1
|
Gaba S, Sahu M, Chauhan N, Jain U. Transforming growth factor alpha: Key insights into physiological role, cancer therapeutics, and biomarker potential (A review). Int J Biol Macromol 2025; 310:143212. [PMID: 40250676 DOI: 10.1016/j.ijbiomac.2025.143212] [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: 01/29/2025] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Transforming Growth Factor Alpha (TGF-α) is a critical member of the epidermal growth factor (EGF) family and a key regulator of various physiological processes, including cellular proliferation, survival, differentiation, wound repair, and tissue regeneration. Deficiencies or mutations in TGF-α have been associated with impaired tissue development and organ growth, underscoring its critical role in maintaining normal and healthy physiology. Alterations in its levels are frequently implicated in the neoplastic transformation of cells, contributing to cancer development. Several strategies for targeting TGF-α in cancer therapy have been explored, such as the use of antibodies, recombinant proteins, oligonucleotide-mediated interference in ligand synthesis, ligand sequestration via binding proteins, and modulation of the signal transduction pathway. Furthermore, there is growing interest in the potential of TGF-α as a diagnostic or prognostic biomarker for cancer. This review delves into the role of TGF-α in normal physiology and its involvement in carcinogenesis. It highlights therapies targeting TGF-α and explores future directions in targeting TGF-α/EGFR signaling using advancing approaches, including nanoparticle-based drug delivery systems, CRISPR-Cas genome editing tool, PROTAC, and combination therapies. By bringing attention to this molecule, we aim to explore its untapped potential in cancer treatment and inspire further research into its promising applications across related fields. While recent studies highlight the promise of TGF-α as a clinical biomarker, further research is needed to validate its specificity and integration into personalized medicine. By providing a comprehensive overview of TGF-α in both normal and pathological contexts, this review aims to offer new insights into its translational applications in cancer therapeutics and biomarker discovery.
Collapse
Affiliation(s)
- Smriti Gaba
- School of Health Sciences and Technology, UPES, Dehradun 248007, India
| | - Mridul Sahu
- School of Health Sciences and Technology, UPES, Dehradun 248007, India
| | - Nidhi Chauhan
- School of Health Sciences and Technology, UPES, Dehradun 248007, India
| | - Utkarsh Jain
- School of Health Sciences and Technology, UPES, Dehradun 248007, India.
| |
Collapse
|
2
|
Montgomery A, Westphal J, Bryan AE, Harris GM. Dynamically changing extracellular matrix stiffness drives Schwann cell phenotype. Matrix Biol Plus 2025; 25:100167. [PMID: 39868413 PMCID: PMC11754676 DOI: 10.1016/j.mbplus.2024.100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025] Open
Abstract
Schwann cells (SCs) hold key roles in axonal function and maintenance in the peripheral nervous system (PNS) and are a critical component to the regeneration process following trauma. Following PNS trauma, SCs respond to both physical and chemical signals to modify phenotype and assist in the regeneration of damaged axons and extracellular matrix (ECM). There is currently a lack of knowledge regarding the SC response to dynamic, temporal changes in the ECM brought on by swelling and the development of scar tissue as part of the body's wound-healing process. Thus, this work seeks to utilize a biocompatible, mechanically tunable biomaterial to mimic changes in the microenvironment following injury and over time. Previously, we have reported that ECM cues such as ligand type and substrate stiffness impact SC phenotype and plasticity, which was demonstrated by SCs on mechanically stable biomaterials. However, to better realize SC potential for plasticity following traumatic injury, a UV-tunable polydimethylsiloxane (PDMS) substrate with dynamically changing stiffness was utilized to mimic changes over time in the microenvironment. The dynamic biomaterial showed an increase in stress fibers, greater YAP expression, and fluctuations in c-Jun production in SCs in comparison to stiff and soft static controls. Utilizing biomaterials to better understand the role between temporal mechanical dynamics and SC phenotype holds a very high potential for developing future PNS therapies.
Collapse
Affiliation(s)
- Alyssa Montgomery
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jennifer Westphal
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Andrew E. Bryan
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Greg M. Harris
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Cincinnati VA Medical Center, Cincinnati, OH 45220, USA
| |
Collapse
|
3
|
Ramos-Alvarez I, Jensen RT. The Important Role of p21-Activated Kinases in Pancreatic Exocrine Function. BIOLOGY 2025; 14:113. [PMID: 40001881 PMCID: PMC11851965 DOI: 10.3390/biology14020113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/27/2025]
Abstract
The p21-activated kinases (PAKs) are a conserved family of serine/threonine protein kinases, which are effectors for the Rho family GTPases, namely, Rac/Cdc42. PAKs are divided into two groups: group I (PAK1-3) and group II (PAK4-6). Both groups of PAKs have been well studied in apoptosis, protein synthesis, glucose homeostasis, growth (proliferation and survival) and cytoskeletal regulation, as well as in cell motility, proliferation and cycle control. However, little is known about the role of PAKs in the secretory tissues, including in exocrine tissue, such as the exocrine pancreas (except for islet function and pancreatic cancer growth). Recent studies have provided insights supporting the importance of PAKs in exocrine pancreas. This review summarizes the recent insights into the importance of PAKs in the exocrine pancreas by reviewing their presence and activation; the ability of GI hormones/neurotransmitters/GFs/post-receptor activators to activate them; the kinetics of their activation; the participation of exocrine-tissue PAKs in activating the main growth-signaling cascade; their roles in the stimulation of enzyme secretion; finally, their roles in pancreatitis. These insights suggest that PAKs could be more important in exocrine/secretory tissues than currently appreciated and that their roles should be explored in more detail in the future.
Collapse
Affiliation(s)
| | - Robert T. Jensen
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20812-1804, USA;
| |
Collapse
|
4
|
Patel Y, Prajapati A. Unveiling LGR5: Prostate cancer's hidden stem cell and treatment target. Urol Oncol 2024; 42:438-446. [PMID: 39406640 DOI: 10.1016/j.urolonc.2024.10.001] [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: 07/24/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 11/20/2024]
Abstract
Prostate cancer poses a significant risk to the well-being and way of life of countless men, with an increased likelihood of relapse recorded following modern treatment. This highlights the need for innovative approaches, specifically targeting LGR5. This systematic review aims to establish a connection between LGR5 and the various signaling pathways involved in the progression of prostate cancer. LGR5, a gene targeted by Wnt signaling, encodes a receptor protein that serves as a prognostic biomarker for stem cells and indicates the presence of cancer stem cells in colorectal and gastrointestinal cancers. The functions of LGR5 include processes such as cell proliferation, differentiation, and signaling pathways. Any modifications to the LGR5 gene, whether caused by mutations or mechanical stimuli, can lead to the development of treatment-resistant stem cell cancers. This review examines the molecular mechanisms associated with LGR5 and emphasizes methodologies aimed at targeting LGR5 to enhance understanding and promote the development of LGR5-specific therapies.
Collapse
Affiliation(s)
- Yashvi Patel
- Department of Life Science, Biotechnology Division, School of Science, GSFC University, Vadodara, 391750, Gujarat, India
| | - Akhilesh Prajapati
- Department of Life Science, Biotechnology Division, School of Science, GSFC University, Vadodara, 391750, Gujarat, India.
| |
Collapse
|
5
|
Liu X, Lai J, Zhang X, Wu A, Zhou L, Li Y, Huang Q, Huang X, Li H, Lan C, Liu J, Huang F, Wu J. Harmine promotes megakaryocyte differentiation and thrombopoiesis by activating the Rac1/Cdc42/JNK pathway through a potential target of 5-HTR2A. Phytother Res 2024; 38:5134-5149. [PMID: 39152726 DOI: 10.1002/ptr.8317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 08/19/2024]
Abstract
Harmine (HM), a β-carboline alkaloid extracted from plants, is a crucial component of traditional Chinese medicine (TCM) known for its diverse pharmacological activities. Thrombocytopenia, a common and challenging hematological disorder, often coexists with serious illnesses. Previous research has shown a correlation between HM and thrombocytopenia, but the mechanism needs further elucidation. The aim of this study was to clarify the mechanisms underlying the effects of HM on thrombocytopenia and to develop new therapeutic strategies. Flow cytometry, Giemsa staining, and Phalloidin staining were used to assess HM's impact on Meg-01 and HEL cell differentiation and maturation in vitro. A radiation-induced thrombocytopenic mouse model was employed to evaluate HM's effect on platelet production in vivo. Network pharmacology, molecular docking, and protein blotting were utilized to investigate HM's targets and mechanisms. The results demonstrated that HM dose-dependently promoted Meg-01 and HEL cell differentiation and maturation in vitro and restored platelet levels in irradiated mice in vivo. Subsequently, HM was found to be involved in the biological process of platelet production by upregulating the expressions of Rac1, Cdc42, JNK, and 5-HTR2A. Furthermore, the targeting of HM to 5-HTR2A and its correlation with downstream Rac1/Cdc42/JNK were also confirmed. In conclusion, HM regulates megakaryocyte differentiation and thrombopoiesis through the 5-HTR2A and Rac1/Cdc42/JNK pathways, providing a potential treatment strategy for thrombocytopenia.
Collapse
Affiliation(s)
- Xiaoxi Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jia Lai
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, China
| | - Xiaoqin Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Anguo Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Ling Zhou
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yueyue Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qianqian Huang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xinwu Huang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hua Li
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Cai Lan
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Feihong Huang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianming Wu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, China
| |
Collapse
|
6
|
de Carvalho ABG, Rahimnejad M, Oliveira RLMS, Sikder P, Saavedra GSFA, Bhaduri SB, Gawlitta D, Malda J, Kaigler D, Trichês ES, Bottino MC. Personalized bioceramic grafts for craniomaxillofacial bone regeneration. Int J Oral Sci 2024; 16:62. [PMID: 39482290 PMCID: PMC11528123 DOI: 10.1038/s41368-024-00327-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 11/03/2024] Open
Abstract
The reconstruction of craniomaxillofacial bone defects remains clinically challenging. To date, autogenous grafts are considered the gold standard but present critical drawbacks. These shortcomings have driven recent research on craniomaxillofacial bone reconstruction to focus on synthetic grafts with distinct materials and fabrication techniques. Among the various fabrication methods, additive manufacturing (AM) has shown significant clinical potential. AM technologies build three-dimensional (3D) objects with personalized geometry customizable from a computer-aided design. These layer-by-layer 3D biomaterial structures can support bone formation by guiding cell migration/proliferation, osteogenesis, and angiogenesis. Additionally, these structures can be engineered to degrade concomitantly with the new bone tissue formation, making them ideal as synthetic grafts. This review delves into the key advances of bioceramic grafts/scaffolds obtained by 3D printing for personalized craniomaxillofacial bone reconstruction. In this regard, clinically relevant topics such as ceramic-based biomaterials, graft/scaffold characteristics (macro/micro-features), material extrusion-based 3D printing, and the step-by-step workflow to engineer personalized bioceramic grafts are discussed. Importantly, in vitro models are highlighted in conjunction with a thorough examination of the signaling pathways reported when investigating these bioceramics and their effect on cellular response/behavior. Lastly, we summarize the clinical potential and translation opportunities of personalized bioceramics for craniomaxillofacial bone regeneration.
