1
|
Zaniboni BA, Oliveira VSD, Leite GAA, Grinevicius VMADS, Pedrosa RC, Silva FRMB. Pyriproxyfen Disrupts the Ongoing Spermatogenesis Wave in Danio rerio Potentially Mediated by Voltage-Dependent Calcium Channels and Protein Kinase C. J Appl Toxicol 2025. [PMID: 40308215 DOI: 10.1002/jat.4801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/14/2025] [Accepted: 04/19/2025] [Indexed: 05/02/2025]
Abstract
Pyriproxyfen (PPX) is an analog of the juvenile hormone from insects. Following our previous studies, for the ex vivo short-term effect, we chose 10-9 M pyriproxyfen to analyze the morphology of spermatogenesis wave cells. In silico docking and ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) studies were carried out to preliminarily predict possible interaction modes between PPX and the T-type voltage-dependent calcium channel (T-VDCC), as well as with protein kinase C (PKC), as we previously reported by using pharmacological approach. The in silico ADMET evaluations revealed that PPX demonstrates notable lipophilicity. Moreover, PPX is predicted to inhibit the enzymatic activity of CYP1A2, CYP2C19, CYP2C9, and CYP2D6. Furthermore, in silico molecular docking analyses revealed that PPX has the potential to interact with the T-VDCC through hydrogen bonds with Gln1653 and hydrophobic interactions with Leu291, Phe322, Phe1607, and Leu1656. Possible interactions of PPX with PKC involve ionic bonding with Lys463, hydrogen bonds with His592, and hydrophobic interactions with Lys463, Val596, Gly591, Phe593, Lys611, Asp711, and Leu714 reinforcing these both targets to PPX. In summary, short-term PPX exposure influenced the morphology of testicular cells (spermatids, spermatozoa, and Leydig cells) through interactions with molecular targets. Findings reveal the bimodal effects (on morphology and signaling) of this compound on specific cells within the spermatogenic wave, endocrine cells, and signal transduction proteins. This interference may impair reproduction and lead to male infertility. In addition, the prediction from both molecular docking and ADMET supported our in vitro mechanistic analysis firstly reported in the testis of Danio rerio.
Collapse
Affiliation(s)
- Bruna Antunes Zaniboni
- Instituto de Bioeletricidade Celular (IBIOCEL): Ciência & Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Vanessa Staldoni de Oliveira
- Instituto de Bioeletricidade Celular (IBIOCEL): Ciência & Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Gabriel Adan Araujo Leite
- Instituto de Bioeletricidade Celular (IBIOCEL): Ciência & Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | - Rozangela Curi Pedrosa
- Instituto de Bioeletricidade Celular (IBIOCEL): Ciência & Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Fátima Regina Mena Barreto Silva
- Instituto de Bioeletricidade Celular (IBIOCEL): Ciência & Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| |
Collapse
|
2
|
Alshehri SA, Wahab S, Almoyad MAA. In silico identification of potential protein kinase C alpha inhibitors from phytochemicals from IMPPAT database for anticancer therapeutics: a virtual screening approach. J Biomol Struct Dyn 2024; 42:9463-9474. [PMID: 37643015 DOI: 10.1080/07391102.2023.2252086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023]
Abstract
Protein Kinase C alpha (PKCα) is a critical signaling molecule that plays a crucial role in various physiological processes, including cell growth, differentiation, and survival. Over the years, there has been a growing interest in targeting PKCα as a promising drug target for the treatment of various diseases, including cancer. Targeting PKCα can, therefore, serve as a potential strategy to prevent cancer progression and enhance the efficacy of conventional anticancer therapies. We conducted a systematic search for promising compounds for their anticancer potential that target PKCα using natural compounds from the IMPPAT database. The initial compounds were screened through various tests, including analysis of their physical and chemical properties, PAINS filter, ADMET analysis, PASS analysis, and specific interaction analysis. We selected those that showed high binding affinity and specificity to PKCα from the screened compounds, and we further analyzed them using molecular dynamics simulations (MDS) and principal component analysis (PCA). Various systematic parameters from the MDS analyses suggested that the protein-ligand complexes were stabilized throughout the simulation trajectories of 100 nanoseconds (ns). Our findings indicated that compounds Nicandrenone and Withaphysalin D bind to PKCα with high stability and affinity, making them potential candidates for further research in cancer therapeutics innovation in clinical contexts.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saad Ali Alshehri
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Ali Abdullah Almoyad
- Department of Basic Medical Sciences, College of Applied Medical Sciences in Khamis Mushyt, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
3
|
Singh N, Nandy SK, Jyoti A, Saxena J, Sharma A, Siddiqui AJ, Sharma L. Protein Kinase C (PKC) in Neurological Health: Implications for Alzheimer's Disease and Chronic Alcohol Consumption. Brain Sci 2024; 14:554. [PMID: 38928554 PMCID: PMC11201589 DOI: 10.3390/brainsci14060554] [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: 05/07/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Protein kinase C (PKC) is a diverse enzyme family crucial for cell signalling in various organs. Its dysregulation is linked to numerous diseases, including cancer, cardiovascular disorders, and neurological problems. In the brain, PKC plays pivotal roles in synaptic plasticity, learning, memory, and neuronal survival. Specifically, PKC's involvement in Alzheimer's Disease (AD) pathogenesis is of significant interest. The dysregulation of PKC signalling has been linked to neurological disorders, including AD. This review elucidates PKC's pivotal role in neurological health, particularly its implications in AD pathogenesis and chronic alcohol addiction. AD, characterised by neurodegeneration, implicates PKC dysregulation in synaptic dysfunction and cognitive decline. Conversely, chronic alcohol consumption elicits neural adaptations intertwined with PKC signalling, exacerbating addictive behaviours. By unravelling PKC's involvement in these afflictions, potential therapeutic avenues emerge, offering promise for ameliorating their debilitating effects. This review navigates the complex interplay between PKC, AD pathology, and alcohol addiction, illuminating pathways for future neurotherapeutic interventions.
Collapse
Affiliation(s)
- Nishtha Singh
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| | - Shouvik Kumar Nandy
- School of Pharmacy, Techno India University, Sector-V, Kolkata 700091, West Bengal, India;
| | - Anupam Jyoti
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara 391760, Gujarat, India;
| | - Juhi Saxena
- Department of Biotechnology, Parul Institute of Technology, Parul University, Vadodara 391760, Gujarat, India;
| | - Aditi Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail 55476, Saudi Arabia
| | - Lalit Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| |
Collapse
|
4
|
Chen H, Li G, Liu Y, Ji S, Li Y, Xiang J, Zhou L, Gao H, Zhang W, Sun X, Fu X, Li B. Pleiotropic Roles of CXCR4 in Wound Repair and Regeneration. Front Immunol 2021; 12:668758. [PMID: 34122427 PMCID: PMC8194072 DOI: 10.3389/fimmu.2021.668758] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/26/2021] [Indexed: 12/27/2022] Open
Abstract
Wound healing is a multi-step process that includes multiple cellular events such as cell proliferation, cell adhesion, and chemotactic response as well as cell apoptosis. Accumulating studies have documented the significance of stromal cell-derived factor-1 (SDF-1)/C-X-C chemokine receptor 4 (CXCR4) signaling in wound repair and regeneration. However, the molecular mechanism of regeneration is not clear. This review describes various types of tissue regeneration that CXCR4 participates in and how the efficiency of regeneration is increased by CXCR4 overexpression. It emphasizes the pleiotropic effects of CXCR4 in regeneration. By delving into the specific molecular mechanisms of CXCR4, we hope to provide a theoretical basis for tissue engineering and future regenerative medicine.
Collapse
Affiliation(s)
- Huating Chen
- Department of Wound Repair Surgery, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | | | - Yiqiong Liu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuaifei Ji
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Li
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China.,Department of Southern Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Jiangbing Xiang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China.,Department of School of Biological Engineering, Chongqing University, Chongqing, China
| | - Laixian Zhou
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Huanhuan Gao
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenwen Zhang
- Department of Wound Repair Surgery, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Binghui Li
- Department of Wound Repair Surgery, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Mukherjee A, Roy S, Patidar A, Bodhale N, Dandapat J, Saha B, Sarkar A. TLR2 dimer-specific ligands selectively activate protein kinase C isoforms in Leishmania infection. Immunology 2021; 164:318-331. [PMID: 34021910 DOI: 10.1111/imm.13373] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 04/18/2021] [Accepted: 05/10/2021] [Indexed: 11/30/2022] Open
Abstract
Of the thirteen Toll-like receptors (TLRs) in mice, TLR2 has a unique ability of forming heterodimers with TLR1 and TLR6. Such associations lead to selective cellular signalling and cellular responses such as cytokine expression. One of the signalling intermediates is protein kinase C (PKC); of which, eight isoforms are expressed in macrophages. Leishmania-a protozoan parasite that resides and replicates in macrophages-selectively modulates PKC-α, PKC-β, PKC-δ and PKC-ζ isoforms in macrophages. As TLR2 plays significant roles in Leishmania infection, we examined whether these PKC isoforms play selective roles in TLR2 signalling and TLR2-induced anti-leishmanial functions. We observed that the TLR2 ligands-Pam3 CSK4 (TLR1/2), PGN (TLR2/2) and FSL (TLR2/6)-differentially phosphorylated and translocated PKC-α, PKC-β, PKC-δ and PKC-ζ isoforms to cell membrane in uninfected and L. major-infected macrophages. The PKC isoform-specific inhibitors differentially altered IL-10 and IL-12 expression, Th1 and Th2 responses and anti-leishmanial effects in macrophages and in BALB/c mice. While PKC isoforms' inhibitors had insignificant effects on the Pam3CSK4-induced anti-leishmanial functions, PGN-induced pro-leishmanial effects were enhanced by PKC-(α + β) inhibitors, whereas PKC-(δ + ζ) inhibitors enhanced the anti-leishmanial effects of FSL. These results indicated that the ligand-induced TLR2 dimerization triggered differential dose-dependent and kinetic profiles of PKC isoform activation and that selective targeting of PKC isoforms using their respective inhibitors in combination significantly modulated TLR2-induced anti-leishmanial functions. To the best of our knowledge, this is the first demonstration of TLR2 dimer signalling through PKC isoforms and TLR2-induced PKC isoform-targeted anti-leishmanial therapy.
Collapse
Affiliation(s)
| | - Sayoni Roy
- National Centre for Cell Science [NCCS], Pune, India
| | - Ashok Patidar
- National Centre for Cell Science [NCCS], Pune, India
| | - Neelam Bodhale
- National Centre for Cell Science [NCCS], Pune, India.,JBNSTS, Calcutta, India
| | | | - Bhaskar Saha
- Trident Academy of Creative Technology [TACT], Bhubaneswar, India.,National Centre for Cell Science [NCCS], Pune, India
| | - Arup Sarkar
- Trident Academy of Creative Technology [TACT], Bhubaneswar, India
| |
Collapse
|
6
|
Signal Transduction in Immune Cells and Protein Kinases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:133-149. [PMID: 33539014 DOI: 10.1007/978-3-030-49844-3_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Immune response relies upon several intracellular signaling events. Among the protein kinases involved in these pathways, members of the protein kinase C (PKC) family are prominent molecules because they have the capacity to acutely and reversibly modulate effector protein functions, controlling both spatial distribution and dynamic properties of the signals. Different PKC isoforms are involved in distinct signaling pathways, with selective functions in a cell-specific manner.In innate system, Toll-like receptor signaling is the main molecular event triggering effector functions. Various isoforms of PKC can be common to different TLRs, while some of them are specific for a certain type of TLR. Protein kinases involvement in innate immune cells are presented within the chapter emphasizing their coordination in many aspects of immune cell function and, as important players in immune regulation.In adaptive immunity T-cell receptor and B-cell receptor signaling are the main intracellular pathways involved in seminal immune specific cellular events. Activation through TCR and BCR can have common intracellular pathways while others can be specific for the type of receptor involved or for the specific function triggered. Various PKC isoforms involvement in TCR and BCR Intracellular signaling will be presented as positive and negative regulators of the immune response events triggered in adaptive immunity.
Collapse
|
7
|
Li XP, Zhang J. A live attenuated Edwardsiella tarda vaccine induces immunological expression pattern in Japanese flounder (Paralichthys olivaceus) in the early phase of immunization. Comp Biochem Physiol C Toxicol Pharmacol 2021; 239:108872. [PMID: 32814144 DOI: 10.1016/j.cbpc.2020.108872] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/03/2020] [Accepted: 08/09/2020] [Indexed: 12/29/2022]
Abstract
A previous study showed that an attenuated Edwardsiella tarda strain, TXhfq, as a live vaccine could elicit protective immune effects in fish against E. tarda infection. In the current study, in order to clarify the molecular mechanism of fish immune response at the early stage after TXhfq vaccination, RNA-Seq technology was used to compare the transcriptomes of skin, intestine, and spleen between bath-vaccinated and unvaccinated Japanese flounder (Paralichthys olivaceus). An average of 46.6 million clean reads per library was obtained, ~88.04% of which were successfully mapped to the reference genome, and approximately 24,600 genes were detected in each sample. A total of 565, 878, and 1258 differential expression genes (DEGs) were found in skin, intestine, and spleen, respectively, including 1263 up-regulated genes and 1438 down-regulated genes. The DEGs exhibited different characteristics in each tissue. One hundred and sixteen DEGs belonging to six immune related categories were scrutinized, i.e., inflammatory factors, cytokines, complement and coagulation system, mucins, phagocytosis, and antigen processing and presentation. A protein-protein interaction network was constructed to get the interaction network between immune genes during the early stage of immunization. The top six hub genes highly regulated by TXhfq formed complicated interaction relationship with each other, which were involved in immune processes, notably inflammation and phagocytosis. Our results provide valuable information for the understanding of the immune mechanism underlying the protection of live attenuated vaccines in fish.
