/25/2017] [Accepted: 09/08/2017] [Indexed: 12/12/2022]
Abstract
Tertiary butylhydroquinone (TBHQ) is a food additive and has various beneficial actions under in vitro and in vivo experimental conditions. Therefore, it is necessary to collect additional data on the toxicity of TBHQ in order to avoid adverse effects during clinical applications. Changes in plasma membrane potential are associated with changes in physiological functions even in non-excitable cells such as lymphocytes. Thus, compounds that affect membrane potential may modify some lymphocytic functions. The effect of TBHQ on plasma membrane potential was examined in rat thymocytes using flow cytometric techniques. Treatment of rat thymocytes with TBHQ caused hyperpolarization and then depolarization. The TBHQ-induced hyperpolarization was due to the activation of Ca2+-dependent K+ channels. TBHQ elevated intracellular Ca2+ levels. The depolarization by TBHQ was caused by a nonspecific increase in membrane ionic permeability. Both the sustained depolarization and elevation of intracellular Ca2+ level by TBHQ are thought to be adverse for thymocytes because such changes disturb membrane and intracellular signaling. The thymus is most active during neonatal and pre-adolescent periods. If TBHQ exerts adverse actions on thymocytes, it may result in an immunotoxic effect in neonates and adolescents.
Collapse
Affiliation(s)
- Maki Takeda
- Faculty of Integrated Arts and Sciences, Tokushima University, Tokushima 770-8502, Japan
| | - Keisuke Oyama
- Osaka University Hospital, Suita, Osaka 565-0871, Japan
| | - Norio Kamemura
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Kaori Kanemaru
- Faculty of Integrated Arts and Sciences, Tokushima University, Tokushima 770-8502, Japan; Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Keizo Yuasa
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Kumio Yokoigawa
- Faculty of Integrated Arts and Sciences, Tokushima University, Tokushima 770-8502, Japan; Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Yasuo Oyama
- Faculty of Integrated Arts and Sciences, Tokushima University, Tokushima 770-8502, Japan; Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan.
| |
Collapse
|
45
|
Sim JH, Kim KS, Park H, Kim KJ, Lin H, Kim TJ, Shin HM, Kim G, Lee DS, Park CW, Lee DH, Kang I, Kim SJ, Cho CH, Doh J, Kim HR. Differentially Expressed Potassium Channels Are Associated with Function of Human Effector Memory CD8 + T Cells. Front Immunol 2017; 8:859. [PMID: 28791017 PMCID: PMC5522836 DOI: 10.3389/fimmu.2017.00859] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 07/07/2017] [Indexed: 12/12/2022] Open
Abstract
The voltage-gated potassium channel, Kv1.3, and the Ca2+-activated potassium channel, KCa3.1, regulate membrane potentials in T cells, thereby controlling T cell activation and cytokine production. However, little is known about the expression and function of potassium channels in human effector memory (EM) CD8+ T cells that can be further divided into functionally distinct subsets based on the expression of the interleukin (IL)-7 receptor alpha (IL-7Rα) chain. Herein, we investigated the functional expression and roles of Kv1.3 and KCa3.1 in EM CD8+ T cells that express high or low levels of the IL-7 receptor alpha chain (IL-7Rαhigh and IL-7Rαlow, respectively). In contrast to the significant activity of Kv1.3 and KCa3.1 in IL-7Rαhigh EM CD8+ T cells, IL-7Rαlow EM CD8+ T cells showed lower expression of Kv1.3 and insignificant expression of KCa3.1. Kv1.3 was involved in the modulation of cell proliferation and IL-2 production, whereas KCa3.1 affected the motility of EM CD8+ T cells. The lower motility of IL-7Rαlow EM CD8+ T cells was demonstrated using transendothelial migration and motility assays with intercellular adhesion molecule 1- and/or chemokine stromal cell-derived factor-1α-coated surfaces. Consistent with the lower migration property, IL-7Rαlow EM CD8+ T cells were found less frequently in human skin. Stimulating IL-7Rαlow EM CD8+ T cells with IL-2 or IL-15 increased their motility and recovery of KCa3.1 activity. Our findings demonstrate that Kv1.3 and KCa3.1 are differentially involved in the functions of EM CD8+ T cells. The weak expression of potassium channels in IL-7Rαlow EM CD8+ T cells can be revived by stimulation with IL-2 or IL-15, which restores the associated functions. This study suggests that IL-7Rαhigh EM CD8+ T cells with functional potassium channels may serve as a reservoir for effector CD8+ T cells during peripheral inflammation.