Collapse
Affiliation(s)
- Ana Beatriz G de Carvalho
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Department of Dental Materials and Prosthodontics, São Paulo State University, São José dos Campos, SP, Brazil
| | - Maedeh Rahimnejad
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Rodrigo L M S Oliveira
- Federal University of São Paulo, Institute of Science and Technology, São José dos Campos, SP, Brazil
| | - Prabaha Sikder
- Department of Mechanical Engineering, Cleveland State University, Cleveland, OH, USA
| | - Guilherme S F A Saavedra
- Department of Dental Materials and Prosthodontics, São Paulo State University, São José dos Campos, SP, Brazil
| | - Sarit B Bhaduri
- Department of Mechanical, Industrial and Manufacturing Engineering, University of Toledo, Toledo, OH, USA
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, The Netherlands
| | - Jos Malda
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Darnell Kaigler
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Eliandra S Trichês
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Federal University of São Paulo, Institute of Science and Technology, São José dos Campos, SP, Brazil
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Didan Y, Ghomlaghi M, Nguyen LK, Ng DCH. Stress pathway outputs are encoded by pH-dependent clustering of kinase components. Nat Commun 2024; 15:6614. [PMID: 39103333 DOI: 10.1038/s41467-024-50638-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Signal processing by intracellular kinases controls near all biological processes but how signal pathway functions evolve with changed cellular context is poorly understood. Functional specificity of c-Jun N-terminal Kinases (JNK) are partly encoded by signal strength. Here we reveal that intracellular pH (pHi) is a significant component of the JNK network and defines signal response to specific stimuli. We show pHi regulates JNK activity in response to cell stress, with the relationship between pHi and JNK activity dependent on specific stimuli and upstream kinases activated. Using the optogenetic clustering tag CRY2, we show that an increase in pHi promotes the light-induced phase transition of ASK1 to augment JNK activation. While increased pHi similarly promoted CRY2-tagged JNK2 to form light-induced condensates, this attenuated JNK activity. Mathematical modelling of feedback signalling incorporating pHi and differential contributions by ASK1 and JNK2 condensates was sufficient to delineate signal responses to specific stimuli. Taking pHi and ASK1/JNK2 signal contributions into consideration may delineate oncogenic versus tumour suppressive JNK functions and cancer cell drug responses.
Collapse
Affiliation(s)
- Yuliia Didan
- School of Biomedical Science, Faculty of Medicine, University of Queensland; St Lucia, Brisbane, Australia
| | - Milad Ghomlaghi
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Lan K Nguyen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Dominic C H Ng
- School of Biomedical Science, Faculty of Medicine, University of Queensland; St Lucia, Brisbane, Australia.
| |
Collapse
|
8
|
Lemche E, Killick R, Mitchell J, Caton PW, Choudhary P, Howard JK. Molecular mechanisms linking type 2 diabetes mellitus and late-onset Alzheimer's disease: A systematic review and qualitative meta-analysis. Neurobiol Dis 2024; 196:106485. [PMID: 38643861 DOI: 10.1016/j.nbd.2024.106485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/23/2024] Open
Abstract
Research evidence indicating common metabolic mechanisms through which type 2 diabetes mellitus (T2DM) increases risk of late-onset Alzheimer's dementia (LOAD) has accumulated over recent decades. The aim of this systematic review is to provide a comprehensive review of common mechanisms, which have hitherto been discussed in separate perspectives, and to assemble and evaluate candidate loci and epigenetic modifications contributing to polygenic risk linkages between T2DM and LOAD. For the systematic review on pathophysiological mechanisms, both human and animal studies up to December 2023 are included. For the qualitative meta-analysis of genomic bases, human association studies were examined; for epigenetic mechanisms, data from human studies and animal models were accepted. Papers describing pathophysiological studies were identified in databases, and further literature gathered from cited work. For genomic and epigenomic studies, literature mining was conducted by formalised search codes using Boolean operators in search engines, and augmented by GeneRif citations in Entrez Gene, and other sources (WikiGenes, etc.). For the systematic review of pathophysiological mechanisms, 923 publications were evaluated, and 138 gene loci extracted for testing candidate risk linkages. 3 57 publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight insulin signalling, inflammation and inflammasome pathways, proteolysis, gluconeogenesis and glycolysis, glycosylation, lipoprotein metabolism and oxidation, cell cycle regulation or survival, autophagic-lysosomal pathways, and energy. Documented findings suggest interplay between brain insulin resistance, neuroinflammation, insult compensatory mechanisms, and peripheral metabolic dysregulation in T2DM and LOAD linkage. The results allow for more streamlined longitudinal studies of T2DM-LOAD risk linkages.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom.
| | - Richard Killick
- Section of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom
| | - Jackie Mitchell
- Department of Basic and Clinical Neurosciences, Maurice Wohl CIinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom
| | - Paul W Caton
- Diabetes Research Group, School of Life Course Sciences, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, United Kingdom
| | - Pratik Choudhary
- Diabetes Research Group, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, Hodgkin Building, Guy's Campus, King's College London, Great Maze Pond, London SE1 1UL, United Kingdom
| |
Collapse
|
9
|
Ahmed-de-Prado S, Estella C, Baonza A. Temporal dynamics of apoptosis-induced proliferation in pupal wing development: implications for regenerative ability. BMC Biol 2024; 22:98. [PMID: 38679694 PMCID: PMC11057159 DOI: 10.1186/s12915-024-01894-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND The ability of animals to regenerate damaged tissue is a complex process that involves various cellular mechanisms. As animals age, they lose their regenerative abilities, making it essential to understand the underlying mechanisms that limit regenerative ability during aging. Drosophila melanogaster wing imaginal discs are epithelial structures that can regenerate after tissue injury. While significant research has focused on investigating regenerative responses during larval stages our comprehension of the regenerative potential of pupal wings and the underlying mechanisms contributing to the decline of regenerative responses remains limited. RESULTS Here, we explore the temporal dynamics during pupal development of the proliferative response triggered by the induction of cell death, a typical regenerative response. Our results indicate that the apoptosis-induced proliferative response can continue until 34 h after puparium formation (APF), beyond this point cell death alone is not sufficient to induce a regenerative response. Under normal circumstances, cell proliferation ceases around 24 h APF. Interestingly, the failure of reinitiating the cell cycle beyond this time point is not attributed to an incapacity to activate the JNK pathway. Instead, our results suggest that the function of the ecdysone-responsive transcription factor E93 is involved in limiting the apoptosis-induced proliferative response during pupal development. CONCLUSIONS Our study shows that apoptosis can prolong the proliferative period of cells in the wing during pupal development as late as 34 h APF, at least 10 h longer than during normal development. After this time point, the regenerative response is diminished, a process mediated in part by the ecdysone-responsive transcription factor E93.
Collapse
Affiliation(s)
| | - Carlos Estella
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM), C/Nicolás Cabrera 1, Madrid, 28049, Spain
| | - Antonio Baonza
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM), C/Nicolás Cabrera 1, Madrid, 28049, Spain.
| |
Collapse
|
10
|
Ma Y, Xiao Q, Kang C, Yuan L, Zhao Z, Wang X, Liu Q, Guo W, Wei X, Hao W. Effect of lanthanum nitrate on adipogenesis in mice. J Appl Toxicol 2024; 44:542-552. [PMID: 37908164 DOI: 10.1002/jat.4558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/02/2023]
Abstract
Lanthanum (La) is widely used in modern industry and agriculture because of its unique physicochemical properties and is broadly exposed in the population. Some studies have shown that La may have some effects on adipogenesis, but there is a lack of related in vivo evidence. In this study, the effects of La(NO3 )3 on adipogenesis and its associated mechanism were studied using C57BL/6J mouse model. The results showed that La(NO3 )3 exposure caused a decrease in body weight and the percentage of fat content in mice. In addition, the adipose marker molecules and specific adipogenic transcription factors decreased in both white adipose tissue (WAT) and brown adipose tissue (BAT). Detection of signaling pathway-related molecules revealed that canonical wnt/β-catenin pathway-related molecules were upregulated in both adipose tissues. In summary, in vivo exposure to La(NO3 )3 might inhibited adipogenesis in mice, possibly through upregulation of the canonical Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yuhong Ma
- Department of Toxicology, School of Public Health, Peking University, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China
| | - Qianqian Xiao
- Department of Toxicology, School of Public Health, Peking University, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China
| | - Chenping Kang
- Department of Toxicology, School of Public Health, Peking University, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China
| | - Lilan Yuan
- Department of Toxicology, School of Public Health, Peking University, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China
| | - Zhe Zhao
- Department of Toxicology, School of Public Health, Peking University, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China
| | - Xiaoxia Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China
| | - Qianyi Liu
- Department of Toxicology, School of Public Health, Peking University, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China
| | - Wanqian Guo
- Department of Toxicology, School of Public Health, Peking University, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Peking University, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China
| | - Weidong Hao
- Department of Toxicology, School of Public Health, Peking University, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China
| |
Collapse
|
11
|
Yan H, He L, Lv D, Yang J, Yuan Z. The Role of the Dysregulated JNK Signaling Pathway in the Pathogenesis of Human Diseases and Its Potential Therapeutic Strategies: A Comprehensive Review. Biomolecules 2024; 14:243. [PMID: 38397480 PMCID: PMC10887252 DOI: 10.3390/biom14020243] [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: 12/06/2023] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
JNK is named after c-Jun N-terminal kinase, as it is responsible for phosphorylating c-Jun. As a member of the mitogen-activated protein kinase (MAPK) family, JNK is also known as stress-activated kinase (SAPK) because it can be activated by extracellular stresses including growth factor, UV irradiation, and virus infection. Functionally, JNK regulates various cell behaviors such as cell differentiation, proliferation, survival, and metabolic reprogramming. Dysregulated JNK signaling contributes to several types of human diseases. Although the role of the JNK pathway in a single disease has been summarized in several previous publications, a comprehensive review of its role in multiple kinds of human diseases is missing. In this review, we begin by introducing the landmark discoveries, structures, tissue expression, and activation mechanisms of the JNK pathway. Next, we come to the focus of this work: a comprehensive summary of the role of the deregulated JNK pathway in multiple kinds of diseases. Beyond that, we also discuss the current strategies for targeting the JNK pathway for therapeutic intervention and summarize the application of JNK inhibitors as well as several challenges now faced. We expect that this review can provide a more comprehensive insight into the critical role of the JNK pathway in the pathogenesis of human diseases and hope that it also provides important clues for ameliorating disease conditions.