Collapse
Affiliation(s)
- Xue-Peng Li
- CAS Key Laboratory of Experimental Marine Biology, CAS Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; School of Ocean, Yantai University, Yantai, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jian Zhang
- CAS Key Laboratory of Experimental Marine Biology, CAS Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; School of Ocean, Yantai University, Yantai, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
8
|
Deka SJ, Trivedi V. Potentials of PKC in Cancer Progression and Anticancer Drug Development. Curr Drug Discov Technol 2020; 16:135-147. [PMID: 29468974 DOI: 10.2174/1570163815666180219113614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/29/2018] [Accepted: 02/12/2018] [Indexed: 01/07/2023]
Abstract
PKC is a family of serine-threonine kinases which play crucial roles in the regulation of important signal transduction pathways in mammalian cell-biology. These enzymes are themselves regulated by various molecules that can serve as ligands to the regulatory domains and translocate PKC to membrane for activity. The role of PKC in the modulation of both proliferative and apoptotic signaling in cancer has become a subject of immense interest after it was discovered that PKC regulates a myriad of enzymes and transcription factors involved in carcinogenic signaling. Therefore, PKC has served as an attractive target for the development of newer generation of anti-cancer drugs. The following review discusses the potential of PKC to be regarded as a target for anti-cancer therapy. We also review all the molecules that have been discovered so far to be regulators/activators/inhibitors of PKC and also how far these molecules can be considered as potential candidates for anti-cancer drug development based on PKC.
Collapse
Affiliation(s)
- Suman J Deka
- Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati-781039, Assam, India
| | - Vishal Trivedi
- Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati-781039, Assam, India
| |
Collapse
|
9
|
Kobayakawa T, Takano H, Ishii T, Tsuji K, Ohashi N, Nomura W, Furuta T, Tamamura H. Synthesis of hydrophilic caged DAG-lactones for chemical biology applications. Org Biomol Chem 2020; 18:4217-4223. [PMID: 32432608 DOI: 10.1039/d0ob00807a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The 6-bromo-7-hydroxy-coumarin-4-ylmethyl (Bhc) group has been used widely in cage chemistry because of its high molar absorptivity and photolytic efficiency. One of the drawbacks of coumarins however is their low aqueous solubility. Aqueous solubility is important in the behavior of caged compounds because hydrophobic caged compounds might be aggregated in physiological conditions and consequently the photocleavage would be impaired. The 8-azacoumarin-4-ylmethyl derivatives with bromine (8-aza-Bhc) or iodine (8-aza-Ihc), which were previously developed in this laboratory, have aqueous solubilities that are higher than those of related coumarins. Here, to improve the hydrophilicity and management of caged diacylglycerol lactones (DAG-lactones), 8-aza-Bhc and 8-aza-Ihc were introduced into the DAG-lactone structure. The synthesized caged compounds showed high hydrophilicity compared with the parent Bhc-caged DAG-lactone, and the 8-aza-Ihc-caged DAG-lactone (2) showed excellent photolytic efficiency, which allows rapid release of the DAG-lactone (1) by brief photoirradiation. The 8-aza-7-hydroxy-6-iodo-coumarin-4-ylmethyl group might be useful for caging of bioactive compounds, especially hydrophobic compounds.
Collapse
Affiliation(s)
- Takuya Kobayakawa
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 3-10 Kandasurugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Hikaru Takano
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 3-10 Kandasurugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Takahiro Ishii
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 3-10 Kandasurugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Kohei Tsuji
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 3-10 Kandasurugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Nami Ohashi
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 3-10 Kandasurugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Wataru Nomura
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 3-10 Kandasurugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Toshiaki Furuta
- Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi 274-8510, Japan
| | - Hirokazu Tamamura
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 3-10 Kandasurugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| |
Collapse
|
10
|
Knoell DL, Smith DA, Sapkota M, Heires AJ, Hanson CK, Smith LM, Poole JA, Wyatt TA, Romberger DJ. Insufficient zinc intake enhances lung inflammation in response to agricultural organic dust exposure. J Nutr Biochem 2019; 70:56-64. [PMID: 31153019 PMCID: PMC10575612 DOI: 10.1016/j.jnutbio.2019.04.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/18/2019] [Accepted: 04/25/2019] [Indexed: 11/15/2022]
Abstract
Organic dust exposure particularly within hog confinement facilities is a significant cause of airway inflammation and lung disease. In a cohort of Midwestern veterans with COPD and agricultural work exposure we observed reduced zinc intakes which were associated with decreased lung function. Because insufficient zinc intake is common within the U.S. and a potent modulator of innate immune function, we sought to determine whether deficits in zinc intake would impact the airway inflammatory response to hog confinement facility dust extract (HDE). Adult male C57BL/6 mice were randomized to zinc deficient or matched zinc sufficient diets for 3 weeks and subsequently treated with intranasal HDE inhalation or saline once or daily for 3 weeks while maintained on specific diets. Lavage fluid and lung tissue was collected. Conditions of zinc deficiency were also studied in macrophages exposed to HDE. Single and repetitive HDE inhalation exposure resulted in increased influx of total cells and neutrophils, increased mediator hyper-responsiveness (TNFα, IL-6, CXCL1, and amphiregulin), and enhanced tissue pathology that was more pronounced in zinc deficient mice compared to normal dietary counterparts. Airway inflammation was most pronounced in zinc deficient mice treated with repetitive HDE for 3 weeks. Similarly, macrophages maintained in a zinc deficient environment exhibited increased CXCL1 and IL-23 production as a result of increased NF-κB activation. Conclusion: Given the relatively high incidence of dietary deficiencies in agriculture workers, we anticipate that zinc intake, or a lack thereof, may play an important role in modulating the host response to organic dust exposure.
Collapse
Affiliation(s)
- Daren L Knoell
- The University of Nebraska Medical Center College of Pharmacy, Omaha, NE 68198.
| | - Deandra A Smith
- The University of Nebraska Medical Center College of Pharmacy, Omaha, NE 68198.
| | - Muna Sapkota
- The University of Nebraska Medical Center College of Pharmacy, Omaha, NE 68198.
| | - Art J Heires
- The University of Nebraska Medical Center College of Medicine, Omaha, NE 68198.
| | - Corrine K Hanson
- The University of Nebraska Medical Center College of Allied Health, Omaha, NE 68198.
| | - Lynette M Smith
- The University of Nebraska Medical Center College of Public Health, Omaha, NE 68198
| | - Jill A Poole
- The University of Nebraska Medical Center College of Medicine, Omaha, NE 68198.
| | - Todd A Wyatt
- The University of Nebraska Medical Center College of Medicine, Omaha, NE 68198; The University of Nebraska Medical Center College of Public Health, Omaha, NE 68198; VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105.
| | - Debra J Romberger
- The University of Nebraska Medical Center College of Medicine, Omaha, NE 68198.
| |
Collapse
|
11
|
Abstract
Doxorubicin-induced cardiotoxicity in childhood cancer survivors is a growing problem. The population of patients at risk for cardiovascular disease is steadily increasing, as five-year survival rates for all types of childhood cancers continue to improve. Doxorubicin affects the developing heart differently from the adult heart and in a subset of exposed patients, childhood exposure leads to late, irreversible cardiomyopathy. Notably, the prevalence of late-onset toxicity is increasing in parallel with improved survival. By the year 2020, it is estimated that there will be 500,000 childhood cancer survivors and over 50,000 of them will suffer from doxorubicin-induced cardiotoxicity. The majority of the research to-date, concentrated on childhood cancer survivors, has focused mostly on clinical outcomes through well-designed epidemiological and retrospective cohort studies. Preclinical studies have elucidated many of the cellular mechanisms that elicit acute toxicity in cardiomyocytes. However, more research is needed in the areas of early- and late-onset cardiotoxicity and more importantly improving the scientific understanding of how other cells present in the cardiac milieu are impacted by doxorubicin exposure. The overall goal of this review is to succinctly summarize the major clinical and preclinical studies focused on doxorubicin-induced cardiotoxicity. As the prevalence of patients affected by doxorubicin exposure continues to increase, it is imperative that the major gaps in existing research are identified and subsequently utilized to develop appropriate research priorities for the coming years. Well-designed preclinical research models will enhance our understanding of the pathophysiology of doxorubicin-induced cardiotoxicity and directly lead to better diagnosis, treatment, and prevention. © 2019 American Physiological Society. Compr Physiol 9:905-931, 2019.
Collapse
Affiliation(s)
- Trevi R. Mancilla
- University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Brian Iskra
- University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Gregory J. Aune
- University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| |
Collapse
|
12
|
Other Forms of Immunosuppression. KIDNEY TRANSPLANTATION - PRINCIPLES AND PRACTICE 2019. [PMCID: PMC7152196 DOI: 10.1016/b978-0-323-53186-3.00020-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
13
|
STIM- and Orai-mediated calcium entry controls NF-κB activity and function in lymphocytes. Cell Calcium 2018; 74:131-143. [PMID: 30048879 DOI: 10.1016/j.ceca.2018.07.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 12/24/2022]
Abstract
The central role of Ca2+ signaling in the development of functional immunity and tolerance is well established. These signals are initiated by antigen binding to cognate receptors on lymphocytes that trigger store operated Ca2+ entry (SOCE). The underlying mechanism of SOCE in lymphocytes involves TCR and BCR mediated activation of Stromal Interaction Molecule 1 and 2 (STIM1/2) molecules embedded in the ER membrane leading to their activation of Orai channels in the plasma membrane. STIM/Orai dependent Ca2+ signals guide key antigen induced lymphocyte development and function principally through direct regulation of Ca2+ dependent transcription factors. The role of Ca2+ signaling in NFAT activation and signaling is well known and has been studied extensively, but a wide appreciation and mechanistic understanding of how Ca2+ signals also shape the activation and specificity of NF-κB dependent gene expression has lagged. Here we discuss and interpret what is known about Ca2+ dependent mechanisms of NF-kB activation, including what is known and the gaps in our understanding of how these signals control lymphocyte development and function.
Collapse
|
14
|
Gao J, Sun Y, Sun Y, Chen C, Kausar S, Tian J, Zhu B, Liu C. Identification and function of cAMP response element binding protein in Oak silkworm Antheraea pernyi. J Invertebr Pathol 2018; 151:14-20. [DOI: 10.1016/j.jip.2017.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/17/2017] [Accepted: 10/23/2017] [Indexed: 12/23/2022]
|
15
|
Zinc Signals and Immunity. Int J Mol Sci 2017; 18:ijms18102222. [PMID: 29064429 PMCID: PMC5666901 DOI: 10.3390/ijms18102222] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/13/2017] [Accepted: 10/19/2017] [Indexed: 01/11/2023] Open
Abstract
Zinc homeostasis is crucial for an adequate function of the immune system. Zinc deficiency as well as zinc excess result in severe disturbances in immune cell numbers and activities, which can result in increased susceptibility to infections and development of especially inflammatory diseases. This review focuses on the role of zinc in regulating intracellular signaling pathways in innate as well as adaptive immune cells. Main underlying molecular mechanisms and targets affected by altered zinc homeostasis, including kinases, caspases, phosphatases, and phosphodiesterases, will be highlighted in this article. In addition, the interplay of zinc homeostasis and the redox metabolism in affecting intracellular signaling will be emphasized. Key signaling pathways will be described in detail for the different cell types of the immune system. In this, effects of fast zinc flux, taking place within a few seconds to minutes will be distinguish from slower types of zinc signals, also designated as “zinc waves”, and late homeostatic zinc signals regarding prolonged changes in intracellular zinc.