Collapse
Affiliation(s)
- Ji Hyun Sim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyung Soo Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyoungjun Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Kyung-Jin Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea
| | - Haiyue Lin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, South Korea
| | - Tae-Joo Kim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyun Mu Shin
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.,Medical Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Gwanghun Kim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong-Sup Lee
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Chan-Wook Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, South Korea
| | - Insoo Kang
- Department of Internal Medicine, Section of Rheumatology, Yale University School of Medicine, New Haven, CT, United States
| | - Sung Joon Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.,Medical Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Chung-Hyun Cho
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.,Medical Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Junsang Doh
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Hang-Rae Kim
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.,Medical Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
46
|
Bergmann R, Kubeil M, Zarschler K, Chhabra S, Tajhya RB, Beeton C, Pennington MW, Bachmann M, Norton RS, Stephan H. Distribution and kinetics of the Kv1.3-blocking peptide HsTX1[R14A] in experimental rats. Sci Rep 2017. [PMID: 28623364 PMCID: PMC5473807 DOI: 10.1038/s41598-017-03998-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The peptide HsTX1[R14A] is a potent and selective blocker of the voltage-gated potassium channel Kv1.3, which is a highly promising target for the treatment of autoimmune diseases and other conditions. In order to assess the biodistribution of this peptide, it was conjugated with NOTA and radiolabelled with copper-64. [64Cu]Cu-NOTA-HsTX1[R14A] was synthesised in high radiochemical purity and yield. The radiotracer was evaluated in vitro and in vivo. The biodistribution and PET studies after intravenous and subcutaneous injections showed similar patterns and kinetics. The hydrophilic peptide was rapidly distributed, showed low accumulation in most of the organs and tissues, and demonstrated high molecular stability in vitro and in vivo. The most prominent accumulation occurred in the epiphyseal plates of trabecular bones. The high stability and bioavailability, low normal-tissue uptake of [64Cu]Cu-NOTA-HsTX1[R14A], and accumulation in regions of up-regulated Kv channels both in vitro and in vivo demonstrate that HsTX1[R14A] represents a valuable lead for conditions treatable by blockade of the voltage-gated potassium channel Kv1.3. The pharmacokinetics shows that both intravenous and subcutaneous applications are viable routes for the delivery of this potent peptide.
Collapse
Affiliation(s)
- Ralf Bergmann
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, D-01328, Germany
| | - Manja Kubeil
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, D-01328, Germany.,School of Chemistry, Monash University, Melbourne, Victoria, 3800, Australia
| | - Kristof Zarschler
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, D-01328, Germany
| | - Sandeep Chhabra
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Rajeev B Tajhya
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Michael Bachmann
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, D-01328, Germany
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia.
| | - Holger Stephan
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, D-01328, Germany.