Collapse
Affiliation(s)
- Huaying Yan
- Department of Ultrasound, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (H.Y.); (L.H.)
| | - Lanfang He
- Department of Ultrasound, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; (H.Y.); (L.H.)
| | - De Lv
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Jun Yang
- Cancer Center and State Key Laboratory of Biotherapy, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Zhu Yuan
- Cancer Center and State Key Laboratory of Biotherapy, Department of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China;
| |
Collapse
|
12
|
Martínez-Esquivias F, Perez-Larios A, Guzmán-Flores JM. Effect of Administration of Selenium Nanoparticles Synthesized Using Onion Extract on Biochemical and Inflammatory Parameters in Mice Fed with High-Fructose Diet: In Vivo and In Silico Analysis. Biol Trace Elem Res 2024; 202:558-568. [PMID: 37119340 DOI: 10.1007/s12011-023-03685-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/26/2023] [Indexed: 05/01/2023]
Abstract
Fructose consumption has increased globally and has been linked to obesity, insulin resistance, and diabetes. Selenium nanoparticles (SeNPs) can regulate glucose and lipid concentrations and have immunoregulatory properties. Four study groups (n = 7/group) of eight-week-old male mice (Balb/c) were formed for this investigation. One group received a standard diet (C), another standard diet plus SeNPs (C + SeNPs), a high fructose diet (F), and a group with a high fructose diet plus SeNPs (F + SeNPs). Weight, glucose, triglycerides, and cholesterol were evaluated. In the end, mice were sacrificed, blood samples were obtained to assess cytokine profile, and liver, kidney, and pancreas were removed for histological examination. The study was complemented with an in silico analysis where the CTD, STITCH, ToppGene Suite, ShinyGO 0.76.3 databases, and Cytoscape software were implemented. The results of in vivo analysis showed that SeNPs regulated biochemical parameters and showed anti-inflammatory effects by decreasing the concentrations of TNF-alpha, IL-1beta, and IFN-gamma and increasing IL-10. No damage was observed in the studied organs. In addition, SeNPs regulate oxidative stress, preserve cell organelles, and regulate metabolic pathways to avoid the adverse effects of fructose consumption, according to bioinformatics analysis. In conclusion, SeNPs protect against the undesirable effects of a diet rich in fructose.
Collapse
Affiliation(s)
- Fernando Martínez-Esquivias
- Instituto de Investigación en Biociencias, Centro Universitario de Los Altos, Universidad de Guadalajara, Av. Rafael Casillas Aceves 1200, Tepatitlán de Morelos, 47600, Jalisco, México
| | - Alejandro Perez-Larios
- Laboratorio de Materiales, Centro Universitario de Los Altos, Universidad de Guadalajara, Agua y Energía, Tepatitlán de Morelos, Jalisco, México
| | - Juan Manuel Guzmán-Flores
- Instituto de Investigación en Biociencias, Centro Universitario de Los Altos, Universidad de Guadalajara, Av. Rafael Casillas Aceves 1200, Tepatitlán de Morelos, 47600, Jalisco, México.
| |
Collapse
|
13
|
Sharma C, Mazumder A. A Comprehensive Review on Potential Molecular Drug Targets for the Management of Alzheimer's Disease. Cent Nerv Syst Agents Med Chem 2024; 24:45-56. [PMID: 38305393 DOI: 10.2174/0118715249263300231116062740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/25/2023] [Accepted: 10/04/2023] [Indexed: 02/03/2024]
Abstract
Alzheimer's disease (AD) is an onset and incurable neurodegenerative disorder that has been linked to various genetic, environmental, and lifestyle factors. Recent research has revealed several potential targets for drug development, such as the prevention of Aβ production and removal, prevention of tau hyperphosphorylation, and keeping neurons alive. Drugs that target numerous ADrelated variables have been developed, and early results are encouraging. This review provides a concise map of the different receptor signaling pathways associated with Alzheimer's Disease, as well as insight into drug design based on these pathways. It discusses the molecular mechanisms of AD pathogenesis, such as oxidative stress, aging, Aβ turnover, thiol groups, and mitochondrial activities, and their role in the disease. It also reviews the potential drug targets, in vivo active agents, and docking studies done in AD and provides prospects for future drug development. This review intends to provide more clarity on the molecular processes that occur in Alzheimer's patient's brains, which can be of use in diagnosing and preventing the condition.
Collapse
Affiliation(s)
- Chanchal Sharma
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida-201306, Uttar Pradesh, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida-201306, Uttar Pradesh, India
| |
Collapse
|
14
|
Bahar ME, Kim HJ, Kim DR. Targeting the RAS/RAF/MAPK pathway for cancer therapy: from mechanism to clinical studies. Signal Transduct Target Ther 2023; 8:455. [PMID: 38105263 PMCID: PMC10725898 DOI: 10.1038/s41392-023-01705-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/03/2023] [Accepted: 11/12/2023] [Indexed: 12/19/2023] Open
Abstract
Metastatic dissemination of solid tumors, a leading cause of cancer-related mortality, underscores the urgent need for enhanced insights into the molecular and cellular mechanisms underlying metastasis, chemoresistance, and the mechanistic backgrounds of individuals whose cancers are prone to migration. The most prevalent signaling cascade governed by multi-kinase inhibitors is the mitogen-activated protein kinase (MAPK) pathway, encompassing the RAS-RAF-MAPK kinase (MEK)-extracellular signal-related kinase (ERK) pathway. RAF kinase is a primary mediator of the MAPK pathway, responsible for the sequential activation of downstream targets, such as MEK and the transcription factor ERK, which control numerous cellular and physiological processes, including organism development, cell cycle control, cell proliferation and differentiation, cell survival, and death. Defects in this signaling cascade are associated with diseases such as cancer. RAF inhibitors (RAFi) combined with MEK blockers represent an FDA-approved therapeutic strategy for numerous RAF-mutant cancers, including melanoma, non-small cell lung carcinoma, and thyroid cancer. However, the development of therapy resistance by cancer cells remains an important barrier. Autophagy, an intracellular lysosome-dependent catabolic recycling process, plays a critical role in the development of RAFi resistance in cancer. Thus, targeting RAF and autophagy could be novel treatment strategies for RAF-mutant cancers. In this review, we delve deeper into the mechanistic insights surrounding RAF kinase signaling in tumorigenesis and RAFi-resistance. Furthermore, we explore and discuss the ongoing development of next-generation RAF inhibitors with enhanced therapeutic profiles. Additionally, this review sheds light on the functional interplay between RAF-targeted therapies and autophagy in cancer.
Collapse
Affiliation(s)
- Md Entaz Bahar
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea.
| |
Collapse
|
15
|
Mersal KI, Abdel-Maksoud MS, Ali EMH, Ammar UM, Zaraei SO, Haque MM, Das T, Hassan NF, Kim EE, Lee JS, Park H, Lee KH, El-Gamal MI, Kim HK, Ibrahim TM, Oh CH. Evaluation of novel pyrazol-4-yl pyridine derivatives possessing arylsulfonamide tethers as c-Jun N-terminal kinase (JNK) inhibitors in leukemia cells. Eur J Med Chem 2023; 261:115779. [PMID: 37776574 DOI: 10.1016/j.ejmech.2023.115779] [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: 05/18/2023] [Revised: 08/14/2023] [Accepted: 08/29/2023] [Indexed: 10/02/2023]
Abstract
A series of 36 pyrazol-4-yl pyridine derivatives (8a-i, 9a-i, 10a-i, and 11a-i) was designed, synthesized, and evaluated for its antiproliferative activity over NCI-60 cancer cell line panel and inhibitory effect against JNK isoforms (JNK1, JNK2, and JNK3). All the synthesized compounds were tested against the NCI-60 cancer cell line panel. Compounds 11b, 11c, 11g, and 11i were selected to determine their GI50s and exerted a superior potency over the reference standard SP600125 against the tested cell lines. 11c showed a GI50 of 1.28 μM against K562 leukemic cells. Vero cells were used to assess 11c cytotoxicity compared to the tested cancer cells. The target compounds were tested against hJNK isoforms in which compound 11e exhibited the highest potency against JNK isoforms with IC50 values of 1.81, 12.7, and 10.5 nM against JNK1, JNK2, and JNK3, respectively. Kinase profiling of 11e showed higher JNK selectivity in 50 kinase panels. Compounds 11c and 11e showed cell population arrest at the G2/M phase, induced early apoptosis, and slightly inhibited beclin-1 production at higher concentrations in K562 leukemia cells relative to SP600125. NanoBRET assay of 11e showed intracellular JNK1 inhibition with an IC50 of 2.81 μM. Also, it inhibited CYP2D6 and 3A4 with different extent and its hERG activity showed little cardiac toxicity with an IC50 of 4.82 μM. hJNK3 was used as a template to generate the hJNK1 crystal structure to explore the binding mode of 11e (PDB ID: 8ENJ) with a resolution of 2.8 °A and showed a typical type I kinase inhibition against hJNK1. Binding energy scores showed that selectivity of 11e towards JNK1 could be attributed to additional hydrophobic interactions relative to JNK3.