Collapse
|
16
|
Application of vitamin D and vitamin D analogs in acute myelogenous leukemia. Exp Hematol 2017; 50:1-12. [DOI: 10.1016/j.exphem.2017.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/16/2017] [Accepted: 01/26/2017] [Indexed: 12/18/2022]
|
17
|
Zuidscherwoude M, Dunlock VME, van den Bogaart G, van Deventer SJ, van der Schaaf A, van Oostrum J, Goedhart J, In 't Hout J, Hämmerling GJ, Tanaka S, Nadler A, Schultz C, Wright MD, Adjobo-Hermans MJW, van Spriel AB. Tetraspanin microdomains control localized protein kinase C signaling in B cells. Sci Signal 2017; 10:eaag2755. [PMID: 28487417 DOI: 10.1126/scisignal.aag2755] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Activation of B cells by the binding of antigens to the B cell receptor (BCR) requires the protein kinase C (PKC) family member PKCβ. Because PKCs must translocate to the plasma membrane to become activated, we investigated the mechanisms regulating their spatial distribution in mouse and human B cells. Through live-cell imaging, we showed that BCR-stimulated production of the second messenger diacylglycerol (DAG) resulted in the translocation of PKCβ from the cytosol to plasma membrane regions containing the tetraspanin protein CD53. CD53 was specifically enriched at sites of BCR signaling, suggesting that BCR-dependent PKC signaling was initiated at these tetraspanin microdomains. Fluorescence lifetime imaging microscopy studies confirmed the molecular recruitment of PKC to CD53-containing microdomains, which required the amino terminus of CD53. Furthermore, we showed that Cd53-deficient B cells were defective in the phosphorylation of PKC substrates. Consistent with this finding, PKC recruitment to the plasma membrane was impaired in both mouse and human CD53-deficient B cells compared to that in their wild-type counterparts. These data suggest that CD53 promotes BCR-dependent PKC signaling by recruiting PKC to the plasma membrane so that it can phosphorylate its substrates and that tetraspanin-containing microdomains can act as signaling hotspots in the plasma membrane.
Collapse
Affiliation(s)
- Malou Zuidscherwoude
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Vera-Marie E Dunlock
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Sjoerd J van Deventer
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Alie van der Schaaf
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Jenny van Oostrum
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Joachim Goedhart
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, 1098 XH Amsterdam, Netherlands
| | - Joanna In 't Hout
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, 6500 HB Nijmegen, Netherlands
| | - Günter J Hämmerling
- Department of Molecular Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Satoshi Tanaka
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Japan
| | - André Nadler
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Carsten Schultz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Mark D Wright
- Department of Immunology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia
| | - Merel J W Adjobo-Hermans
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Annemiek B van Spriel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands.
| |
Collapse
|
18
|
Palm E, Demirel I, Bengtsson T, Khalaf H. The role of toll-like and protease-activated receptors and associated intracellular signaling in Porphyromonas gingivalis-infected gingival fibroblasts. APMIS 2017; 125:157-169. [PMID: 28120492 DOI: 10.1111/apm.12645] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/04/2016] [Indexed: 11/28/2022]
Abstract
Porphyromonas gingivalis, which is considered a keystone agent in periodontitis, has evolved elaborate mechanisms to grow and survive in a hostile milieu. The gingival fibroblast is the major cell type in the gingiva and is considered to be important in the periodontitis-associated inflammation. As a part of the innate immune response, they produce cytokines such as CXCL8 and interleukin (IL)-6 which are believed to contribute to the destruction of the tooth-supporting tissues. This study investigates how the expression of protease-activated receptors (PAR1, PAR2) and toll-like receptors (TLR2, TLR4) changes with P. gingivalis exposure and how silencing of one receptor affects the expression of the other receptors. The importance of protein kinase C (PKC) and p38 in the regulation of CXCL8 and IL-6 was also examined. Receptors were knockdown with small-interfering RNA. PKC or p38 was blocked prior to stimulation with P. gingivalis. Fibroblasts were able to compensate for PAR1 knockdown with increased expression of PAR2. PKC and p38 were involved in the regulation of P. gingivalis-induced CXCL8 and IL-6. Our results indicate that PAR1 and PAR2 could be implicated in periodontitis and that PKC and P38 play a role in the inflammatory response in P. gingivalis-infected gingival fibroblasts.
Collapse
Affiliation(s)
- Eleonor Palm
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Isak Demirel
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Torbjörn Bengtsson
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Hazem Khalaf
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
19
|
Eason RJ, Bell KS, Marshall FA, Rodgers DT, Pineda MA, Steiger CN, Al-Riyami L, Harnett W, Harnett MM. The helminth product, ES-62 modulates dendritic cell responses by inducing the selective autophagolysosomal degradation of TLR-transducers, as exemplified by PKCδ. Sci Rep 2016; 6:37276. [PMID: 27869138 PMCID: PMC5116678 DOI: 10.1038/srep37276] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/27/2016] [Indexed: 12/25/2022] Open
Abstract
We have previously shown that ES-62, a phosphorylcholine (PC)-containing glycoprotein secreted by the parasitic filarial nematode Acanthocheilonema viteae targets dendritic cell (DC) responses, specifically by suppressing TLR4 signalling to inhibit Th1/Th17-driven inflammation. We have now investigated the molecular mechanisms underpinning such immunomodulation and show here that ES-62-mediated downregulation of protein kinase C-δ (PKC-δ), a TLR4-associated signalling mediator required for full activation of LPS-driven pro-inflammatory responses, is associated with induction of a low level of autophagic flux, as evidenced by upregulation and trafficking of p62 and LC3 and their consequent autophagolysosomal degradation. By contrast, the classical TLR4 ligand LPS, strongly upregulates p62 and LC3 expression but under such canonical TLR4 signalling this upregulation appears to reflect a block in autophagic flux, with these elements predominantly degraded in a proteasomal manner. These data are consistent with autophagic flux acting to homeostatically suppress proinflammatory DC responses and indeed, blocking of PKC-δ degradation by the autophagolysosomal inhibitors, E64d plus pepstatin A, results in abrogation of the ES-62-mediated suppression of LPS-driven release of IL-6, IL-12p70 and TNF-α by DCs. Thus, by harnessing this homeostatic regulatory mechanism, ES-62 can protect against aberrant inflammation, either to promote parasite survival or serendipitously, exhibit therapeutic potential in inflammatory disease.
Collapse
Affiliation(s)
- Russell J. Eason
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - Kara S. Bell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Fraser A. Marshall
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - David T. Rodgers
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - Miguel A. Pineda
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - Christina N. Steiger
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - Lamyaa Al-Riyami
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - William Harnett
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Margaret M. Harnett
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| |
Collapse
|
20
|
Liu S, Feng L, Jiang WD, Liu Y, Jiang J, Wu P, Zeng YY, Xu SD, Kuang SY, Tang L, Tang WN, Zhang YA, Zhou XQ. Impact of exogenous lipase supplementation on growth, intestinal function, mucosal immune and physical barrier, and related signaling molecules mRNA expression of young grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2016; 55:88-105. [PMID: 27164217 DOI: 10.1016/j.fsi.2016.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 04/30/2016] [Accepted: 05/04/2016] [Indexed: 06/05/2023]
Abstract
This study investigated the effects of exogenous lipase supplementation on the growth performance, intestinal growth and function, immune response and physical barrier function, and related signaling molecules mRNA expression of young grass carp (Ctenopharyngodon idella). A total of 450 grass carp (255.02 ± 0.34 g) were fed five diets for 60 days. There were 5 dietary treatments that included a normal protein and lipid diet containing 30% crude protein (CP) with 5% ether extract (EE), and the low-protein and high-lipid diets (28% CP, 6% EE) supplemented with graded levels of exogenous lipase supplementation activity at 0, 1193, 2560 and 3730 U/kg diet. The results indicated that compared with a normal protein and lipid diet (30% CP, 5% EE), a low-protein and high-lipid diet (28% CP, 6% EE) (un-supplemented lipase) improved lysozyme activities and complement component 3 contents in the distal intestine (DI), interleukin 10 mRNA expression in the proximal intestine (PI), and glutathione S-transferases activity and glutathione content in the intestine of young grass carp. In addition, in low-protein and high-lipid diets, optimal exogenous lipase supplementation significantly increased acid phosphatase (ACP) activities and complement component 3 (C3) contents (P < 0.05), up-regulated the relative mRNA levels of antimicrobial peptides (liver expressed antimicrobial peptide 2 and hepcidin) and anti-inflammatory cytokines (interleukin 10 and transforming growth factor β1) and signaling molecules inhibitor protein-κBα (IκBα) and target of rapamycin (TOR) (P < 0.05), down-regulated the mRNA levels of pro-inflammatory cytokines (tumor necrosis factor α, interleukin 8, interferon γ2, and interleukin 1β), and signaling molecules (nuclear factor kappa B p65, IκB kinase β, IκB kinase γ) (P < 0.05) in the intestine of young grass carp. Moreover, optimal exogenous lipase supplementation significantly decreased reactive oxygen species (ROS), malondialdehyde (MDA) and protein carbonyl (PC) contents (P < 0.05), improved the activities of anti-superoxide anion (ASA) and anti-hydroxyl radical (AHR), glutathione content, and the activities and mRNA levels of antioxidant enzymes (copper/zinc superoxide dismutase, manganese superoxide dismutase, catalase, glutathione peroxidase, glutathione S-transferases and glutathione reductase) (P < 0.05), up-regulated signaling molecule NF-E2-related factor 2 (Nrf2) (P < 0.05), down-regulated signaling molecules (Kelch-like-ECH-associated protein 1a, Kelch-like-ECH-associated protein 1b) (P < 0.05) in the intestine of young grass carp. Furthermore, optimal exogenous lipase supplementation significantly elevated the mRNA levels of tight junction proteins (Occludin, zonula occludens 1, Claudin b, Claudin c and Claudin 3) (P < 0.05), down-regulated the mRNA levels of tight junction proteins (Claudin 12 and Claudin 15a) (P < 0.05), down-regulated signaling molecules myosin light chain kinase (P < 0.05) in the intestine of young grass carp. In conclusion, dietary lipid could partially spare protein, and the low-protein and high-lipid diet could improve growth, intestinal growth and function, immune response and antioxidant capability of fish. Meanwhile, in high-fat and low-protein diets, optimal exogenous lipase supplementation improved growth, intestinal growth and function, intestinal immunity, physical barrier, and regulated the mRNA expression of related signal molecules of fish. The optimal level of exogenous lipase supplementation in young grass carp (255-771 g) was estimated to be 1193 U kg(-1) diet.
Collapse
Affiliation(s)
- Sen Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yun-Yun Zeng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Shu-De Xu
- Guangdong Vtr Bio-tech Co., Ltd., Zhuhai 519060, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Wu-Neng Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
21
|
Choi JY, Jeon SJ, Son KH, Park YI, Dong MS. Induction of mast cell degranulation by triterpenoidal saponins obtained from Cimicifugae rhizoma. Immunopharmacol Immunotoxicol 2016; 38:311-8. [PMID: 27310149 DOI: 10.1080/08923973.2016.1201101] [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: 10/21/2022]
Abstract
Cimicifugae rhizoma has been widely used as a traditional herbal medicine to treat inflammation and menopausal symptoms. In this study, we found that some of the triterpenoidal saponins purified from the ethanol extract of Cimicifugae rhizoma dramatically induced histamine release. The structure-related induction of mast cell degranulation by them and the mechanism of action were determined. β-Hexosaminidase release in HMC-1 cells was increased in a concentration-dependent manner, with maximal 6.5- and 8.5-fold increases, by 200 μg/mL 24-epi-7,8-didehydrocimigenol-3-O-xyloside (comp 1) and cimigenol 3-O-beta-d-xyloside (comp 4) compared with those treated with phorbol 12-myristate 13-acetate and A23187 (PMACI), respectively. However, β-hexosaminidase release was not changed by 7,8-dihydrocimigenol (comp 3), or 23-OAc-shengmanol-3-O-xyloside (comp 7). These triterpenoidal saponins changed neither the intracellular Ca(2+ )level nor the activation of PKC, both of which play essential roles in histamine release. However, cromolyn and ketotifen, membrane stabilizers, effectively inhibited the β-hexosaminidase release induced by comp 1 or comp 4 by 39 and 45%, respectively. Collectively, xylose on the cimigenol-related backbone among triterpene glycosides isolated from Cimicifugae rhizoma may play an important role in activating mast cells and induction of degranulation partly via membrane destabilization of mast cells.
Collapse
Affiliation(s)
- Ji-Yoon Choi
- a School of Life Sciences and Biotechnology, Korea University , Seoul , Republic of Korea
| | - Su Jin Jeon
- b Department of Food and Nutrition , Andong National University , Andong , Republic of Korea
| | - Kun Ho Son
- b Department of Food and Nutrition , Andong National University , Andong , Republic of Korea
| | - Young In Park
- c College of Pharmacy, Korea University , Sejong , Republic of Korea
| | - Mi-Sook Dong
- a School of Life Sciences and Biotechnology, Korea University , Seoul , Republic of Korea
| |
Collapse
|
22
|
Najib A, Kim MS, Choi SH, Kang YJ, Kim KH. Changes in microRNAs expression profile of olive flounder (Paralichthys olivaceus) in response to viral hemorrhagic septicemia virus (VHSV) infection. FISH & SHELLFISH IMMUNOLOGY 2016; 51:384-391. [PMID: 26975411 DOI: 10.1016/j.fsi.2016.03.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 03/03/2016] [Accepted: 03/10/2016] [Indexed: 06/05/2023]
Abstract
To know the effect of viral hemorrhagic septicemia virus (VHSV) infection on the cellular microRNA expression profile in olive flounder (Paralichthys olivaceus), fish were infected with VHSV, and cellular microRNAs expression was analyzed at 0 (control), 6, 12, 24, 48 and 72 h post-infection (h.p.i.) by the high-throughput sequencing. A total of 372 mature miRNAs were identified, and, among them, 63 miRNAs were differentially expressed during VHSV infection. The differentially expressed microRNAs number was greatly increased from 24 h.p.i. compared to the number at 6 and 12 h.p.i., suggesting that the alteration of microRNAs expression by VHSV infection may be related to the progression of VHSV disease. The target prediction analysis, the GO enrichment analysis, and the KEGG pathway analysis of the predicted target genes showed that various biological pathways could be affected by VHSV infection through the down-regulation or up-regulation of host miRNAs. The present results provide a basic information on the microRNAs related to VHSV infection in olive flounder. Considering broad effects of microRNAs on various biological pathways, data in this study can be used to interpret the mechanism of VHSV pathogenesis, which, vice versa, can be used to develop control measures against VHSV.