| |
Collapse
|
47
|
Ohanyan V, Yin L, Bardakjian R, Kolz C, Enrick M, Hakobyan T, Luli J, Graham K, Khayata M, Logan S, Kmetz J, Chilian WM. Kv1.3 channels facilitate the connection between metabolism and blood flow in the heart. Microcirculation 2017; 24:10.1111/micc.12334. [PMID: 28504408 PMCID: PMC5433265 DOI: 10.1111/micc.12334] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 10/23/2016] [Accepted: 11/01/2016] [Indexed: 12/17/2022]
Abstract
The connection between metabolism and flow in the heart, metabolic dilation, is essential for cardiac function. We recently found redox-sensitive Kv1.5 channels play a role in coronary metabolic dilation; however, more than one ion channel likely plays a role in this process as animals null for these channels still showed limited coronary metabolic dilation. Accordingly, we examined the role of another Kv1 family channel, the energetically linked Kv1.3 channel, in coronary metabolic dilation. We measured myocardial blood flow (contrast echocardiography) during norepinephrine-induced increases in cardiac work (heart rate x mean arterial pressure) in WT, WT mice given correolide (preferential Kv1.3 antagonist), and Kv1.3-null mice (Kv1.3-/- ). We also measured relaxation of isolated small arteries mounted in a myograph. During increased cardiac work, myocardial blood flow was attenuated in Kv1.3-/- and in correolide-treated mice. In isolated vessels from Kv1.3-/- mice, relaxation to H2 O2 was impaired (vs WT), but responses to adenosine and acetylcholine were equivalent to WT. Correolide reduced dilation to adenosine and acetylcholine in WT and Kv1.3-/- , but had no effect on H2 O2 -dependent dilation in vessels from Kv1.3-/- mice. We conclude that Kv1.3 channels participate in the connection between myocardial blood flow and cardiac metabolism.
Collapse
Affiliation(s)
- Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | | | - Christopher Kolz
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Molly Enrick
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Tatevik Hakobyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Jordan Luli
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Kathleen Graham
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | | | - Suzanna Logan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - John Kmetz
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| |
Collapse
|
48
|
Schwartz AB, Kapur A, Wang W, Huang Z, Fardone E, Palui G, Mattoussi H, Fadool DA. Margatoxin-bound quantum dots as a novel inhibitor of the voltage-gated ion channel Kv1.3. J Neurochem 2016; 140:404-420. [PMID: 27861889 DOI: 10.1111/jnc.13891] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 10/24/2016] [Accepted: 10/31/2016] [Indexed: 01/01/2023]
Abstract
Venom-derived ion channel inhibitors have strong channel selectivity, potency, and stability; however, tracking delivery to their target can be challenging. Herein, we utilized luminescent quantum dots (QDs) conjugated to margatoxin (MgTx) as a traceable vehicle to target a voltage-dependent potassium channel, Kv1.3, which has a select distribution and well-characterized role in immunity, glucose metabolism, and sensory ability. We screened both unconjugated (MgTx) and conjugated MgTx (QD-MgTx) for their ability to inhibit Shaker channels Kv1.1 to Kv1.7 using patch-clamp electrophysiology in HEK293 cells. Our data indicate that MgTx inhibits 79% of the outward current in Kv1.3-transfected cells and that the QD-MgTx conjugate is able to achieve a similar level of block, albeit a slightly reduced efficacy (66%) and at a slower time course (50% block by 10.9 ± 1.1 min, MgTx; vs. 15.3 ± 1.2 min, QD-MgTx). Like the unbound peptide, the QD-MgTx conjugate inhibits both Kv1.3 and Kv1.2 at a 1 nM concentration, whereas it does not inhibit other screened Shaker channels. We tested the ability of QD-MgTx to inhibit native Kv1.3 expressed in the mouse olfactory bulb (OB). In brain slices of the OB, the conjugate acted similarly to MgTx to inhibit Kv1.3, causing an increased action potential firing frequency attributed to decreased intraburst duration rather than interspike interval. Our data demonstrate a retention of known biophysical properties associated with block of the vestibule of Kv1.3 by QD-MgTx conjugate compared to that of MgTx, inferring QDs could provide a useful tool to deliver ion channel inhibitors to targeted tissues in vivo.