Collapse
Affiliation(s)
- Karim I Mersal
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, 12055, Egypt; University of Science & Technology (UST), Daejeon, Yuseong-gu, 34113, Republic of Korea; Center of Biomaterials, Korea Institute of Science & Technology (KIST School), Seoul, Seongbuk-gu, 02792, Republic of Korea
| | - Mohammed S Abdel-Maksoud
- Medicinal & Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre NRC (ID: 60014618), Dokki, Giza, 12622, Egypt
| | - Eslam M H Ali
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, 12055, Egypt; Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN, 47907, USA
| | - Usama M Ammar
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, 9 Sighthill Court, Edinburgh, EH11 4BN, United Kingdom
| | - Seyed-Omar Zaraei
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Md Mamunul Haque
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, 21201, USA
| | - Tanuza Das
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Noha F Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, 12055, Egypt
| | - Eunice EunKyeong Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, South Korea
| | - Jun-Seok Lee
- Department of Pharmacology, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - HaJeung Park
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, USA
| | - Kwan Hyi Lee
- Center for Advanced Biomolecular Recognition, Korea Institute of Science & Technology (KIST School), Seoul, Seongbuk-gu, 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Mohammed I El-Gamal
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Hee-Kwon Kim
- Department of Nuclear Medicine, Molecular Imaging & Therapeutic Medicine Research Center, Jeonbuk National University Medical School and Hospital, 20 Geonji-ro, Deokjin-gu, Jeonju, 54907, Republic of Korea; Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, 20 Geonji-ro, Deokjin-gu, Jeonju, 54907, Republic of Korea.
| | - Tamer M Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt
| | - Chang-Hyun Oh
- University of Science & Technology (UST), Daejeon, Yuseong-gu, 34113, Republic of Korea; Center of Biomaterials, Korea Institute of Science & Technology (KIST School), Seoul, Seongbuk-gu, 02792, Republic of Korea.
| |
Collapse
|
16
|
Narożna M, Krajka-Kuźniak V, Bednarczyk-Cwynar B, Baer-Dubowska W. Unlocking the Potential: Novel NSAIDs Hybrids Unleash Chemopreventive Power toward Liver Cancer Cells through Nrf2, NF-κB, and MAPK Signaling Pathways. Molecules 2023; 28:5759. [PMID: 37570726 PMCID: PMC10420225 DOI: 10.3390/molecules28155759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
HCC is a highly aggressive malignancy with limited treatment options. In this study, novel conjugates of non-steroidal anti-inflammatory drugs (NSAIDs)-Ibuprofen and Ketoprofen-with oleanolic acid oximes derivatives (OAO) were synthesized, and their activity as modulators of signaling pathways involved in HCC pathogenesis was evaluated in normal THLE-2 liver cells, and HCC-derived HepG2 cells. The results demonstrated that conjugation with OAO derivatives reduces the cytotoxicity of parent compounds in both cell lines. In THLE-2 cells, treatment with conjugates resulted in increased activation of the Nrf2-ARE pathway. An opposite effect was observed in HepG2 cells. In the later reduction of NF-κB, it was observed along with modulation of MAPK signaling pathways (AKT, ERK, p38, p70S6K, and JNK). Moreover, STAT3, STAT5, and CREB transcription factors on protein levels were significantly reduced as a result of treatment with IBU- and KET-OAO derivatives conjugates. The most active were conjugates with OAO-morpholide. Overall, the findings of this study demonstrate that IBU-OAO and KET-OAO derivative conjugates modulate the key signaling pathways involved in hepatic cancer development. Their effect on specific signaling pathways varied depending on the structure of the conjugate. Since the conjugation of IBU and KET with OAO derivatives reduced their cytotoxicity, the conjugates may be considered good candidates for the prevention of liver cancer.
Collapse
Affiliation(s)
- Maria Narożna
- Program in Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, 825, NE 13th Street, Oklahoma City, OK 73104, USA;
| | - Violetta Krajka-Kuźniak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, 4, Święcicki Street, 60-781 Poznań, Poland;
| | - Barbara Bednarczyk-Cwynar
- Department of Organic Chemistry, Poznan University of Medical Sciences, 6, Grunwaldzka Street, 60-780 Poznań, Poland;
| | - Wanda Baer-Dubowska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, 4, Święcicki Street, 60-781 Poznań, Poland;
| |
Collapse
|
17
|
Chowdhury I, Dashi G, Keskitalo S. CMGC Kinases in Health and Cancer. Cancers (Basel) 2023; 15:3838. [PMID: 37568654 PMCID: PMC10417348 DOI: 10.3390/cancers15153838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/18/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
CMGC kinases, encompassing cyclin-dependent kinases (CDKs), mitogen-activated protein kinases (MAPKs), glycogen synthase kinases (GSKs), and CDC-like kinases (CLKs), play pivotal roles in cellular signaling pathways, including cell cycle regulation, proliferation, differentiation, apoptosis, and gene expression regulation. The dysregulation and aberrant activation of these kinases have been implicated in cancer development and progression, making them attractive therapeutic targets. In recent years, kinase inhibitors targeting CMGC kinases, such as CDK4/6 inhibitors and BRAF/MEK inhibitors, have demonstrated clinical success in treating specific cancer types. However, challenges remain, including resistance to kinase inhibitors, off-target effects, and the need for better patient stratification. This review provides a comprehensive overview of the importance of CMGC kinases in cancer biology, their involvement in cellular signaling pathways, protein-protein interactions, and the current state of kinase inhibitors targeting these kinases. Furthermore, we discuss the challenges and future perspectives in targeting CMGC kinases for cancer therapy, including potential strategies to overcome resistance, the development of more selective inhibitors, and novel therapeutic approaches, such as targeting protein-protein interactions, exploiting synthetic lethality, and the evolution of omics in the study of the human kinome. As our understanding of the molecular mechanisms and protein-protein interactions involving CMGC kinases expands, so too will the opportunities for the development of more selective and effective therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Iftekhar Chowdhury
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland; (I.C.)
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Giovanna Dashi
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland; (I.C.)
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Salla Keskitalo
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland; (I.C.)
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
18
|
Valverde-Salazar V, Ruiz-Gabarre D, García-Escudero V. Alzheimer's Disease and Green Tea: Epigallocatechin-3-Gallate as a Modulator of Inflammation and Oxidative Stress. Antioxidants (Basel) 2023; 12:1460. [PMID: 37507998 PMCID: PMC10376369 DOI: 10.3390/antiox12071460] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, characterised by a marked decline of both memory and cognition, along with pathophysiological hallmarks including amyloid beta peptide (Aβ) accumulation, tau protein hyperphosphorylation, neuronal loss and inflammation in the brain. Additionally, oxidative stress caused by an imbalance between free radicals and antioxidants is considered one of the main risk factors for AD, since it can result in protein, lipid and nucleic acid damage and exacerbate Aβ and tau pathology. To date, there is a lack of successful pharmacological approaches to cure or even ameliorate the terrible impact of this disease. Due to this, dietary compounds with antioxidative and anti-inflammatory properties acquire special relevance as potential therapeutic agents. In this context, green tea, and its main bioactive compound, epigallocatechin-3-gallate (EGCG), have been targeted as a plausible option for the modulation of AD. Specifically, EGCG acts as an antioxidant by regulating inflammatory processes involved in neurodegeneration such as ferroptosis and microglia-induced cytotoxicity and by inducing signalling pathways related to neuronal survival. Furthermore, it reduces tau hyperphosphorylation and aggregation and promotes the non-amyloidogenic route of APP processing, thus preventing the formation of Aβ and its subsequent accumulation. Taken together, these results suggest that EGCG may be a suitable candidate in the search for potential therapeutic compounds for neurodegenerative disorders involving inflammation and oxidative stress, including Alzheimer's disease.
Collapse
Affiliation(s)
- Víctor Valverde-Salazar
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Daniel Ruiz-Gabarre
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Vega García-Escudero
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, 28031 Madrid, Spain
- Institute for Molecular Biology-IUBM, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
19
|
Ardanaz CG, Ezkurdia A, Bejarano A, Echarte B, Smerdou C, Martisova E, Martínez-Valbuena I, Luquin MR, Ramírez MJ, Solas M. JNK3 Overexpression in the Entorhinal Cortex Impacts on the Hippocampus and Induces Cognitive Deficiencies and Tau Misfolding. ACS Chem Neurosci 2023. [PMID: 37236204 DOI: 10.1021/acschemneuro.3c00092] [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: 05/28/2023] Open
Abstract
c-Jun N-terminal kinases (JNKs) are a family of protein kinases activated by a myriad of stimuli consequently modulating a vast range of biological processes. In human postmortem brain samples affected with Alzheimer's disease (AD), JNK overactivation has been described; however, its role in AD onset and progression is still under debate. One of the earliest affected areas in the pathology is the entorhinal cortex (EC). Noteworthy, the deterioration of the projection from EC to hippocampus (Hp) point toward the idea that the connection between EC and Hp is lost in AD. Thus, the main objective of the present work is to address if JNK3 overexpression in the EC could impact on the hippocampus, inducing cognitive deficits. Data obtained in the present work suggest that JNK3 overexpression in the EC influences the Hp leading to cognitive impairment. Moreover, proinflammatory cytokine expression and Tau immunoreactivity were increased both in the EC and in the Hp. Therefore, activation of inflammatory signaling and induction of Tau aberrant misfolding caused by JNK3 could be responsible for the observed cognitive impairment. Altogether, JNK3 overexpression in the EC may impact on the Hp inducing cognitive dysfunction and underlie the alterations observed in AD.
Collapse
Affiliation(s)
- Carlos G Ardanaz
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Amaia Ezkurdia
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Arantza Bejarano
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
| | - Beatriz Echarte
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
| | - Cristian Smerdou
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Eva Martisova
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Iván Martínez-Valbuena
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Neurosciences Division, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, M5S 1A8 Toronto, Canada
| | - María-Rosario Luquin
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Neurosciences Division, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Neurology Department, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - María J Ramírez
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Maite Solas
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| |
Collapse
|
20
|
García-Flores N, Jiménez-Suárez J, Garnés-García C, Fernández-Aroca DM, Sabater S, Andrés I, Fernández-Aramburo A, Ruiz-Hidalgo MJ, Belandia B, Sanchez-Prieto R, Cimas FJ. P38 MAPK and Radiotherapy: Foes or Friends? Cancers (Basel) 2023; 15:861. [PMID: 36765819 PMCID: PMC9913882 DOI: 10.3390/cancers15030861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/16/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Over the last 30 years, the study of the cellular response to ionizing radiation (IR) has increased exponentially. Among the various signaling pathways affected by IR, p38 MAPK has been shown to be activated both in vitro and in vivo, with involvement in key processes triggered by IR-mediated genotoxic insult, such as the cell cycle, apoptosis or senescence. However, we do not yet have a definitive clue about the role of p38 MAPK in terms of radioresistance/sensitivity and its potential use to improve current radiotherapy. In this review, we summarize the current knowledge on this family of MAPKs in response to IR as well as in different aspects related to radiotherapy, such as their role in the control of REDOX, fibrosis, and in the radiosensitizing effect of several compounds.