Collapse
Affiliation(s)
- Abdellaoui Najib
- Department of Aquatic Life Medicine, Pukyong National University, Busan, 608-737, South Korea
| | - Min Sun Kim
- Department of Aquatic Life Medicine, Pukyong National University, Busan, 608-737, South Korea
| | - Seung Hyuk Choi
- Department of Aquatic Life Medicine, Pukyong National University, Busan, 608-737, South Korea
| | - Yue Jai Kang
- Department of Aquatic Life and Medical Sciences, Sun Moon University, Asan-si, Chungnam, 336-708, South Korea
| | - Ki Hong Kim
- Department of Aquatic Life Medicine, Pukyong National University, Busan, 608-737, South Korea.
| |
Collapse
|
23
|
Abstract
Epstein-Barr virus (EBV) infection is a common feature of B cell lymphoproliferative disorders (LPDs), including diffuse large B cell lymphoma. Approximately 10 % of DLBCLs are EBV-positive, with the highest incidence in immunocompromised and elderly patients. Here, we review the clinical, genetic, and pathologic characteristics of DLBCL and discuss the molecular role of EBV in lymphoma tumorigenesis. Using EBV-positive DLBCL of the elderly as a model, we describe the key features of EBV-positive DLBCL. Studies of EBV-positive DLBCL of the elderly demonstrate that EBV-positive DLBCL has a distinct biology, related to both viral and host factors. The pathogenic mechanisms noted in EBV-positive DLBCL of the elderly, including enhanced NFκB activity, are likely to be a generalizable feature of EBV-positive DLBCL. Therefore, we review how this information might be used to target the EBV or its host response for the development of novel treatment strategies.
Collapse
|
24
|
Global Mapping of Traditional Chinese Medicine into Bioactivity Space and Pathways Annotation Improves Mechanistic Understanding and Discovers Relationships between Therapeutic Action (Sub)classes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:2106465. [PMID: 26989424 PMCID: PMC4775820 DOI: 10.1155/2016/2106465] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/03/2015] [Indexed: 02/08/2023]
Abstract
Traditional Chinese medicine (TCM) still needs more scientific rationale to be proven for it to be accepted further in the West. We are now in the position to propose computational hypotheses for the mode-of-actions (MOAs) of 45 TCM therapeutic action (sub)classes from in silico target prediction algorithms, whose target was later annotated with Kyoto Encyclopedia of Genes and Genomes pathway, and to discover the relationship between them by generating a hierarchical clustering. The results of 10,749 TCM compounds showed 183 enriched targets and 99 enriched pathways from Estimation Score ≤ 0 and ≥ 5% of compounds/targets in a (sub)class. The MOA of a (sub)class was established from supporting literature. Overall, the most frequent top three enriched targets/pathways were immune-related targets such as tyrosine-protein phosphatase nonreceptor type 2 (PTPN2) and digestive system such as mineral absorption. We found two major protein families, G-protein coupled receptor (GPCR), and protein kinase family contributed to the diversity of the bioactivity space, while digestive system was consistently annotated pathway motif, which agreed with the important treatment principle of TCM, “the foundation of acquired constitution” that includes spleen and stomach. In short, the TCM (sub)classes, in many cases share similar targets/pathways despite having different indications.
Collapse
|
25
|
Tebay LE, Robertson H, Durant ST, Vitale SR, Penning TM, Dinkova-Kostova AT, Hayes JD. Mechanisms of activation of the transcription factor Nrf2 by redox stressors, nutrient cues, and energy status and the pathways through which it attenuates degenerative disease. Free Radic Biol Med 2015; 88:108-146. [PMID: 26122708 PMCID: PMC4659505 DOI: 10.1016/j.freeradbiomed.2015.06.021] [Citation(s) in RCA: 645] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) regulates the basal and stress-inducible expression of a battery of genes encoding key components of the glutathione-based and thioredoxin-based antioxidant systems, as well as aldo-keto reductase, glutathione S-transferase, and NAD(P)H quinone oxidoreductase-1 drug-metabolizing isoenzymes along with multidrug-resistance-associated efflux pumps. It therefore plays a pivotal role in both intrinsic resistance and cellular adaptation to reactive oxygen species (ROS) and xenobiotics. Activation of Nrf2 can, however, serve as a double-edged sword because some of the genes it induces may contribute to chemical carcinogenesis by promoting futile redox cycling of polycyclic aromatic hydrocarbon metabolites or confer resistance to chemotherapeutic drugs by increasing the expression of efflux pumps, suggesting its cytoprotective effects will vary in a context-specific fashion. In addition to cytoprotection, Nrf2 also controls genes involved in intermediary metabolism, positively regulating those involved in NADPH generation, purine biosynthesis, and the β-oxidation of fatty acids, while suppressing those involved in lipogenesis and gluconeogenesis. Nrf2 is subject to regulation at multiple levels. Its ability to orchestrate adaptation to oxidants and electrophiles is due principally to stress-stimulated modification of thiols within one of its repressors, the Kelch-like ECH-associated protein 1 (Keap1), which is present in the cullin-3 RING ubiquitin ligase (CRL) complex CRLKeap1. Thus modification of Cys residues in Keap1 blocks CRLKeap1 activity, allowing newly translated Nrf2 to accumulate rapidly and induce its target genes. The ability of Keap1 to repress Nrf2 can be attenuated by p62/sequestosome-1 in a mechanistic target of rapamycin complex 1 (mTORC1)-dependent manner, thereby allowing refeeding after fasting to increase Nrf2-target gene expression. In parallel with repression by Keap1, Nrf2 is also repressed by β-transducin repeat-containing protein (β-TrCP), present in the Skp1-cullin-1-F-box protein (SCF) ubiquitin ligase complex SCFβ-TrCP. The ability of SCFβ-TrCP to suppress Nrf2 activity is itself enhanced by prior phosphorylation of the transcription factor by glycogen synthase kinase-3 (GSK-3) through formation of a DSGIS-containing phosphodegron. However, formation of the phosphodegron in Nrf2 by GSK-3 is inhibited by stimuli that activate protein kinase B (PKB)/Akt. In particular, PKB/Akt activity can be increased by phosphoinositide 3-kinase and mTORC2, thereby providing an explanation of why antioxidant-responsive element-driven genes are induced by growth factors and nutrients. Thus Nrf2 activity is tightly controlled via CRLKeap1 and SCFβ-TrCP by oxidative stress and energy-based signals, allowing it to mediate adaptive responses that restore redox homeostasis and modulate intermediary metabolism. Based on the fact that Nrf2 influences multiple biochemical pathways in both positive and negative ways, it is likely its dose-response curve, in terms of susceptibility to certain degenerative disease, is U-shaped. Specifically, too little Nrf2 activity will lead to loss of cytoprotection, diminished antioxidant capacity, and lowered β-oxidation of fatty acids, while conversely also exhibiting heightened sensitivity to ROS-based signaling that involves receptor tyrosine kinases and apoptosis signal-regulating kinase-1. By contrast, too much Nrf2 activity disturbs the homeostatic balance in favor of reduction, and so may have deleterious consequences including overproduction of reduced glutathione and NADPH, the blunting of ROS-based signal transduction, epithelial cell hyperplasia, and failure of certain cell types to differentiate correctly. We discuss the basis of a putative U-shaped Nrf2 dose-response curve in terms of potentially competing processes relevant to different stages of tumorigenesis.
Collapse
Affiliation(s)
- Lauren E Tebay
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Holly Robertson
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Stephen T Durant
- AstraZeneca Oncology Innovative Medicines, Bioscience, 33F197 Mereside, Alderley Park, Cheshire SK10 4TG, UK
| | - Steven R Vitale
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6160, USA
| | - Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6160, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK.
| |
Collapse
|
26
|
Mohamed W, Ray S, Brazill D, Baskar R. Absence of catalytic domain in a putative protein kinase C (PkcA) suppresses tip dominance in Dictyostelium discoideum. Dev Biol 2015; 405:10-20. [PMID: 26183108 DOI: 10.1016/j.ydbio.2015.05.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 04/06/2015] [Accepted: 05/28/2015] [Indexed: 12/22/2022]
Abstract
A number of organisms possess several isoforms of protein kinase C but little is known about the significance of any specific isoform during embryogenesis and development. To address this we characterized a PKC ortholog (PkcA; DDB_G0288147) in Dictyostelium discoideum. pkcA expression switches from prestalk in mound to prespore in slug, indicating a dynamic expression pattern. Mutants lacking the catalytic domain of PkcA (pkcA(-)) did not exhibit tip dominance. A striking phenotype of pkcA- was the formation of an aggregate with a central hollow, and aggregates later fragmented to form small mounds, each becoming a fruiting body. Optical density wave patterns of cAMP in the late aggregates showed several cAMP wave generation centers. We attribute these defects in pkcA(-) to impaired cAMP signaling, altered cell motility and decreased expression of the cell adhesion molecules - CadA and CsaA. pkcA(-) slugs showed ectopic expression of ecmA in the prespore region. Further, the use of a PKC-specific inhibitor, GF109203X that inhibits the activity of catalytic domain phenocopied pkcA(-).
Collapse
Affiliation(s)
- Wasima Mohamed
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Sibnath Ray
- Department of Biological Sciences, Center for Translational and Basic Research, Hunter College and The Graduate Center of the City University of New York, New York, NY 10065, USA
| | - Derrick Brazill
- Department of Biological Sciences, Center for Translational and Basic Research, Hunter College and The Graduate Center of the City University of New York, New York, NY 10065, USA
| | - Ramamurthy Baskar
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India.
| |
Collapse
|
27
|
López-Rodríguez C, Aramburu J, Berga-Bolaños R. Transcription factors and target genes of pre-TCR signaling. Cell Mol Life Sci 2015; 72:2305-21. [PMID: 25702312 PMCID: PMC11113633 DOI: 10.1007/s00018-015-1864-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/22/2015] [Accepted: 02/16/2015] [Indexed: 11/27/2022]
Abstract
Almost 30 years ago pioneering work by the laboratories of Harald von Boehmer and Susumo Tonegawa provided the first indications that developing thymocytes could assemble a functional TCRβ chain-containing receptor complex, the pre-TCR, before TCRα expression. The discovery and study of the pre-TCR complex revealed paradigms of signaling pathways in control of cell survival and proliferation, and culminated in the recognition of the multifunctional nature of this receptor. As a receptor integrated in a dynamic developmental process, the pre-TCR must be viewed not only in the light of the biological outcomes it promotes, but also in context with those molecular processes that drive its expression in thymocytes. This review article focuses on transcription factors and target genes activated by the pre-TCR to drive its different outcomes.
Collapse
Affiliation(s)
- Cristina López-Rodríguez
- Immunology Unit, Department of Experimental and Health Sciences and Barcelona Biomedical Research Park, Universitat Pompeu Fabra, C/Doctor Aiguader Nº88, 08003, Barcelona, Barcelona, Spain,
| | | | | |
Collapse
|
28
|
Abstract
Chronic inflammatory diseases, such as asthma and chronic obstructive pulmonary disease (COPD), are clinically and socioeconomically important diseases globally. Currently the mainstay of anti-inflammatory therapy in respiratory diseases is corticosteroids. Although corticosteroids have proven clinical efficacy in asthma, many asthmatic inflammatory conditions (e.g., infection, exacerbation, and severe asthma) are not responsive to corticosteroids. Moreover, despite an understanding that COPD progression is driven by inflammation, we currently do not have effective anti-inflammatory strategies to combat this disease. Hence, alternative anti-inflammatory strategies are required. p38 mitogen-activated protein kinase (MAPK) has emerged as an important signaling molecule driving airway inflammation, and pharmacological inhibitors against p38 MAPK may provide potential therapies for chronic respiratory disease. In this review, we discuss some of the recent in vitro and in vivo studies targeting p38 MAPK, but suggest that p38 MAPK inhibitors may prove less effective than originally considered because they may block anti-inflammatory molecules along with proinflammatory responses. We propose that an alternative strategy may be to target an anti-inflammatory molecule farther downstream of p38 MAPK, i.e., tristetraprolin (TTP). TTP is an mRNA-destabilizing, RNA-binding protein that enhances the decay of mRNAs, including those encoding proteins implicated in chronic respiratory diseases. We suggest that understanding the molecular mechanism of TTP expression and its temporal regulation will guide future development of novel anti-inflammatory pharmacotherapeutic approaches to combat respiratory disease.