Collapse
Affiliation(s)
- Austin B Schwartz
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| | - Anshika Kapur
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, USA
| | - Wentao Wang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, USA
| | - Zhenbo Huang
- Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Erminia Fardone
- Program in Neuroscience, Florida State University, Tallahassee, Florida, USA.,Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Goutam Palui
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, USA
| | - Hedi Mattoussi
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, USA
| | - Debra Ann Fadool
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA.,Program in Neuroscience, Florida State University, Tallahassee, Florida, USA.,Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
49
|
Huang SS, Zhang QB, Yuan QY, He SL, Zhang YM. Inhibitory effects of telmisartan on culture and proliferation of and Kv1.3 potassium channel expression in peripheral blood CD4+ T lymphocytes from Xinjiang Kazakh patients with hypertension. J Renin Angiotensin Aldosterone Syst 2016; 17:1470320316674876. [PMID: 27765883 PMCID: PMC5843919 DOI: 10.1177/1470320316674876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/16/2016] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION Activation of T lymphocytes, for which potassium channels are essential, is involved in the development of hypertension. In this study, we explored the inhibitory effects of telmisartan on the culture and proliferation of and Kv1.3 potassium channel expression in peripheral blood CD4+ T lymphocytes derived from Xinjiang Kazakh patients with hypertension. METHODS CD4+ T-cell samples from hypertensive Kazakh patients and healthy Kazakh people were divided into healthy control, case control, telmisartan, and 4-aminopytidine groups. Changes in the expression levels of interleukin (IL)-6 and IL-17 in the blood of the healthy control and case control subjects were detected by enzyme-linked immunosorbent assay. Peripheral blood CD4+ T lymphocytes were first activated and proliferated in vitro and then incubated for 0, 24, and 48 h under various treatment conditions. Thereafter, changes in CD4+ T-lymphocytic proliferation were determined using Cell Counting Kit-8 and microscope photography. Changes in messenger RNA (mRNA) and protein expression of the Kv1.3 potassium channel in CD4+ T lymphocytes were detected using real-time quantitative polymerase chain reaction and Western blots, respectively. RESULTS The IL-6 and IL-17 expression levels were significantly higher in the blood of the hypertensive Kazakh patients than in the healthy Kazakh people. Telmisartan inhibited T-lymphocytic proliferation, as well as the mRNA and protein expression of the Kv1.3 potassium channel in CD4+ T lymphocytes, and the inhibitory effects were time-dependent, with the strongest inhibition observed after 48 h and significantly weaker inhibition observed after 24 h of treatment. CONCLUSIONS Telmisartan may potentially regulate hypertensive inflammatory responses by inhibiting T-lymphocytic proliferation and Kv1.3 potassium channel expression in CD4+ T lymphocytes.
Collapse
Affiliation(s)
- Sha-Sha Huang
- Heart Centre, First Affiliated Hospital of Xinjiang Medical University, China
| | - Qiu-Bing Zhang
- Heart Centre, First Affiliated Hospital of Xinjiang Medical University, China
| | - Qing-Yan Yuan
- Heart Centre, First Affiliated Hospital of Xinjiang Medical University, China
| | - Si-Li He
- Heart Centre, First Affiliated Hospital of Xinjiang Medical University, China
| | - Yuan-Ming Zhang
- Heart Centre, First Affiliated Hospital of Xinjiang Medical University, China
| |
Collapse
|
50
|
Dale E, Staal RGW, Eder C, Möller T. KCa 3.1-a microglial target ready for drug repurposing? Glia 2016; 64:1733-41. [PMID: 27121595 DOI: 10.1002/glia.22992] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 03/29/2016] [Accepted: 04/03/2016] [Indexed: 01/25/2023]
Abstract
Over the past decade, glial cells have attracted attention for harboring unexploited targets for drug discovery. Several glial targets have attracted de novo drug discovery programs, as highlighted in this GLIA Special Issue. Drug repurposing, which has the objective of utilizing existing drugs as well as abandoned, failed, or not yet pursued clinical development candidates for new indications, might provide a faster opportunity to bring drugs for glial targets to patients with unmet needs. Here, we review the potential of the intermediate-conductance calcium-activated potassium channels KCa 3.1 as the target for such a repurposing effort. We discuss the data on KCa 3.1 expression on microglia in vitro and in vivo and review the relevant literature on the two KCa 3.1 inhibitors TRAM-34 and Senicapoc. Finally, we provide an outlook of what it might take to harness the potential of KCa 3.1 as a bona fide microglial drug target. GLIA 2016;64:1733-1741.
Collapse
Affiliation(s)
- Elena Dale
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey
| | - Roland G W Staal
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey
| | - Claudia Eder
- Institute for Infection and Immunity, St. George's, University of London, United Kingdom
| | - Thomas Möller
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey
| |
Collapse
|