Collapse
Affiliation(s)
- Natalia García-Flores
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Jaime Jiménez-Suárez
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Cristina Garnés-García
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Diego M. Fernández-Aroca
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Sebastia Sabater
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Servicio de Oncología Radioterápica, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
| | - Ignacio Andrés
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Servicio de Oncología Radioterápica, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
| | - Antonio Fernández-Aramburo
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Servicio de Oncología Médica, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
| | - María José Ruiz-Hidalgo
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Borja Belandia
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain
| | - Ricardo Sanchez-Prieto
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain
- Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Francisco J. Cimas
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| |
Collapse
|
21
|
Craig RA, Fox BM, Hu C, Lexa KW, Osipov M, Thottumkara AP, Larhammar M, Miyamoto T, Rana A, Kane LA, Yulyaningsih E, Solanoy H, Nguyen H, Chau R, Earr T, Kajiwara Y, Fleck D, Lucas A, Haddick PCG, Takahashi RH, Tong V, Wang J, Canet MJ, Poda SB, Scearce-Levie K, Srivastava A, Sweeney ZK, Xu M, Zhang R, He J, Lei Y, Zhuo Z, de Vicente J. Discovery of Potent and Selective Dual Leucine Zipper Kinase/Leucine Zipper-Bearing Kinase Inhibitors with Neuroprotective Properties in In Vitro and In Vivo Models of Amyotrophic Lateral Sclerosis. J Med Chem 2022; 65:16290-16312. [PMID: 36469401 DOI: 10.1021/acs.jmedchem.2c01056] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Dual leucine zipper kinase (DLK) and leucine zipper-bearing kinase (LZK) are regulators of neuronal degeneration and axon growth. Therefore, there is a considerable interest in developing DLK/LZK inhibitors for neurodegenerative diseases. Herein, we use ligand- and structure-based drug design approaches for identifying novel amino-pyrazine inhibitors of DLK/LZK. DN-1289 (14), a potent and selective dual DLK/LZK inhibitor, demonstrated excellent in vivo plasma half-life across species and is anticipated to freely penetrate the central nervous system with no brain impairment based on in vivo rodent pharmacokinetic studies and human in vitro transporter data. Proximal target engagement and disease relevant pathway biomarkers were also favorably regulated in an in vivo model of amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Robert A Craig
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Brian M Fox
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Cheng Hu
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Katrina W Lexa
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Maksim Osipov
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Arun P Thottumkara
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Martin Larhammar
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Takashi Miyamoto
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Anil Rana
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Lesley A Kane
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Ernie Yulyaningsih
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Hilda Solanoy
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Hoang Nguyen
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Roni Chau
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Timothy Earr
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Yuji Kajiwara
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Daniel Fleck
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Anthony Lucas
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Patrick C G Haddick
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Ryan H Takahashi
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Vincent Tong
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Jing Wang
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Mark J Canet
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Suresh B Poda
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Kimberly Scearce-Levie
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Ankita Srivastava
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Zachary K Sweeney
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| | - Musheng Xu
- Department of Chemistry, WuXi AppTec Co., Ltd., 168 Nanhai Road, 10th Avenue, Tianjin TEDA, Tianjin 300457, China
| | - Rui Zhang
- Department of Chemistry, WuXi AppTec Co., Ltd., 168 Nanhai Road, 10th Avenue, Tianjin TEDA, Tianjin 300457, China
| | - Jianrong He
- Department of Chemistry, WuXi AppTec Co., Ltd., 168 Nanhai Road, 10th Avenue, Tianjin TEDA, Tianjin 300457, China
| | - Yanan Lei
- Department of Chemistry, WuXi AppTec Co., Ltd., 168 Nanhai Road, 10th Avenue, Tianjin TEDA, Tianjin 300457, China
| | - Zheng Zhuo
- Department of Chemistry, WuXi AppTec Co., Ltd., 168 Nanhai Road, 10th Avenue, Tianjin TEDA, Tianjin 300457, China
| | - Javier de Vicente
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, California 94080, United States
| |
Collapse
|
22
|
Metwally NH, Eldaly SM. Design, Synthesis of New Pyrazoles and Chromenes as ERK‐2 Inhibitors, Apoptosis inducers and Cell cycle interrupters Based on Thiophene‐Chalcone Scaffold. ChemistrySelect 2022. [DOI: 10.1002/slct.202202257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
| | - Salwa Magdy Eldaly
- Department of Chemistry Faculty of Science Cairo University POX. 12613 Giza Egypt
| |
Collapse
|
23
|
Gannam ZTK, Jamali H, Kweon OS, Herrington J, Shillingford SR, Papini C, Gentzel E, Lolis E, Bennett AM, Ellman JA, Anderson KS. Defining the structure-activity relationship for a novel class of allosteric MKP5 inhibitors. Eur J Med Chem 2022; 243:114712. [PMID: 36116232 PMCID: PMC9830533 DOI: 10.1016/j.ejmech.2022.114712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 01/13/2023]
Abstract
Mitogen-activated protein kinase (MAPK) phosphatase 5 (MKP5) is responsible for regulating the activity of the stress-responsive MAPKs and has been put forth as a potential therapeutic target for a number of diseases, including dystrophic muscle disease a fatal rare disease which has neither a treatment nor cure. In previous work, we identified Compound 1 (3,3-dimethyl-1-((9-(methylthio)-5,6-dihydrothieno[3,4-h]quinazolin-2-yl)thio)butan-2-one) as the lead compound of a novel class of MKP5 inhibitors. In this work, we explore the structure-activity relationship for inhibition of MKP5 through modifications to the scaffold and functional groups present in 1. A series of derivative compounds was designed, synthesized, and evaluated for inhibition of MKP5. In addition, the X-ray crystal structures of six enzyme-inhibitor complexes were solved, further elucidating the necessary requirements for MKP5 inhibition. We found that the parallel-displaced π-π interaction between the inhibitor three-ring core and Tyr435 is critical for modulating potency, and that modifications to the core and functionalization at the C-9 position are essential for ensuring proper positioning of the core for this interaction. These results lay the foundation from which more potent MKP5 allosteric inhibitors can be developed for potential therapeutics towards the treatment of dystrophic muscle disease.
Collapse
Affiliation(s)
- Zira T K Gannam
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Haya Jamali
- Department of Chemistry, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Oh Sang Kweon
- Department of Chemistry, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - James Herrington
- Yale Center for Molecular Discovery, Yale University School of Medicine, New Haven, CT, 06520, USA
| | | | - Christina Papini
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Erik Gentzel
- Department of Chemistry, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Elias Lolis
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Anton M Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Jonathan A Ellman
- Department of Chemistry, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Karen S Anderson
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA; Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
24
|
Kim J, Kim S, Uddin S, Lee SS, Park S. Microfabricated Stretching Devices for Studying the Effects of Tensile Stress on Cells and Tissues. BIOCHIP JOURNAL 2022; 16:366-375. [DOI: 10.1007/s13206-022-00073-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/05/2022] [Accepted: 07/12/2022] [Indexed: 11/02/2022]
|
25
|
Wang J, Kan X, Li X, Sun J, Xu X. Porcine epidemic diarrhoea virus (PEDV) infection activates AMPK and JNK through TAK1 to induce autophagy and enhance virus replication. Virulence 2022; 13:1697-1712. [PMID: 36168145 PMCID: PMC9543055 DOI: 10.1080/21505594.2022.2127192] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Autophagy plays an important role in defending against invading microbes. However, numerous viruses can subvert autophagy to benefit their replication. Porcine epidemic diarrhoea virus (PEDV) is an aetiological agent that causes severe porcine epidemic diarrhoea. How PEDV infection regulates autophagy and its role in PEDV replication are inadequately understood. Herein, we report that PEDV induced complete autophagy in Vero and IPEC-DQ cells, as evidenced by increased LC3 lipidation, p62 degradation, and the formation of autolysosomes. The lysosomal protease inhibitors chloroquine (CQ) or bafilomycin A and Beclin-1 or ATG5 knockdown blocked autophagic flux and inhibited PEDV replication. PEDV infection activated AMP-activated protein kinase (AMPK) and c-Jun terminal kinase (JNK) by activating TGF-beta-activated kinase 1 (TAK1). Compound C (CC), an AMPK inhibitor, and SP600125, a JNK inhibitor, inhibited PEDV-induced autophagy and virus replication. AMPK activation led to increased ULK1S777 phosphorylation and activation. Inhibition of ULK1 activity by SBI-0206965 (SBI) and TAK1 activity by 5Z-7-Oxozeaenol (5Z) or by TAK1 siRNA led to the suppression of autophagy and virus replication. Our study provides mechanistic insights into PEDV-induced autophagy and how PEDV infection leads to JNK and AMPK activation.
Collapse
Affiliation(s)
- Jingxiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, P. R. China
| | - Xianjin Kan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, P. R. China
| | - Xiaomei Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, P. R. China
| | - Jing Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, P. R. China.,Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, P. R. China
| | - Xiulong Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province, P. R. China.,Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province, P. R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu Province, China
| |
Collapse
|
26
|
Kim E, Suh JS, Jang YK, Kim H, Choi G, Kim TJ. Harmine inhibits proliferation and migration of glioblastoma via ERK signalling. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
27
|
Klymenko A, Lutz D. Melatonin signalling in Schwann cells during neuroregeneration. Front Cell Dev Biol 2022; 10:999322. [PMID: 36299487 PMCID: PMC9589221 DOI: 10.3389/fcell.2022.999322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
It has widely been thought that in the process of nerve regeneration Schwann cells populate the injury site with myelinating, non–myelinating, phagocytic, repair, and mesenchyme–like phenotypes. It is now clear that the Schwann cells modify their shape and basal lamina as to accommodate re–growing axons, at the same time clear myelin debris generated upon injury, and regulate expression of extracellular matrix proteins at and around the lesion site. Such a remarkable plasticity may follow an intrinsic functional rhythm or a systemic circadian clock matching the demands of accurate timing and precision of signalling cascades in the regenerating nervous system. Schwann cells react to changes in the external circadian clock clues and to the Zeitgeber hormone melatonin by altering their plasticity. This raises the question of whether melatonin regulates Schwann cell activity during neurorepair and if circadian control and rhythmicity of Schwann cell functions are vital aspects of neuroregeneration. Here, we have focused on different schools of thought and emerging concepts of melatonin–mediated signalling in Schwann cells underlying peripheral nerve regeneration and discuss circadian rhythmicity as a possible component of neurorepair.