Collapse
Affiliation(s)
- Pavan Prabhala
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
| | - Alaina J Ammit
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
29
|
Durpès MC, Morin C, Paquin-Veillet J, Beland R, Paré M, Guimond MO, Rekhter M, King GL, Geraldes P. PKC-β activation inhibits IL-18-binding protein causing endothelial dysfunction and diabetic atherosclerosis. Cardiovasc Res 2015; 106:303-13. [PMID: 25808972 DOI: 10.1093/cvr/cvv107] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 03/06/2015] [Indexed: 12/19/2022] Open
Abstract
AIMS Clinical observations showed a correlation between accelerated atherosclerosis in diabetes and high plasmatic level of IL-18, a pro-inflammatory cytokine. IL-18 enhances the production of inflammatory cytokines and cellular adhesion molecules contributing to atherosclerotic plaque formation and instability. Previous studies indicated that protein kinase C (PKC)-β inhibition prevented macrophage-induced cytokine expression involved in diabetic (DM) atherosclerotic plaque development. However, the role of PKC-β activation on IL-18/IL-18-binding protein (IL-18BP) pathway causing endothelial dysfunction and monocyte adhesion in diabetes has never been explored. METHODS AND RESULTS Apoe(-/-) mice were rendered DM and fed with western diet containing ruboxistaurin (RBX), a PKC-β inhibitor. After 20 weeks, atherosclerotic plaque composition was quantified. Compared with non-diabetic, DM mice exhibited elevated atherosclerotic plaque formation, cholestoryl ester content and macrophage infiltration, as well as reduced IL-18BP expression in the aorta which was prevented with RBX treatment. Endothelial cells (ECs) and macrophages were exposed to normal or high glucose (HG) levels with or without palmitate and recombinant IL-18 for 24 h. The combined HG and palmitate condition was required to increase IL-18 expression and secretion in macrophages, while it reduced IL-18BP expression in EC causing up-regulation of the vascular cell adhesion molecule (VCAM)-1 and monocyte adhesion. Elevated VCAM-1 expression and monocyte adherence were prevented by siRNA, RBX, and IL-18 neutralizing antibody. CONCLUSION Our study unrevealed a new mechanism by which PKC-β activation promotes EC dysfunction caused by the de-regulation of the IL-18/IL-18BP pathway, leading to increased VCAM-1 expression, monocyte/macrophage adhesion, and accelerated atherosclerotic plaque formation in diabetes.
Collapse
Affiliation(s)
- Marie-Claude Durpès
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, 3001 12e Avenue Nord, QC, Canada J1H 5N4
| | - Catherine Morin
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, 3001 12e Avenue Nord, QC, Canada J1H 5N4
| | - Judith Paquin-Veillet
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, 3001 12e Avenue Nord, QC, Canada J1H 5N4
| | - Raphaël Beland
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, 3001 12e Avenue Nord, QC, Canada J1H 5N4
| | - Martin Paré
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, 3001 12e Avenue Nord, QC, Canada J1H 5N4
| | - Marie-Odile Guimond
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, 3001 12e Avenue Nord, QC, Canada J1H 5N4
| | - Mark Rekhter
- Cardiometabolic Diseases and Complications of Diabetes, Lilly Research Laboratories, Indianapolis, IN, USA
| | - George L King
- Research Division, Joslin Diabetes Center, Boston, MA, USA
| | - Pedro Geraldes
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, 3001 12e Avenue Nord, QC, Canada J1H 5N4
| |
Collapse
|
30
|
Fukahori H, Chida N, Maeda M, Tasaki M, Kawashima T, Matsuoka H, Suzuki K, Ishikawa T, Tanaka A, Higashi Y. Effect of AS2521780, a novel PKCθ selective inhibitor, on T cell-mediated immunity. Eur J Pharmacol 2014; 745:217-22. [DOI: 10.1016/j.ejphar.2014.10.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 10/23/2014] [Accepted: 10/28/2014] [Indexed: 01/29/2023]
|
31
|
Neafsey DE, Waterhouse RM, Abai MR, Aganezov SS, Alekseyev MA, Allen JE, Amon J, Arcà B, Arensburger P, Artemov G, Assour LA, Basseri H, Berlin A, Birren BW, Blandin SA, Brockman AI, Burkot TR, Burt A, Chan CS, Chauve C, Chiu JC, Christensen M, Costantini C, Davidson VLM, Deligianni E, Dottorini T, Dritsou V, Gabriel SB, Guelbeogo WM, Hall AB, Han MV, Hlaing T, Hughes DST, Jenkins AM, Jiang X, Jungreis I, Kakani EG, Kamali M, Kemppainen P, Kennedy RC, Kirmitzoglou IK, Koekemoer LL, Laban N, Langridge N, Lawniczak MKN, Lirakis M, Lobo NF, Lowy E, MacCallum RM, Mao C, Maslen G, Mbogo C, McCarthy J, Michel K, Mitchell SN, Moore W, Murphy KA, Naumenko AN, Nolan T, Novoa EM, O'Loughlin S, Oringanje C, Oshaghi MA, Pakpour N, Papathanos PA, Peery AN, Povelones M, Prakash A, Price DP, Rajaraman A, Reimer LJ, Rinker DC, Rokas A, Russell TL, Sagnon N, Sharakhova MV, Shea T, Simão FA, Simard F, Slotman MA, Somboon P, Stegniy V, Struchiner CJ, Thomas GWC, Tojo M, Topalis P, Tubio JMC, Unger MF, Vontas J, Walton C, Wilding CS, Willis JH, Wu YC, Yan G, Zdobnov EM, Zhou X, Catteruccia F, Christophides GK, Collins FH, Cornman RS, et alNeafsey DE, Waterhouse RM, Abai MR, Aganezov SS, Alekseyev MA, Allen JE, Amon J, Arcà B, Arensburger P, Artemov G, Assour LA, Basseri H, Berlin A, Birren BW, Blandin SA, Brockman AI, Burkot TR, Burt A, Chan CS, Chauve C, Chiu JC, Christensen M, Costantini C, Davidson VLM, Deligianni E, Dottorini T, Dritsou V, Gabriel SB, Guelbeogo WM, Hall AB, Han MV, Hlaing T, Hughes DST, Jenkins AM, Jiang X, Jungreis I, Kakani EG, Kamali M, Kemppainen P, Kennedy RC, Kirmitzoglou IK, Koekemoer LL, Laban N, Langridge N, Lawniczak MKN, Lirakis M, Lobo NF, Lowy E, MacCallum RM, Mao C, Maslen G, Mbogo C, McCarthy J, Michel K, Mitchell SN, Moore W, Murphy KA, Naumenko AN, Nolan T, Novoa EM, O'Loughlin S, Oringanje C, Oshaghi MA, Pakpour N, Papathanos PA, Peery AN, Povelones M, Prakash A, Price DP, Rajaraman A, Reimer LJ, Rinker DC, Rokas A, Russell TL, Sagnon N, Sharakhova MV, Shea T, Simão FA, Simard F, Slotman MA, Somboon P, Stegniy V, Struchiner CJ, Thomas GWC, Tojo M, Topalis P, Tubio JMC, Unger MF, Vontas J, Walton C, Wilding CS, Willis JH, Wu YC, Yan G, Zdobnov EM, Zhou X, Catteruccia F, Christophides GK, Collins FH, Cornman RS, Crisanti A, Donnelly MJ, Emrich SJ, Fontaine MC, Gelbart W, Hahn MW, Hansen IA, Howell PI, Kafatos FC, Kellis M, Lawson D, Louis C, Luckhart S, Muskavitch MAT, Ribeiro JM, Riehle MA, Sharakhov IV, Tu Z, Zwiebel LJ, Besansky NJ. Mosquito genomics. Highly evolvable malaria vectors: the genomes of 16 Anopheles mosquitoes. Science 2014; 347:1258522. [PMID: 25554792 DOI: 10.1126/science.1258522] [Show More Authors] [Citation(s) in RCA: 391] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Variation in vectorial capacity for human malaria among Anopheles mosquito species is determined by many factors, including behavior, immunity, and life history. To investigate the genomic basis of vectorial capacity and explore new avenues for vector control, we sequenced the genomes of 16 anopheline mosquito species from diverse locations spanning ~100 million years of evolution. Comparative analyses show faster rates of gene gain and loss, elevated gene shuffling on the X chromosome, and more intron losses, relative to Drosophila. Some determinants of vectorial capacity, such as chemosensory genes, do not show elevated turnover but instead diversify through protein-sequence changes. This dynamism of anopheline genes and genomes may contribute to their flexible capacity to take advantage of new ecological niches, including adapting to humans as primary hosts.
Collapse
Affiliation(s)
- Daniel E Neafsey
- Genome Sequencing and Analysis Program, Broad Institute, 415 Main Street, Cambridge, MA 02142, USA.
| | - Robert M Waterhouse
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, 32 Vassar Street, Cambridge, MA 02139, USA. The Broad Institute of Massachusetts Institute of Technology and Harvard, 415 Main Street, Cambridge, MA 02142, USA. Department of Genetic Medicine and Development, University of Geneva Medical School, Rue Michel-Servet 1, 1211 Geneva, Switzerland. Swiss Institute of Bioinformatics, Rue Michel-Servet 1, 1211 Geneva, Switzerland
| | - Mohammad R Abai
- Department of Medical Entomology and Vector Control, School of Public Health and Institute of Health Researches, Tehran University of Medical Sciences, Tehran, Iran
| | - Sergey S Aganezov
- George Washington University, Department of Mathematics and Computational Biology Institute, 45085 University Drive, Ashburn, VA 20147, USA
| | - Max A Alekseyev
- George Washington University, Department of Mathematics and Computational Biology Institute, 45085 University Drive, Ashburn, VA 20147, USA
| | - James E Allen
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - James Amon
- National Vector Borne Disease Control Programme, Ministry of Health, Tafea Province, Vanuatu
| | - Bruno Arcà
- Department of Public Health and Infectious Diseases, Division of Parasitology, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Peter Arensburger
- Department of Biological Sciences, California State Polytechnic-Pomona, 3801 West Temple Avenue, Pomona, CA 91768, USA
| | - Gleb Artemov
- Tomsk State University, 36 Lenina Avenue, Tomsk, Russia
| | - Lauren A Assour
- Department of Computer Science and Engineering, Eck Institute for Global Health, 211B Cushing Hall, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Hamidreza Basseri
- Department of Medical Entomology and Vector Control, School of Public Health and Institute of Health Researches, Tehran University of Medical Sciences, Tehran, Iran
| | - Aaron Berlin
- Genome Sequencing and Analysis Program, Broad Institute, 415 Main Street, Cambridge, MA 02142, USA
| | - Bruce W Birren
- Genome Sequencing and Analysis Program, Broad Institute, 415 Main Street, Cambridge, MA 02142, USA
| | - Stephanie A Blandin
- Inserm, U963, F-67084 Strasbourg, France. CNRS, UPR9022, IBMC, F-67084 Strasbourg, France
| | - Andrew I Brockman
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Thomas R Burkot
- Faculty of Medicine, Health and Molecular Science, Australian Institute of Tropical Health Medicine, James Cook University, Cairns 4870, Australia
| | - Austin Burt
- Department of Life Sciences, Imperial College London, Silwood Park Campus, Ascot SL5 7PY, UK
| | - Clara S Chan
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, 32 Vassar Street, Cambridge, MA 02139, USA. The Broad Institute of Massachusetts Institute of Technology and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Cedric Chauve
- Department of Mathematics, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Joanna C Chiu
- Department of Entomology and Nematology, One Shields Avenue, University of California-Davis, Davis, CA 95616, USA
| | - Mikkel Christensen
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Carlo Costantini
- Institut de Recherche pour le Développement, Unités Mixtes de Recherche Maladies Infectieuses et Vecteurs Écologie, Génétique, Évolution et Contrôle, 911, Avenue Agropolis, BP 64501 Montpellier, France
| | - Victoria L M Davidson
- Division of Biology, Kansas State University, 271 Chalmers Hall, Manhattan, KS 66506, USA
| | - Elena Deligianni
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Hellas, Nikolaou Plastira 100 GR-70013, Heraklion, Crete, Greece
| | - Tania Dottorini
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Vicky Dritsou
- Centre of Functional Genomics, University of Perugia, Perugia, Italy
| | - Stacey B Gabriel
- Genomics Platform, Broad Institute, 415 Main Street, Cambridge, MA 02142, USA
| | - Wamdaogo M Guelbeogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou 01 BP 2208, Burkina Faso
| | - Andrew B Hall
- Program of Genetics, Bioinformatics, and Computational Biology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Mira V Han
- School of Life Sciences, University of Nevada, Las Vegas, NV 89154, USA
| | - Thaung Hlaing
- Department of Medical Research, No. 5 Ziwaka Road, Dagon Township, Yangon 11191, Myanmar
| | - Daniel S T Hughes
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK. Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Adam M Jenkins
- Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA
| | - Xiaofang Jiang
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA. Program of Genetics, Bioinformatics, and Computational Biology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Irwin Jungreis
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, 32 Vassar Street, Cambridge, MA 02139, USA. The Broad Institute of Massachusetts Institute of Technology and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Evdoxia G Kakani
- Harvard School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA 02115, USA. Dipartimento di Medicina Sperimentale e Scienze Biochimiche, Università degli Studi di Perugia, Perugia, Italy
| | - Maryam Kamali
- Department of Entomology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Petri Kemppainen
- Computational Evolutionary Biology Group, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Ryan C Kennedy
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143, USA
| | - Ioannis K Kirmitzoglou
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK. Bioinformatics Research Laboratory, Department of Biological Sciences, New Campus, University of Cyprus, CY 1678 Nicosia, Cyprus
| | - Lizette L Koekemoer
- Wits Research Institute for Malaria, Faculty of Health Sciences, and Vector Control Reference Unit, National Institute for Communicable Diseases of the National Health Laboratory Service, Sandringham 2131, Johannesburg, South Africa