Collapse
|
28
|
Gu Y, Hou T, Qin Y, Dong W. Zoledronate promotes osteoblast differentiation in high-glucose conditions via the p38MAPK pathway. Cell Biol Int 2022; 47:216-227. [PMID: 36193698 DOI: 10.1002/cbin.11921] [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: 11/24/2021] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 11/11/2022]
Abstract
Zoledronate (ZOL) were found to inhibit bone resorption in an animal model of diabetes, high glucose concentrations have been shown to decreased the osteogenesis-related gene expression. But the molecular mechanism by which high glucose levels affect osteoblasts and the effects of ZOL on osteoblast differentiation in a high-glucose environment remain unclear. Therefore, we aimed to investigate the effect of ZOL on osteoblast differentiation in a high-glucose environment and determine the responsible mechanism. Cell proliferation was detected by MTT assay, and cell differentiation was evaluated by immunofluorescence staining for alkaline phosphatase expression, alizarin red staining, cytoskeletal arrangement, and actin fiber formation. Real-time PCR and western blot analyses were performed to detect the mRNA and protein expression of p38MAPK, phosphorylated (p)-p38MAPK, CREB, p-CREB, collagen (COL) I, osteoprotegerin (OPG), and RANKL. The results showed that cell proliferation activity did not differ among the groups. But high glucose inhibited osteoblast differentiation; actin fiber formation; and p38MAPK, p-p38MAPK, CREB, p-CREB, COL I, and OPG expression, while promoting RANKL expression. However, we found that treatment with ZOL reversed these effects of high glucose. And further addition of a p38MAPK inhibitor led to inhibition of osteoblast differentiation and actin fiber formation, and lower p38MAPK, p-p38MAPK, CREB, p-CREB, COL I, and OPG expression than in the high glucose +ZOL group with higher RANKL expression than in the high glucose +ZOL group. Collectively, this study demonstrates that high glucose inhibits the differentiation of osteoblasts, and ZOL could partly overcome these effects by regulating p38MAPK pathway activity.
Collapse
Affiliation(s)
- Yingying Gu
- School of Stomatology, North China University of Science and Technology, Tangshan, China
| | - Tian Hou
- School of Stomatology, North China University of Science and Technology, Tangshan, China
| | - Yazhi Qin
- School of Stomatology, North China University of Science and Technology, Tangshan, China
| | - Wei Dong
- School of Stomatology, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
29
|
Yan J, Chen Q, Tian L, Li K, Lai W, Bian L, Han J, Jia R, Liu X, Xi Z. Intestinal toxicity of micro- and nano-particles of foodborne titanium dioxide in juvenile mice: Disorders of gut microbiota-host co-metabolites and intestinal barrier damage. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 821:153279. [PMID: 35074372 DOI: 10.1016/j.scitotenv.2022.153279] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/09/2022] [Accepted: 01/16/2022] [Indexed: 05/28/2023]
Abstract
The wide use of TiO2 particles in food and the high exposure risk to children have prompted research into the health risks of TiO2. We used the microbiome and targeted metabolomics to explore the potential mechanism of intestinal toxicity of foodborne TiO2 micro-/nanoparticles after oral exposure for 28 days in juvenile mice. Results showed that the gut microbiota-including the abundance of Bacteroides, Bifidobacterium, Lactobacillus, and Prevotella-changed dynamically during exposure. The organic inflammatory response was activated, and lipopolysaccharide levels increased. Intestinal toxicity manifested as increased mucosal permeability, impaired intestinal barrier, immune damage, and pathological changes. The expression of antimicrobial peptides, occludin, and ZO-1 significantly reduced, while that of JNK2 and Src/pSrc increased. Compared with micro-TiO2 particles, the nano-TiO2 particles had strong toxicity. Fecal microbiota transplant confirmed the key role of gut microbiota in intestinal toxicity. The levels of gut microbiota-host co-metabolites, including pyroglutamic acid, L-glutamic acid, phenylacetic acid, and 3-hydroxyphenylacetic acid, changed significantly. Significant changes were observed in the glutathione and propanoate metabolic pathways. There was a significant correlation between the changes in gut microbiota, metabolites, and intestinal cytokine levels. These, together with the intestinal barrier damage signaling pathway, constitute the network mechanism of the intestinal toxicity of TiO2 particles.
Collapse
Affiliation(s)
- Jun Yan
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Qi Chen
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Lei Tian
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Kang Li
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Wenqing Lai
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Liping Bian
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Jie Han
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Rui Jia
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China
| | - Xiaohua Liu
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China.
| | - Zhuge Xi
- Tianjin Institute of Environmental & Operational Medicine, No. 1, Dali Road, Heping District, Tianjin 300050, China.
| |
Collapse
|
30
|
Ali MA, Khalil MM, Al-Mokaddem AK, Aljuaydi SH, Ahmed M, Khalil HM. Differential effects of cancer modifying agents during radiation therapy on Ehrlich solid tumor-bearing mice: A comparative investigation of metformin and ascorbic acid. Appl Radiat Isot 2022; 187:110305. [DOI: 10.1016/j.apradiso.2022.110305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 05/15/2022] [Accepted: 05/18/2022] [Indexed: 11/02/2022]
|
31
|
Ma S, Shu X, Wang WX. Multi-omics reveals the regulatory mechanisms of zinc exposure on the intestine-liver axis of golden pompano Trachinotus ovatus. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 816:151497. [PMID: 34752869 DOI: 10.1016/j.scitotenv.2021.151497] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 06/13/2023]
Abstract
Metal zinc (Zn) has been the focus of many environmental toxicological studies, but there are limited studies on its potential dietary molecular toxicity and physiology. The present study was the first to use multi-omics-based approaches to explore the fish intestine-liver axis under dietary Zn exposure. Golden pompano Trachinotus ovatus were exposed to different dietary concentrations (78.4, 134.6, and 161.4 mg/kg as the control, low-dose Zn, and high-dose Zn groups, respectively) of Zn for 4-week. Low-dose Zn exposure significantly promoted the fish growth, whereas the high-dose Zn exposure reduced the fish growth. Co-analysis of 16S diversity, metagenome and transcriptome showed that the low-dose Zn enriched the intestinal microflora and changed the dominant microflora abundances (Proteobacteria, Fusobacteria, Firmicutes and Bacteroidetes), as well as activated the growth hormone metabolism in the liver. Meanwhile, the high-dose of Zn caused the intestinal microbiota dysbiosis, activated the Type VI secretion systems (T6SSs), and further triggered the oxidative stress response, immunity, and antiviral function of the liver. Multi-omics revealed the interference of long-term Zn dietary exposure on the intestine-liver axis. There was an apparent homeostasis of Zn accumulation in the fish tissues, but the window of dietary Zn nutritional requirements versus toxicity appeared to be narrow for the golden pompano. These results provided new insight into the adverse effects and regulatory mechanisms of dietary Zn requirements and toxicity in marine fish.
Collapse
Affiliation(s)
- Shuoli Ma
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| | - Xugang Shu
- School of Chemistry and Chemical Engineering, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
| | - Wen-Xiong Wang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China.
| |
Collapse
|
32
|
Bai F, Hu N, Yang R, Qu LY, Ma S, Huang J, Wang JH, Yang BF, Li CL. Tongmai granules improve rat hippocampal injury by regulating TLR4/MyD88/AP-1 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 285:114874. [PMID: 34838942 DOI: 10.1016/j.jep.2021.114874] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tongmai granules (TMG) is composed of Salvia miltiorrhiza Bge., Radix puerariae Lobata., and Ligusticum chuanxiong hort. TMG is mainly used for ischemic cardiovascular, cerebrovascular diseases, atherosclerosis, coronary heart disease, cerebral infarction and cerebral ischemia. TMG is a kind of traditional compound granule, which has a protective effect on brain injury. However, the potential protective mechanism of the TMG has not been elucidated. AIM OF THE STUDY TMG has a good effect on brain injury, but its brain protective mechanism is still unclear. The purpose of this study was to confirm the neuroprotective mechanism of TMG, reveal its target genes and identify the active components of TMG. MATERIALS AND METHODS High-performance liquid chromatography (HPLC) was used to identify the fingerprint of TMG. UPLC-Q-TOF-MSE was used to analyze the base peak intensity (BPI) chromatograms of TMG. TMG was pre-administered for one week, brain injury and edema were induced by injection of glutamate (Glu) into the lateral ventricles of rats. HE staining was used to investigate the pathological damage caused by Glu in the hippocampus of rats, and the RNA-seq was used to analyze the changes of different genes before and after TMG treatment. Finally, changes of related proteins were analyzed by qRT-PCR, Western blot, and other molecular biological methods. Dosage of TMG were set to 0.6 g/kg, 1.2 g/kg and 2.4 g/kg. RESULTS We found that TMG contained many active components, including salvianolic acid, puerarin, ferulic acid, etc. TMG could improve cerebral edema and brain injury induced by Glu. After TMG treatment, differential gene analysis showed that differential genes were significantly enriched in toll-like receptor signaling pathway. qRT-PCR validation results were consistent with RNA-Seq analysis results. Combined with Western blot analysis, we found that TMG ultimately regulated the expression of inflammatory cytokines by affecting the TLR4/MyD88/AP-1 pathway. CONCLUSIONS In this study, we combined TMG with RNA-seq analysis to demonstrate that TMG may play a neuroprotective role by regulating Toll-like receptor signaling pathway and down-regulating the expression of inflammatory cytokine. TMG may become a kind of traditional Chinese medicine with neuroprotective potential.
Collapse
Affiliation(s)
- Fei Bai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Nan Hu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Ran Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Li-Yuan Qu
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Shuang Ma
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Jian Huang
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Jin-Hui Wang
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Bao-Feng Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China; School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Chun-Li Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
33
|
Zhu Y, Shuai W, Zhao M, Pan X, Pei J, Wu Y, Bu F, Wang A, Ouyang L, Wang G. Unraveling the Design and Discovery of c-Jun N-Terminal Kinase Inhibitors and Their Therapeutic Potential in Human Diseases. J Med Chem 2022; 65:3758-3775. [PMID: 35200035 DOI: 10.1021/acs.jmedchem.1c01947] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
c-Jun N-terminal kinases (JNKs), members of the mitogen-activated protein kinase (MAPK) family, are encoded by three genes: jnk1, jnk2, and jnk3. JNKs are involved in the pathogenesis and development of many diseases, such as neurodegenerative diseases, inflammation, and cancers. Therefore, JNKs have become important therapeutic targets. Many JNK inhibitors have been discovered, and some have been introduced into clinical trials. However, the study of isoform-selective JNK inhibitors is still a challenging task. To further develop novel JNK inhibitors with clinical value, a comprehensive understanding of JNKs and their corresponding inhibitors is required. In this Perspective, we introduced the JNK signaling pathways and reviewed different chemical types of JNK inhibitors, focusing on their structure-activity relationships and biological activities. The challenges and strategies for the development of JNK inhibitors are also discussed. It is hoped that this Perspective will provide valuable references for the development of novel selective JNK inhibitors.