| | - Njoroge Laban
- National Museums of Kenya, P.O. Box 40658-00100, Nairobi, Kenya
| | - Nicholas Langridge
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Mara K N Lawniczak
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Manolis Lirakis
- Department of Biology, University of Crete, 700 13 Heraklion, Greece
| | - Neil F Lobo
- Eck Institute for Global Health and Department of Biological Sciences, University of Notre Dame, 317 Galvin Life Sciences Building, Notre Dame, IN 46556, USA
| | - Ernesto Lowy
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Robert M MacCallum
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Chunhong Mao
- Virginia Bioinformatics Institute, 1015 Life Science Circle, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Gareth Maslen
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Charles Mbogo
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research - Coast, P.O. Box 230-80108, Kilifi, Kenya
| | - Jenny McCarthy
- Department of Biological Sciences, California State Polytechnic-Pomona, 3801 West Temple Avenue, Pomona, CA 91768, USA
| | - Kristin Michel
- Division of Biology, Kansas State University, 271 Chalmers Hall, Manhattan, KS 66506, USA
| | - Sara N Mitchell
- Harvard School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA 02115, USA
| | - Wendy Moore
- Department of Entomology, 1140 East South Campus Drive, Forbes 410, University of Arizona, Tucson, AZ 85721, USA
| | - Katherine A Murphy
- Department of Entomology and Nematology, One Shields Avenue, University of California-Davis, Davis, CA 95616, USA
| | - Anastasia N Naumenko
- Department of Entomology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Tony Nolan
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Eva M Novoa
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, 32 Vassar Street, Cambridge, MA 02139, USA. The Broad Institute of Massachusetts Institute of Technology and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Samantha O'Loughlin
- Department of Life Sciences, Imperial College London, Silwood Park Campus, Ascot SL5 7PY, UK
| | - Chioma Oringanje
- Department of Entomology, 1140 East South Campus Drive, Forbes 410, University of Arizona, Tucson, AZ 85721, USA
| | - Mohammad A Oshaghi
- Department of Medical Entomology and Vector Control, School of Public Health and Institute of Health Researches, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazzy Pakpour
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Philippos A Papathanos
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK. Centre of Functional Genomics, University of Perugia, Perugia, Italy
| | - Ashley N Peery
- Department of Entomology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Michael Povelones
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Anil Prakash
- Regional Medical Research Centre NE, Indian Council of Medical Research, P.O. Box 105, Dibrugarh-786 001, Assam, India
| | - David P Price
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, USA. Molecular Biology Program, New Mexico State University, Las Cruces, NM 88003, USA
| | - Ashok Rajaraman
- Department of Mathematics, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Lisa J Reimer
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - David C Rinker
- Center for Human Genetics Research, Vanderbilt University Medical Center, Nashville, TN 37235, USA
| | - Antonis Rokas
- Center for Human Genetics Research, Vanderbilt University Medical Center, Nashville, TN 37235, USA. Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| | - Tanya L Russell
- Faculty of Medicine, Health and Molecular Science, Australian Institute of Tropical Health Medicine, James Cook University, Cairns 4870, Australia
| | - N'Fale Sagnon
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou 01 BP 2208, Burkina Faso
| | - Maria V Sharakhova
- Department of Entomology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Terrance Shea
- Genome Sequencing and Analysis Program, Broad Institute, 415 Main Street, Cambridge, MA 02142, USA
| | - Felipe A Simão
- Department of Genetic Medicine and Development, University of Geneva Medical School, Rue Michel-Servet 1, 1211 Geneva, Switzerland. Swiss Institute of Bioinformatics, Rue Michel-Servet 1, 1211 Geneva, Switzerland
| | - Frederic Simard
- Institut de Recherche pour le Développement, Unités Mixtes de Recherche Maladies Infectieuses et Vecteurs Écologie, Génétique, Évolution et Contrôle, 911, Avenue Agropolis, BP 64501 Montpellier, France
| | - Michel A Slotman
- Department of Entomology, Texas A&M University, College Station, TX 77807, USA
| | - Pradya Somboon
- Department of Parasitology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Claudio J Struchiner
- Fundação Oswaldo Cruz, Avenida Brasil 4365, RJ Brazil. Instituto de Medicina Social, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gregg W C Thomas
- School of Informatics and Computing, Indiana University, Bloomington, IN 47405, USA
| | - Marta Tojo
- Department of Physiology, School of Medicine, Center for Research in Molecular Medicine and Chronic Diseases, Instituto de Investigaciones Sanitarias, University of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
| | - Pantelis Topalis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Hellas, Nikolaou Plastira 100 GR-70013, Heraklion, Crete, Greece
| | - José M C Tubio
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Maria F Unger
- Eck Institute for Global Health and Department of Biological Sciences, University of Notre Dame, 317 Galvin Life Sciences Building, Notre Dame, IN 46556, USA
| | - John Vontas
- Department of Biology, University of Crete, 700 13 Heraklion, Greece
| | - Catherine Walton
- Computational Evolutionary Biology Group, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Craig S Wilding
- School of Natural Sciences and Psychology, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - Judith H Willis
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Yi-Chieh Wu
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, 32 Vassar Street, Cambridge, MA 02139, USA. The Broad Institute of Massachusetts Institute of Technology and Harvard, 415 Main Street, Cambridge, MA 02142, USA. Department of Computer Science, Harvey Mudd College, Claremont, CA 91711, USA
| | - Guiyun Yan
- Program in Public Health, College of Health Sciences, University of California, Irvine, Hewitt Hall, Irvine, CA 92697, USA
| | - Evgeny M Zdobnov
- Department of Genetic Medicine and Development, University of Geneva Medical School, Rue Michel-Servet 1, 1211 Geneva, Switzerland. Swiss Institute of Bioinformatics, Rue Michel-Servet 1, 1211 Geneva, Switzerland
| | - Xiaofan Zhou
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| | - Flaminia Catteruccia
- Harvard School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA 02115, USA. Dipartimento di Medicina Sperimentale e Scienze Biochimiche, Università degli Studi di Perugia, Perugia, Italy
| | - George K Christophides
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Frank H Collins
- Eck Institute for Global Health and Department of Biological Sciences, University of Notre Dame, 317 Galvin Life Sciences Building, Notre Dame, IN 46556, USA
| | - Robert S Cornman
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Andrea Crisanti
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK. Centre of Functional Genomics, University of Perugia, Perugia, Italy
| | - Martin J Donnelly
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK. Malaria Programme, Wellcome Trust Sanger Institute, Cambridge CB10 1SJ, UK
| | - Scott J Emrich
- Department of Computer Science and Engineering, Eck Institute for Global Health, 211B Cushing Hall, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Michael C Fontaine
- Eck Institute for Global Health and Department of Biological Sciences, University of Notre Dame, 317 Galvin Life Sciences Building, Notre Dame, IN 46556, USA. Centre of Evolutionary and Ecological Studies (Marine Evolution and Conservation group), University of Groningen, Nijenborgh 7, NL-9747 AG Groningen, Netherlands
| | - William Gelbart
- Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, Cambridge, MA 02138, USA
| | - Matthew W Hahn
- Department of Biology, Indiana University, Bloomington, IN 47405, USA. School of Informatics and Computing, Indiana University, Bloomington, IN 47405, USA
| | - Immo A Hansen
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, USA. Molecular Biology Program, New Mexico State University, Las Cruces, NM 88003, USA
| | - Paul I Howell
- Centers for Disease Control and Prevention, 1600 Clifton Road NE MSG49, Atlanta, GA 30329, USA
| | - Fotis C Kafatos
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, 32 Vassar Street, Cambridge, MA 02139, USA. The Broad Institute of Massachusetts Institute of Technology and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Daniel Lawson
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Christos Louis
- Department of Biology, University of Crete, 700 13 Heraklion, Greece. Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Hellas, Nikolaou Plastira 100 GR-70013, Heraklion, Crete, Greece. Centre of Functional Genomics, University of Perugia, Perugia, Italy
| | - Shirley Luckhart
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Marc A T Muskavitch
- Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA. Biogen Idec, 14 Cambridge Center, Cambridge, MA 02142, USA
| | - José M Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Michael A Riehle
- Department of Entomology, 1140 East South Campus Drive, Forbes 410, University of Arizona, Tucson, AZ 85721, USA
| | - Igor V Sharakhov
- Department of Entomology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA. Program of Genetics, Bioinformatics, and Computational Biology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Zhijian Tu
- Program of Genetics, Bioinformatics, and Computational Biology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA. Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Laurence J Zwiebel
- Departments of Biological Sciences and Pharmacology, Institutes for Chemical Biology, Genetics and Global Health, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Nora J Besansky
- Eck Institute for Global Health and Department of Biological Sciences, University of Notre Dame, 317 Galvin Life Sciences Building, Notre Dame, IN 46556, USA.
| |
Collapse
|
32
|
Calcium mobilization is both required and sufficient for initiating chromatin decondensation during activation of peripheral T-cells. Mol Immunol 2014; 63:540-9. [PMID: 25453467 DOI: 10.1016/j.molimm.2014.10.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 10/06/2014] [Accepted: 10/17/2014] [Indexed: 01/10/2023]
Abstract
Antigen engagement of the T-cell receptor (TCR) induces a rapid and dramatic decondensation of chromatin that is necessary for T-cell activation. This decondensation makes T-cells competent to respond to interleukin-2 providing a mechanism to ensure clonotypic proliferation during an immune response. Using murine T-cells, we investigated the mechanism by which TCR signaling can initiate chromatin decondensation, focusing on the role of calcium mobilization. During T-cell activation, calcium is first released from intracellular stores, followed by influx of extracellular calcium via store operated calcium entry. We show that mobilization of intracellular calcium is required for TCR-induced chromatin decondensation. However, the decondensation is not dependent on the activity of the downstream transcription factor NFAT. Furthermore, we show that the influx of extracellular calcium is dispensable for initiating chromatin decondensation. Finally, we show that mobilization of calcium from intracellular stores is sufficient to induce decondensation, independent of TCR engagement. Collectively, our data suggest that chromatin decondensation in peripheral T-cells is controlled by modulating intracellular calcium levels.
Collapse
|
33
|
Shin J, Jang H, Lin J, Lee SY. PKCβ positively regulates RANKL-induced osteoclastogenesis by inactivating GSK-3β. Mol Cells 2014; 37:747-52. [PMID: 25256217 PMCID: PMC4213766 DOI: 10.14348/molcells.2014.0220] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 08/18/2014] [Accepted: 08/18/2014] [Indexed: 12/14/2022] Open
Abstract
Protein kinase C (PKC) family members phosphorylate a wide variety of protein targets and are known to be involved in diverse cellular signaling pathways. However, the role of PKC in receptor activator of NF-κB ligand (RANKL) signaling has remained elusive. We now demonstrate that PKCβ acts as a positive regulator which inactivates glycogen synthase kinase-3β (GSK-3β) and promotes NFATc1 induction during RANKL-induced osteoclastogenesis. Among PKCs, PKCβ expression is increased by RANKL. Pharmacological inhibition of PKCβ decreased the formation of osteoclasts which was caused by the inhibition of NFATc1 induction. Importantly, the phosphorylation of GSK-3β was decreased by PKCβ inhibition. Likewise, down-regulation of PKCβ by RNA interference suppressed osteoclast differentiation, NFATc1 induction, and GSK-3β phosphorylation. The administration of PKC inhibitor to the RANKL-injected mouse calvaria efficiently protected RANKL-induced bone destruction. Thus, the PKCβ pathway, leading to GSK-3β inactivation and NFATc1 induction, has a key role in the differentiation of osteoclasts. Our results also provide a further rationale for PKCβ's therapeutic targeting to treat inflammation-related bone diseases.