Collapse
Affiliation(s)
- Yumeng Zhu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Wen Shuai
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Min Zhao
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Xiaoli Pan
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Junping Pei
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Yongya Wu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Faqian Bu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Aoxue Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
34
|
Shah K, Al-Haidari A, Sun J, Kazi JU. T cell receptor (TCR) signaling in health and disease. Signal Transduct Target Ther 2021; 6:412. [PMID: 34897277 PMCID: PMC8666445 DOI: 10.1038/s41392-021-00823-w] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Interaction of the T cell receptor (TCR) with an MHC-antigenic peptide complex results in changes at the molecular and cellular levels in T cells. The outside environmental cues are translated into various signal transduction pathways within the cell, which mediate the activation of various genes with the help of specific transcription factors. These signaling networks propagate with the help of various effector enzymes, such as kinases, phosphatases, and phospholipases. Integration of these disparate signal transduction pathways is done with the help of adaptor proteins that are non-enzymatic in function and that serve as a scaffold for various protein-protein interactions. This process aids in connecting the proximal to distal signaling pathways, thereby contributing to the full activation of T cells. This review provides a comprehensive snapshot of the various molecules involved in regulating T cell receptor signaling, covering both enzymes and adaptors, and will discuss their role in human disease.
Collapse
Affiliation(s)
- Kinjal Shah
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Amr Al-Haidari
- Clinical Genetics and Pathology, Skåne University Hospital, Region Skåne, Lund, Sweden
- Clinical Sciences Department, Surgery Research Unit, Lund University, Malmö, Sweden
| | - Jianmin Sun
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Julhash U Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden.
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
35
|
Sasaki S, Negishi T, Tsuzuki T, Yukawa K. Diphenylarsinic acid induced activation of MAP kinases, transcription factors, and oxidative stress-responsive factors and hypersecretion of cytokines in cultured normal human cerebellar astrocytes. Neurotoxicology 2021; 88:196-207. [PMID: 34883095 DOI: 10.1016/j.neuro.2021.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/24/2021] [Accepted: 12/02/2021] [Indexed: 01/09/2023]
Abstract
Diphenylarsinic acid (DPAA) is a non-natural pentavalent organic arsenic and was detected in well water in Kamisu, Ibaraki, Japan in 2003. Individuals that had consumed this arsenic-contaminated water developed cerebellar symptoms such as myoclonus. We previously revealed that DPAA exposure in rats in vitro and in vivo specifically affected astrocytes rather than neurons among cerebellar cells. Here, we evaluated adverse effects of DPAA in cultured normal human cerebellar astrocytes (NHA), which were compared with those in normal rat cerebellar astrocytes (NRA) exposed to DPAA at 10 μM for 96 h, focusing on aberrant activation of astrocytes; increase in cell viability, activation of MAP kinases (ERK1/2, p38MAPK, and SAPK/JNK) and transcription factors (CREB, c-Jun, and c-Fos), upregulation of oxidative stress-responsive factors (Nrf2, HO-1, and Hsp70), and also hypersecretion of brain cytokines (MCP-1, adrenomedullin, FGF-2, CXCL1, and IL-6) as reported in NRA. While DPAA exposure at 10 μM for 96 h had little effect on NHA, a higher concentration (50 μM for 96 h) and longer exposure (10 μM for 288 h) induced similar aberrant activation. Moreover, exposure to DPAA at 50 μM for 96 h or 10 μM for 288 h in NHA induced hypersecretion of cytokines induced in DPAA-exposed NRA (MCP-1, adrenomedullin, FGF-2, CXCL1, and IL-6), and IL-8 besides into culture medium. These results suggested that aberrantly activated human astrocytes by DPAA exposure might play a pivotal role in the pathogenesis of cerebellar symptoms, affecting adjacent neurons, microglia, brain blood vessels, or astrocyte itself through these brain cytokines in human.
Collapse
Affiliation(s)
- Shoto Sasaki
- Department of Physiology, Graduate School of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya-shi, Aichi, 468-8503, Japan
| | - Takayuki Negishi
- Department of Physiology, Graduate School of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya-shi, Aichi, 468-8503, Japan; Department of Physiology, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya-shi, Aichi, 468-8503, Japan.
| | - Takamasa Tsuzuki
- Department of Physiology, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya-shi, Aichi, 468-8503, Japan
| | - Kazunori Yukawa
- Department of Physiology, Graduate School of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya-shi, Aichi, 468-8503, Japan; Department of Physiology, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya-shi, Aichi, 468-8503, Japan
| |
Collapse
|
36
|
Tang Q, Tu B, Jiang X, Zhang J, Bai L, Meng P, Zhang L, Qin X, Wang B, Chen C, Zou Z. Exposure to carbon black nanoparticles during pregnancy aggravates lipopolysaccharide-induced lung injury in offspring: an intergenerational effect. Am J Physiol Lung Cell Mol Physiol 2021; 321:L900-L911. [PMID: 34585979 DOI: 10.1152/ajplung.00545.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Carbon black nanoparticles (CBNPs) are one of the most frequently used nanoparticles. Exposure to CBNPs during pregnancy (PrE to CBNPs) can directly induce inflammation, lung injury, and genotoxicity in dams and results in abnormalities in offspring. However, whether exposure to CBNPs during pregnancy enhances the susceptibility of offspring to environmental stimuli remains unknown. To address this issue, in this study, we intranasally treated pregnant mice with mock or CBNPs from gestational day (GD) 9 to GD18, and F1 and F2 offspring were normally obtained. By intratracheal instillation of mice with lipopolysaccharide (LPS) to trigger a classic animal model for acute lung injury, we intriguingly found that after LPS treatment, F1 and F2 offspring after exposure during pregnancy to CBNPs both exhibited more pronounced lung injury symptoms, including more degenerative histopathological changes, vascular leakage, elevated MPO activity, and activation of inflammation-related signaling transduction, compared with F1 and F2 offspring in the mock group, suggesting PrE to CBNPs would aggravate LPS-induced lung injury in offspring, and this effect was intergenerational. We also observed that PrE to CBNPs upregulated the mRNA expression of DNA methyltransferases (Dnmt) 1/3a/3b and DNA hypermethylation in both F1 and F2 offspring, which might partially account for the intergenerational effect. Together, our study demonstrates for the first time that PrE to CBNPs can enhance sensitivity to LPS in both F1 and F2 offspring, and this intergenerational effect may be related to DNA hypermethylation caused by CBNPs.
Collapse
Affiliation(s)
- Qianghu Tang
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing, People's Republic of China
| | - Baijie Tu
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing, People's Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, People's Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, grid.203458.8Chongqing Medical University, Chongqing, People's Republic of China
| | - Lulu Bai
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing, People's Republic of China
| | - Pan Meng
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing, People's Republic of China
| | - Longbin Zhang
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing, People's Republic of China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Bin Wang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, grid.203458.8Chongqing Medical University, Chongqing, People's Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing, People's Republic of China.,Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, grid.203458.8Chongqing Medical University, Chongqing, People's Republic of China.,Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
37
|
Discovery of a Potent and Selective JNK3 Inhibitor with Neuroprotective Effect Against Amyloid β-Induced Neurotoxicity in Primary Rat Neurons. Int J Mol Sci 2021; 22:ijms222011084. [PMID: 34681742 PMCID: PMC8539420 DOI: 10.3390/ijms222011084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/30/2021] [Accepted: 10/11/2021] [Indexed: 11/28/2022] Open
Abstract
As members of the MAPK family, c-Jun-N-terminal kinases (JNKs) regulate the biological processes of apoptosis. In particular, the isoform JNK3 is expressed explicitly in the brain at high levels and is involved in the pathogenesis of neurodegenerative diseases such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). In this study, we prepared a series of five 6-dihydroxy-1H-benzo[d]imidazoles as JNK3 inhibitors and found them have potential as neuroprotective agents. Following a previous lead scaffold, benzimidazole moiety was modified with various aryl groups and hydroxylation, and the resulting compounds exhibited JNK3 inhibitory activity with improved potency and selectivity. Out of 37 analogues synthesized, (S)-cyclopropyl(3-((4-(2-(2,3-dihydrobenzo[b][1,4]dioxin -6-yl)-5,6-dihydroxy-1H-benzo[d]imidazol-1-yl)pyrimidin-2-yl)amino) piperidin-1-yl)methanone (35b) demonstrated the highest JNK3 inhibition (IC50 = 9.7 nM), as well as neuroprotective effects against Aβ-induced neuronal cell death. As a protein kinase inhibitor, it also showed excellent selectivity over other protein kinases including isoforms JNK1 (>1000 fold) and JNK2 (−10 fold).
Collapse
|
38
|
Shaheryar ZA, Khan MA, Adnan CS, Zaidi AA, Hänggi D, Muhammad S. Neuroinflammatory Triangle Presenting Novel Pharmacological Targets for Ischemic Brain Injury. Front Immunol 2021; 12:748663. [PMID: 34691061 PMCID: PMC8529160 DOI: 10.3389/fimmu.2021.748663] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/15/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke is one of the leading causes of morbidity and mortality globally. Hundreds of clinical trials have proven ineffective in bringing forth a definitive and effective treatment for ischemic stroke, except a myopic class of thrombolytic drugs. That, too, has little to do with treating long-term post-stroke disabilities. These studies proposed diverse options to treat stroke, ranging from neurotropic interpolation to venting antioxidant activity, from blocking specific receptors to obstructing functional capacity of ion channels, and more recently the utilization of neuroprotective substances. However, state of the art knowledge suggests that more pragmatic focus in finding effective therapeutic remedy for stroke might be targeting intricate intracellular signaling pathways of the 'neuroinflammatory triangle': ROS burst, inflammatory cytokines, and BBB disruption. Experimental evidence reviewed here supports the notion that allowing neuroprotective mechanisms to advance, while limiting neuroinflammatory cascades, will help confine post-stroke damage and disabilities.