Collapse
Affiliation(s)
- Jihye Shin
- Department of Life Science and the Research Center for Cellular Home-ostasis, Ewha Womans University, Seoul 120-750,
Korea
| | - Hyunduk Jang
- Department of Life Science and the Research Center for Cellular Home-ostasis, Ewha Womans University, Seoul 120-750,
Korea
- Present address: Department of Neurology, Seoul National University Hospital, and College of Medicine and Neuroscience Research Institute, Medical Research Center, Seoul National University, Seoul 110-749,
Korea
| | - Jingjing Lin
- Department of Life Science and the Research Center for Cellular Home-ostasis, Ewha Womans University, Seoul 120-750,
Korea
| | - Soo Young Lee
- Department of Life Science and the Research Center for Cellular Home-ostasis, Ewha Womans University, Seoul 120-750,
Korea
| |
Collapse
|
34
|
Huang Y, Ladinig A, Ashley C, Haines DM, Harding JCS. Innate and adaptive immune responses of snatch-farrowed porcine-colostrum-deprived pigs to Mycoplasma hyopneumoniae vaccination. BMC Vet Res 2014; 10:219. [PMID: 25239088 PMCID: PMC4180582 DOI: 10.1186/s12917-014-0219-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 09/17/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The snatch-farrowed porcine-colostrum-deprived (SF-pCD) pig model, in which neonates are raised on commercially available bovine colostrum, is an alternative model for porcine infectious disease research. It is not known if SF-pCD pigs possess growth performance and immunity comparable to conventional, farm-raised pigs. The current experiment compared growth performance and immune responses of SF-pCD pigs to their farm-raised siblings following Mycoplasma hyopneumoniae (Mhyo) vaccination. Twelve SF-pCD and 13 farm-raised siblings were vaccinated on day 7 (D7) and D26 of age. Body weights were measured once or twice weekly and average daily gain (ADG) was calculated. Peripheral blood mononuclear cells (PBMC) were isolated on D40. Cytokine secretion from PBMC stimulated with Mhyo antigen or phorbol myristate acetate plus ionomycin (PMA/Iono) was assessed using a multiplexed fluorescent microsphere immunoassay (FMIA). Additionally, interferon gamma (IFNγ) secretion from stimulated PBMC was assessed using ELISPOT. Mhyo IgG titers were measured by an ELISA in D40 sera. RESULTS Growth performance did not differ between groups before weaning, but SF-pCD pigs had higher ADG after weaning. In response to Mhyo stimulation, numbers of IFNγ secreting PBMC and levels of interleukin 8 (IL8) and IL10 in PBMC supernatants were significantly higher in SF-pCD pigs, as were Mhyo antibody levels in sera, and levels of IL1β, IL8 and IL12 in supernatants of PMA/Iono stimulated PBMC. CONCLUSIONS Under the conditions of this experiment, SF-pCD pigs demonstrated superior growth performance and enhanced humoral and cell-mediated immunity following vaccination. Whether or not this reflects greater resistance or tolerance to infection is unknown but the ability to react positively to the vaccination provides evidence that SF-pCD pigs are a suitable alternative model for swine disease research.
Collapse
|
35
|
Ooi EL, Chan ST, Cho NE, Wilkins C, Woodward J, Li M, Kikkawa U, Tellinghuisen T, Gale M, Saito T. Novel antiviral host factor, TNK1, regulates IFN signaling through serine phosphorylation of STAT1. Proc Natl Acad Sci U S A 2014; 111:1909-14. [PMID: 24449862 PMCID: PMC3918791 DOI: 10.1073/pnas.1314268111] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In response to viral infection, the host induces over 300 IFN-stimulated genes (ISGs), which are the central component of intracellular antiviral innate immunity. Inefficient induction of ISGs contributes to poor control and persistence of hepatitis C virus infection. Therefore, further understanding of the hepatocytic ISG regulation machinery will guide us to an improved management strategy against hepatitis C virus infection. In this study, comprehensive genome-wide, high-throughput cDNA screening for genes regulating ISG expression identified a tyrosine kinase nonreceptor 1 (TNK1) as a unique player in the ISG induction pathway. The immune-modulatory function of TNK1 has never been studied, and this study characterizes its significance in antiviral innate immunity. TNK1 is abundantly expressed in hepatocytes and maintains basal ISG expression. More importantly, TNK1 plays a critical role in type I IFN-mediated ISG induction. We discovered that the activated IFN receptor complex recruits TNK1 from the cytoplasm. TNK1 is then phosphorylated to enhance its kinase activity. The activated TNK1 potentiates JAK-STAT signaling through dual phosphorylation of STAT1 at tyrosine 701 and serine 727 amino acid positions. Our loss-of-function approach demonstrated that TNK1 governs a cluster of ISG expression that defines the TNK1 pathway effector genes. More importantly, TNK1 abundance is inversely correlated to viral replication efficiency and is also a determinant factor for the hepatocytic response to antiviral treatment. Taken together, our studies found a critical but unidentified integrated component of the IFN-JAK-STAT signaling cascade.
Collapse
Affiliation(s)
- Ee Lyn Ooi
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Stephanie T. Chan
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Noell E. Cho
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Courtney Wilkins
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA 98195-7650
| | - Jessica Woodward
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA 98195-7650
| | - Meng Li
- Bioinformatics Service, Norris Medical Library, University of Southern California, Los Angeles, CA 90089
| | - Ushio Kikkawa
- Biosignal Research Center, Kobe University, Nada-ku, Kobe 657-8501, Japan; and
| | - Timothy Tellinghuisen
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, FL 33458
| | - Michael Gale
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA 98195-7650
| | - Takeshi Saito
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA 98195-7650
| |
Collapse
|
36
|
Abstract
Bile acids, synthesized from cholesterol, are known to produce beneficial as well as toxic effects in the liver. The beneficial effects include choleresis, immunomodulation, cell survival, while the toxic effects include cholestasis, apoptosis and cellular toxicity. It is believed that bile acids produce many of these effects by activating intracellular signaling pathways. However, it has been a challenge to relate intracellular signaling to specific and at times opposing effects of bile acids. It is becoming evident that bile acids produce different effects by activating different isoforms of phosphoinositide 3-kinase (PI3K), Protein kinase Cs (PKCs), and mitogen activated protein kinases (MAPK). Thus, the apoptotic effect of bile acids may be mediated via PI3K-110γ, while cytoprotection induce by cAMP-GEF pathway involves activation of PI3K-p110α/β isoforms. Atypical PKCζ may mediate beneficial effects and nPKCε may mediate toxic effects, while cPKCα and nPKCδ may be involved in both beneficial and toxic effects of bile acids. The opposing effects of nPKCδ activation may depend on nPKCδ phosphorylation site(s). Activation of ERK1/2 and JNK1/2 pathway appears to mediate beneficial and toxic effects, respectively, of bile acids. Activation of p38α MAPK and p38β MAPK may mediate choleretic and cholestatic effects, respectively, of bile acids. Future studies clarifying the isoform specific effects on bile formation should allow us to define potential therapeutic targets in the treatment of cholestatic disorders.
Collapse
Affiliation(s)
- Mohammed Sawkat Anwer
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, 200 Westboro Road, North Grafton, MA, USA
| |
Collapse
|
37
|
O'Brian CA, Chu F, Bornmann WG, Maxwell DS. Protein kinase Cα and ε small-molecule targeted therapeutics: a new roadmap to two Holy Grails in drug discovery? Expert Rev Anticancer Ther 2014; 6:175-86. [PMID: 16445370 DOI: 10.1586/14737140.6.2.175] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Protein kinase (PK)Calpha and epsilon are rational targets for cancer therapy. However, targeted experimental therapeutics that inhibit PKCalpha or epsilon are unavailable. The authors established recently that covalent modification of an active-site cysteine in human PKCepsilon, Cys452, by small molecules, for example 2-mercaptoethanolamine, is necessary and sufficient to render PKCepsilon kinase-dead. Cys452 is conserved in only eleven human protein kinase genes, including PKCalpha. Therefore, the design of small molecules that bind PKC active sites with an electrophile substituent positioned proximal to the Cys452 side chain may lead to targeted therapeutics that selectively inhibit PKCepsilon, PKCalpha or other PKC isozymes.
Collapse
|
38
|
Koutsogiannaki S, Franzellitti S, Fabbri E, Kaloyianni M. Oxidative stress parameters induced by exposure to either cadmium or 17β-estradiol on Mytilus galloprovincialis hemocytes. The role of signaling molecules. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 146:186-195. [PMID: 24316436 DOI: 10.1016/j.aquatox.2013.11.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/24/2013] [Accepted: 11/07/2013] [Indexed: 06/02/2023]
Abstract
The aim of the present study was to determine and compare the possible effects of exposure to an estrogen, 17β-estradiol and to a metal, cadmium on oxidative parameters of Mytilus galloprovincialis hemocytes and to elucidate the signaling pathways that probably mediate the studied effects exerted by these two chemicals. In addition, it was of interest to investigate if the studied parameters could constitute biomarkers for aquatic pollution monitoring. Our results suggest that micromolar concentrations of either cadmium or 17β-estradiol affected the redox status of mussels by modulating oxidative parameters and antioxidant enzymes gene expression in mussel M. galloprovincialis hemocytes. In particular, our results showed that treatment of hemocytes with either 5 μM of cadmium chloride or with 25 nM of 17β-estradiol for 30 min caused significant increased ROS production; this led to oxidative damage exemplified by significant increased DNA damage, protein carbonylation and lipid peroxidation, as well as increased mRNA levels of the antioxidant enzymes catalase (CAT), superoxide dismoutase (SOD) and glutathione S-transferase (GST). Furthermore, our results suggest that either cadmium or 17β-estradiol signal is mediated either through one of the already known pathways initiated by photatidyl-inositol 3-kinase (PI3K) and reaching Na(+)/H(+) exchanger (NHE) probably through protein kinase C (PKC) or a kinase-mediated signaling pathway that involves in most of the cases NHE, PKC, Ca(2+)-dependent PKC isoforms, PI3-K, NADPH oxidase, nitric oxide (NO) synthase, c-Jun N-terminal kinase (JNK) and cyclic adenosine-3'-5'-monophosphate (cAMP). Our results also attribute a protective role to cAMP, since pre-elevated intracellular cAMP levels inhibited the signal induced by each exposure. Finally, since aquatic invertebrates have been the most widely used monitoring organisms for pollution impact evaluation in marine environments and taking under consideration the positive correlation obtained between the studied parameters, we can suggest the simultaneous use of these oxidative stress parameters offering an effective early warning system in biomonitoring of aquatic environments.
Collapse
Affiliation(s)
- Sophia Koutsogiannaki
- Laboratory of Animal Physiology, Zoology Department, School of Biology, Faculty of Science, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Silvia Franzellitti
- University of Bologna, Interdepartment Centre for Environmental Science Research, via S. Alberto 163, 48123 Ravenna, Italy
| | - Elena Fabbri
- University of Bologna, Interdepartment Centre for Environmental Science Research, via S. Alberto 163, 48123 Ravenna, Italy; University of Bologna, Department of Biological, Geological, and Environmental Sciences, via Selmi 3, 40100 Bologna, Italy
| | - Martha Kaloyianni
- Laboratory of Animal Physiology, Zoology Department, School of Biology, Faculty of Science, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| |
Collapse
|
39
|
|
40
|
Neuroprotective effects of quercetin in chemical hypoxia: in silico evaluation of the hypothesis exploring PKC inhibition-mediated pharmacotherapy. Med Chem Res 2013. [DOI: 10.1007/s00044-013-0503-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
41
|
Kim YD, Kwon MS, Na BR, Kim HR, Lee HS, Jun CD. Swiprosin-1 Expression Is Up-Regulated through Protein Kinase C-θ and NF-κB Pathway in T Cells. Immune Netw 2013; 13:55-62. [PMID: 23700395 PMCID: PMC3659256 DOI: 10.4110/in.2013.13.2.55] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 02/15/2013] [Accepted: 02/21/2013] [Indexed: 12/01/2022] Open
Abstract
Swiprosin-1 exhibits the highest expression in CD8+ T cells and immature B cells and has been proposed to play a role in lymphocyte biology through actin remodeling. However, regulation of swiprosin-1 gene expression is poorly understood. Here we report that swiprosin-1 is up-regulated in T cells by PKC pathway. Targeted inhibition of the specific protein kinase C (PKC) isotypes by siRNA revealed that PKC-θ is involved in the expression of swiprosin-1 in the human T cells. In contrast, down-regulation of swiprosin-1 by A23187 or ionomycin suggests that calcium-signaling plays a negative role. Interestingly, swiprosin-1 expression is only reduced by treatment with NF-κB inhibitors but not by NF-AT inhibitor, suggesting that the NF-κB pathway is critical for regulation of swiprosin-1 expression. Collectively, these results suggest that swiprosin-1 is a PKC-θ-inducible gene and that it may modulate the late phase of T cell activation after antigen challenge.
Collapse
Affiliation(s)
- Young-Dae Kim
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | | | | | | | | | | |
Collapse
|
42
|
Lutz-Nicoladoni C, Christina LN, Thuille N, Nikolaus T, Wachowicz K, Katarzyna W, Gruber T, Thomas G, Leitges M, Michael L, Baier G, Gottfried B. PKCα and PKCβ cooperate functionally in CD3-induced de novo IL-2 mRNA transcription. Immunol Lett 2013; 151:31-8. [PMID: 23439007 PMCID: PMC3641392 DOI: 10.1016/j.imlet.2013.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 01/29/2013] [Accepted: 02/05/2013] [Indexed: 12/12/2022]
Abstract
The physiological functions of PKCα and PKCθ isotypes downstream of the antigen receptor have been defined in CD3(+) T cells. In contrast, no function of the second conventional PKC member, PKCβ, has been described yet in T cell antigen receptor signalling. To investigate the hypothesis that both conventional PKCα and PKCβ isotypes may have overlapping functions in T cell activation signalling, we generated mice that lacked the genes for both isotypes. We found that PKCα(-/-)/β(-/-) animals are viable, live normal life spans and display normal T cell development. However, these animals possess additive defects in T cell responses in comparison to animals that carry single mutations in these genes. Our studies demonstrate that the activities of PKCα and PKCβ converge to regulate IL-2 cytokine responses in anti-CD3 stimulated primary mouse T cells. Here, we present genetic evidence that PKCα and PKCβ cooperate in IL-2 transcriptional transactivation in primary mouse T cells independently of the actions of PKCθ.
Collapse
|
43
|
Host signal transduction and protein kinases implicated in Legionella infection. Curr Top Microbiol Immunol 2013; 376:249-69. [PMID: 23918173 DOI: 10.1007/82_2013_342] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Modulation of the phosphorylation status of proteins by both kinases and phosphatases plays an important role in cellular signal transduction. Challenge of host cells by Legionella pneumophila manipulates the phosphorylation state of multiple host factors. These changes play roles in bacterial uptake, vacuole modification, cellular survival, and the immune response. In addition to modification by host cell kinases in response to the bacterium, L. pneumophila translocates bacterial kinases into the host cell that may contribute to further signaling modifications. Proper regulation of host cell signaling by L. pneumophila is necessary for its ability to replicate intracellulary, while avoiding host defenses.
Collapse
|
44
|
Joo DJ, Fang YH, Huh KH, Kim MS, Suh H, Kim YS. Changes in Serum Cytokine Profile after AEB071 (Sotrastaurin) or Tacrolimus versus Their Combinations in Rat Heterotopic Cardiac Allografts. KOREAN JOURNAL OF TRANSPLANTATION 2012. [DOI: 10.4285/jkstn.2012.26.4.248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Dong Jin Joo
- The Research Institute for Transplantation,Yonsei University College of Medicine, Seoul, Korea
- Department of Transplantation Surgery, Yonsei University Health System, Seoul, Korea
| | - Yu Hui Fang
- The Research Institute for Transplantation,Yonsei University College of Medicine, Seoul, Korea
- Graduate Program of Nanoscience and Technology, Yonsei University Health System, Seoul, Korea
| | - Kyu Ha Huh
- The Research Institute for Transplantation,Yonsei University College of Medicine, Seoul, Korea
- Department of Transplantation Surgery, Yonsei University Health System, Seoul, Korea
| | - Myoung Soo Kim
- The Research Institute for Transplantation,Yonsei University College of Medicine, Seoul, Korea
- Department of Transplantation Surgery, Yonsei University Health System, Seoul, Korea
| | - Hwal Suh
- Graduate Program of Nanoscience and Technology, Yonsei University Health System, Seoul, Korea
| | - Yu Seun Kim
- The Research Institute for Transplantation,Yonsei University College of Medicine, Seoul, Korea
- Department of Transplantation Surgery, Yonsei University Health System, Seoul, Korea
- Graduate Program of Nanoscience and Technology, Yonsei University Health System, Seoul, Korea
- Yonsei University, BK for Medical Science, Yonsei University Health System, Seoul, Korea
| |
Collapse
|
45
|
Chan CN, McMonagle EL, Hosie MJ, Willett BJ. Prostratin exhibits both replication enhancing and inhibiting effects on FIV infection of feline CD4+ T-cells. Virus Res 2012. [PMID: 23201205 PMCID: PMC3566544 DOI: 10.1016/j.virusres.2012.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The phorbol ester Prostratin may either stimulate or inhibit human immunodeficiency virus-1 (HIV-1) replication. Here we report that Prostratin also exhibits a similar dual action upon feline immunodeficiency virus (FIV) replication in an IL-2-dependent feline CD4+ T-cell line (MYA-1). While withdrawal of IL-2 halted FIV spread, Prostratin rescued virus production and cell viability, mimicking the functions of the cytokine. Conversely, FIV grew rapidly in the presence of IL-2 and this was inhibited by Prostratin. In contrast to HIV-1, Prostratin mediated inhibition of FIV through means other than blocking virus entry. Co-application of the protein kinase C (PKC) inhibitor Gö6850 with Prostratin reversed both the inhibitory and stimulatory effects, suggesting that PKC is crucial for FIV replication.
Collapse
Affiliation(s)
- Chi Ngai Chan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom.
| | | | | | | |
Collapse
|
46
|
Wang C, Shang Y, Yu J, Zhang M. Substrate recognition mechanism of atypical protein kinase Cs revealed by the structure of PKCι in complex with a substrate peptide from Par-3. Structure 2012; 20:791-801. [PMID: 22579248 DOI: 10.1016/j.str.2012.02.022] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 02/24/2012] [Accepted: 02/27/2012] [Indexed: 01/24/2023]
Abstract
Protein kinase C (PKC) play critical roles in many cellular functions including differentiation, proliferation, growth, and survival. However, the molecular bases governing PKC's substrate recognitions remain poorly understood. Here we determined the structure of PKCι in complex with a peptide from Par-3 at 2.4 Å. PKCι in the complex adopts catalytically competent, closed conformation without phosphorylation of Thr402 in the activation loop. The Par-3 peptide binds to an elongated groove formed by the N- and C-lobes of the kinase domain. The PKCι/Par-3 complex structure, together with extensive biochemical studies, reveals a set of substrate recognition sites common to all PKC isozymes as well as a hydrophobic pocket unique to aPKC. A consensus aPKC's substrate recognition sequence pattern can be readily identified based on the complex structure. Finally, we demonstrate that the pseudosubstrate sequence of PKCι resembles its substrate sequence, directly binds to and inhibits the activity of the kinase.
Collapse
Affiliation(s)
- Chihao Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | | | | | | |
Collapse
|
47
|
Martin P, Moscat J. Th1/Th2 Differentiation and B Cell Function by the Atypical PKCs and Their Regulators. Front Immunol 2012; 3:241. [PMID: 22888333 PMCID: PMC3412266 DOI: 10.3389/fimmu.2012.00241] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 07/17/2012] [Indexed: 12/24/2022] Open
Abstract
The members of the atypical Protein Kinase Cs (aPKC) kinase subfamily, PKCζ and PKCλ/ɩ, as well as their adapters, p62 and Par-6, form part of the PB1-domain-containing group of signaling regulators. Both adapters serve to locate through heterotypic interactions the aPKCs into the NF-κB and cell polarity pathways, respectively. Both signaling cascades have been critically implicated in T cell function in vitro and in vivo. The analysis of gene-knockout (KO) mice deficient in the different PB1 molecules is providing more definitive information on the actual role that the aPKCs and other PB1-containing molecules play in B cell biology and T cell polarity, survival, and differentiation toward the different effector lineages in vivo and at the cellular ex vivo level. Here we discuss recent data generated from the analysis of KO mice linking the control of cell polarity by PKCλ/ɩ and PKCζ, their adapter p62, and the Par-4 inhibitor, in the control of B and T cell signaling and differentiation. Altogether, these genetic and biochemical evidences reveal the existence of a PB1-orchestrated signaling network that acts to control Th2 differentiation in vitro and in vivo, and the gene transcriptional programs that are essential during the B cell maturation and function and Th2 differentiation.
Collapse
Affiliation(s)
- Pilar Martin
- Department of Vascular Biology and Inflammation, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III Madrid, Spain
| | | |
Collapse
|
48
|
Nakagawa R, Vukovic M, Cosimo E, Michie AM. Modulation of PKC-α promotes lineage reprogramming of committed B lymphocytes. Eur J Immunol 2012; 42:1005-15. [PMID: 22531924 DOI: 10.1002/eji.201141442] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
During hematopoietic lineage development, hematopoietic stem cells sequentially commit toward myeloid or lymphoid lineages in a tightly regulated manner, which under normal circumstances is irreversible. However, studies have established that targeted deletion of the B-lineage specific transcription factor, paired box gene 5 (Pax5), enables B cells to differentiate toward other hematopoietic lineages, in addition to generating progenitor B-cell lymphomas. Our previous studies showed that subversion of protein kinase C (PKC)-α in developing B cells transformed B-lineage cells. Here, we demonstrate that PKC-α modulation in committed CD19(+) B lymphocytes also promoted lineage conversion toward myeloid, NK-, and T-cell lineages upon Notch ligation. This occurred via a reduction in Pax5 expression resulting from a downregulation of E47, a product of the E2A gene. T-cell lineage commitment was indicated by the expression of T-cell associated genes Ptcra, Cd3e, and gene rearrangement at the Tcrb gene locus. Importantly, the lineage-converted T cells carried Igh gene rearrangements reminiscent of their B-cell origin. Our findings suggest that modulation of PKC-α induces hematopoietic-lineage plasticity in committed B-lineage cells by perturbing expression of critical B-lineage transcription factors, and deregulation of PKC-α activity/expression represents a potential mechanism for lineage trans-differentiation during malignancies.
Collapse
Affiliation(s)
- Rinako Nakagawa
- Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | | | | | | |
Collapse
|
49
|
Feng X, Zhang J, Smuga-Otto K, Tian S, Yu J, Stewart R, Thomson JA. Protein kinase C mediated extraembryonic endoderm differentiation of human embryonic stem cells. Stem Cells 2012; 30:461-70. [PMID: 22213079 DOI: 10.1002/stem.1018] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Unlike mouse embryonic stem cells (ESCs), which are closely related to the inner cell mass, human ESCs appear to be more closely related to the later primitive ectoderm. For example, human ESCs and primitive ectoderm share a common epithelial morphology, growth factor requirements, and the potential to differentiate to all three embryonic germ layers. However, it has previously been shown that human ESCs can also differentiate to cells expressing markers of trophoblast, an extraembryonic lineage formed before the formation of primitive ectoderm. Here, we show that phorbol ester 12-O-tetradecanoylphorbol 13-acetate causes human ESCs to undergo an epithelial mesenchymal transition and to differentiate into cells expressing markers of parietal endoderm, another extraembryonic lineage. We further confirmed that this differentiation is through the activation of protein kinase C (PKC) pathway and demonstrated that a particular PKC subtype, PKC-δ, is most responsible for this transition.
Collapse
Affiliation(s)
- Xuezhu Feng
- Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Yang Y, Wang X, Zhang L, An H, Zao Z. Inhibitory Effects of Resveratrol on Platelet Activation Induced by Thromboxane A2Receptor Agonist in Human Platelets. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 39:145-59. [PMID: 21213405 DOI: 10.1142/s0192415x11008713] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Resveratrol (RSVL), a polyphenolic compound found in red wine is believed to be a contributor in decreasing the incidence of coronary heart disease. Although its primary target is unknown, it blocks platelet aggregation by an ill-defined mechanism. Protein kinase C (PKC), which would redistribute from the cytosol to the platelet membrane upon platelet stimulation, plays a key role in the signal transduction system of platelets in human. In this study, we investigated the effect of RSVL and a PKC inhibitor (DL-erythro-1,3-Dihydroxy-2-aminooctadecane, PKCI) on platelet aggregation induced by a thromboxane A2receptor agonist (U46619, 9,11-Dideoxy-11α, 9α-epoxymethanoprostaglandin F2α) using a platelet aggregometer. We also studied the platelet membranebound fibrinogen (PFig) content and the activity of protein kinase C (PKC) in platelets from healthy volunteers using flow cytometry, and a phosphorimaging system, respectively. Our results showed that RSVL blocked platelet aggregation and PFig content induced by U46619 in a concentration-dependent manner. PKCI and RSVL had an additive effect in inhibiting platelet aggregation and PFig content. Furthermore, RSVL (final concentration 50 μM) remarkably depressed the activity of PKC in the membrane of platelets and the percentage of membrane PKC activity in total PKC activity. Taken together, these results suggested that RSVL suppressed U46619-induced platelet aggregation and PFig content partially through the inhibition of the activity of PKC in platelets.
Collapse
Affiliation(s)
- Yumin Yang
- Department of Cardiovascular Disease, Traditional Chinese-Mongolian Hospital of Inner Mongolia Autonomous Region, Hohhot 010020, P. R. China
| | - Xiaoling Wang
- Department of Cardiovascular Disease, Traditional Chinese-Mongolian Hospital of Inner Mongolia Autonomous Region, Hohhot 010020, P. R. China
| | - Li Zhang
- Department of Cardiovascular Disease, Traditional Chinese-Mongolian Hospital of Inner Mongolia Autonomous Region, Hohhot 010020, P. R. China
| | - Huiping An
- Department of Pathology and Pathophysiology, Inner Mongolia Medical College, Hohhot 010020, P. R. China
| | - Zhigao Zao
- Department of Pathology and Pathophysiology, Inner Mongolia Medical College, Hohhot 010020, P. R. China
| |
Collapse
|