Collapse
Affiliation(s)
- Zaib A. Shaheryar
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Faculty of Pharmacy, University of Lahore, Lahore, Pakistan
| | - Mahtab A. Khan
- Faculty of Pharmacy, University of Central Punjab, Lahore, Pakistan
| | | | - Awais Ali Zaidi
- Faculty of Pharmacy, University of Lahore, Lahore, Pakistan
- Imran Idrees College of Pharmacy, Lahore, Pakistan
| | - Daniel Hänggi
- Department of Neurosurgery, Faculty of Medicine and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Sajjad Muhammad
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Neurosurgery, Faculty of Medicine and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
39
|
Liakhov SA, Schepetkin IA, Karpenko OS, Duma HI, Haidarzhy NM, Kirpotina LN, Kovrizhina AR, Khlebnikov AI, Bagryanskaya IY, Quinn MT. Novel c-Jun N-Terminal Kinase (JNK) Inhibitors with an 11 H-Indeno[1,2- b]quinoxalin-11-one Scaffold. Molecules 2021; 26:molecules26185688. [PMID: 34577159 PMCID: PMC8464905 DOI: 10.3390/molecules26185688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
c-Jun N-terminal kinase (JNK) plays a central role in stress signaling pathways implicated in important pathological processes, including rheumatoid arthritis and ischemia-reperfusion injury. Therefore, inhibition of JNK is of interest for molecular targeted therapy to treat various diseases. We synthesized 13 derivatives of our reported JNK inhibitor 11H-indeno[1,2-b]quinoxalin-11-one oxime and evaluated their binding to the three JNK isoforms and their biological effects. Eight compounds exhibited submicromolar binding affinity for at least one JNK isoform. Most of these compounds also inhibited lipopolysaccharide (LPS)-induced nuclear factor-κB/activating protein 1 (NF-κB/AP-1) activation and interleukin-6 (IL-6) production in human monocytic THP1-Blue cells and human MonoMac-6 cells, respectively. Selected compounds (4f and 4m) also inhibited LPS-induced c-Jun phosphorylation in MonoMac-6 cells, directly confirming JNK inhibition. We conclude that indenoquinoxaline-based oximes can serve as specific small-molecule modulators for mechanistic studies of JNKs, as well as potential leads for the development of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Serhii A. Liakhov
- A.V. Bogatsky Physico-Chemical Institute, National Academy of Sciences of Ukraine, 65080 Odessa, Ukraine; (S.A.L.); (O.S.K.); (H.I.D.)
| | - Igor A. Schepetkin
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA; (I.A.S.); (L.N.K.)
| | - Olexander S. Karpenko
- A.V. Bogatsky Physico-Chemical Institute, National Academy of Sciences of Ukraine, 65080 Odessa, Ukraine; (S.A.L.); (O.S.K.); (H.I.D.)
| | - Hanna I. Duma
- A.V. Bogatsky Physico-Chemical Institute, National Academy of Sciences of Ukraine, 65080 Odessa, Ukraine; (S.A.L.); (O.S.K.); (H.I.D.)
| | | | - Liliya N. Kirpotina
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA; (I.A.S.); (L.N.K.)
| | - Anastasia R. Kovrizhina
- Kizhner Research Center, Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.R.K.); (A.I.K.)
| | - Andrei I. Khlebnikov
- Kizhner Research Center, Tomsk Polytechnic University, 634050 Tomsk, Russia; (A.R.K.); (A.I.K.)
| | - Irina Y. Bagryanskaya
- Vorozhtsov Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Mark T. Quinn
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA; (I.A.S.); (L.N.K.)
- Correspondence: ; Tel.: +406-994-4707; Fax: +406-994-4303
| |
Collapse
|
40
|
Yu X, Zhou J, Zhao F, Liu X, Mao Y, Diao L, Wen C, Liu M. Tomatidine Suppresses the Destructive Behaviors of Fibroblast-Like Synoviocytes and Ameliorates Type II Collagen-Induced Arthritis in Rats. Front Pharmacol 2021; 12:670707. [PMID: 34512321 PMCID: PMC8426578 DOI: 10.3389/fphar.2021.670707] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/12/2021] [Indexed: 12/29/2022] Open
Abstract
Fibroblast-like synoviocytes (FLSs) are the prominent non-immune cells in synovium and play a pivotal role in rheumatoid arthritis (RA) pathogenesis. Searching for natural compounds that may suppress the pathological phenotypes of FLSs is important for the development of RA treatment. Tomatidine (Td), a steroidal alkaloid derived from the solanaceae family, has been reported to have anti-inflammatory, anti-tumor and immunomodulatory effects. However, its effect on RA remains unknown. Here, we examined the inhibitory effect of Td on TNFα-induced arthritic FLSs, and subsequently investigated its therapeutic effect on collagen-induced arthritis (CIA) rats. Our results revealed that Td significantly inhibited TNFα-induced proliferation and migration of arthritic FLSs. In addition, we found that Td treatment could efficaciously ameliorate synovial inflammation and joint destruction of rats with CIA. Both in vitro and in vivo studies showed that Td significantly suppressed the production of pro-inflammatory cytokines including IL-1β, IL-6 and TNFα, and downregulated the expression of MMP-9 and RANKL. Further molecular mechanism studies revealed that the inhibitory effect of Td on RA might attribute to the decreased activations of MAPKs (ERK and JNK) and NF-κB. These findings provide evidence that Td has the potential to be developed into a complementary or alternative agent for RA therapy.
Collapse
Affiliation(s)
- Xiaolu Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Junnan Zhou
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Fuli Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xuan Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yuhang Mao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Li Diao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chuanjun Wen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Mei Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
41
|
Szegeczki V, Perényi H, Horváth G, Hinnah B, Tamás A, Radák Z, Ábrahám D, Zákány R, Reglodi D, Juhász T. Physical Training Inhibits the Fibrosis Formation in Alzheimer's Disease Kidney Influencing the TGFβ Signaling Pathways. J Alzheimers Dis 2021; 81:1195-1209. [PMID: 33896841 PMCID: PMC8293655 DOI: 10.3233/jad-201206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Alzheimer’s disease (AD) is a neurodegenerative illness, with several peripheral pathological signs such as accumulation of amyloid-β (Aβ) plaques in the kidney. Alterations of transforming growth factor β (TGFβ) signaling in the kidney can induce fibrosis, thus disturbing the elimination of Aβ. Objective: A protective role of increased physical activity has been proven in AD and in kidney fibrosis, but it is not clear whether TGFβ signalization is involved in this effect. Methods: The effects of long-term training on fibrosis were investigated in the kidneys of mice representing a model of AD (B6C3-Tg(APPswe,PSEN1dE9)85Dbo/J) by comparing wild type and AD organs. Alterations of canonical and non-canonical TGFβ signaling pathways were followed with PCR, western blot, and immunohistochemistry. Results: Accumulation of collagen type I and interstitial fibrosis were reduced in kidneys of AD mice after long-term training. AD induced the activation of canonical and non-canonical TGFβ pathways in non-trained mice, while expression levels of signal molecules of both TGFβ pathways became normalized in trained AD mice. Decreased amounts of phosphoproteins with molecular weight corresponding to that of tau and the cleaved C-terminal of AβPP were detected upon exercising, along with a significant increase of PP2A catalytic subunit expression. Conclusion: Our data suggest that physical training has beneficial effects on fibrosis formation in kidneys of AD mice and TGFβ signaling plays a role in this phenomenon.
Collapse
Affiliation(s)
- Vince Szegeczki
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Helga Perényi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gabriella Horváth
- Department of Anatomy, PTE-MTA PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Barbara Hinnah
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andrea Tamás
- Department of Anatomy, PTE-MTA PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Zsolt Radák
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Dóra Ábrahám
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Róza Zákány
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dóra Reglodi
- Department of Anatomy, PTE-MTA PACAP Research Team, University of Pécs Medical School, Pécs, Hungary
| | - Tamás Juhász
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
42
|
Kawasaki S, Ohtsuka H, Sato Y, Douchi D, Sato M, Ariake K, Masuda K, Fukase K, Mizuma M, Nakagawa K, Hayashi H, Morikawa T, Motoi F, Unno M. Silencing of LRRFIP1 enhances the sensitivity of gemcitabine in pancreatic cancer cells by activating JNK/c-Jun signaling. Pancreatology 2021; 21:771-778. [PMID: 33707114 DOI: 10.1016/j.pan.2021.02.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND The epithelial-mesenchymal transition (EMT) in cancer cells has been shown to closely associate with the survival and drug resistance of cancer cells. We recently provided evidence that Wnt signal activator leucine-rich repeat in flightless-1-interacting protein 1 (LRRFIP1) regulates EMT in pancreatic cancer. LRRFIP1 silencing inhibits the translocation of β-catenin to the nucleus, which led to reverse EMT in cancer cells. It was suggested that LRRFIP1 was implicated in gemcitabine sensitivity by regulating EMT signaling. METHODS Gemcitabine chemosensitivity was investigated in LRRFIP1-knockdown pancreatic cancer cells (PANC-1 and MIA Paca-2). In addition, the effects of LRRFIP1 knockdown on JNK/SAPK (stress activated-protein kinase) signaling and apoptosis were evaluated. RESULTS LRRFIP1 silencing accelerates gemcitabine-induced caspase activity and cell death in pancreatic cancer cells. It was also revealed that gemcitabine-induced phosphorylation of c-Jun N-terminal kinase (JNK) and c-Jun were increased in LRRFIP1 knockdown cells. The activation of JNK/c-Jun in LRRFIP1-knockdown cells was significantly diminished by the inhibition of Rac activity. It was confirmed that the acquisition of gemcitabine sensitivity by LRRFIP1 silencing largely depends on the stimulation of JNK/SAPK (stress activated-protein kinase) signaling. CONCLUSIONS Our findings suggest that reversing EMT and transient activation of JNK might be essential for the gemcitabine sensitivity in LRRFIP1 knockdown pancreatic cancer cells. Our discoveries highlight the potential role of LRRFIP1 in the chemosensitivity related to the regulation of EMT signaling.
Collapse
Affiliation(s)
- Shuhei Kawasaki
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Hideo Ohtsuka
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan.
| | - Yoshihiro Sato
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Daisuke Douchi
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Masaki Sato
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Kyohei Ariake
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Kunihiro Masuda
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Koji Fukase
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Masamichi Mizuma
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Kei Nakagawa
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Hiroki Hayashi
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Takanori Morikawa
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Fuyuhiko Motoi
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| |
Collapse
|
43
|
Tang Y, Yang G, Li Y, Wang M, Li G, Hu Y. Protective effects of SP600125 on mice infected with H1N1 influenza A virus. Arch Virol 2021; 166:2151-2158. [PMID: 34014386 PMCID: PMC8134817 DOI: 10.1007/s00705-021-05103-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 03/07/2021] [Indexed: 11/30/2022]
Abstract
Influenza A virus (IAV) can cause high morbidity and mortality globally every year. Myriad host kinases and their related signaling pathways are involved in IAV infection, and the important role of the c-Jun N-terminal kinase signaling pathway during infection has been demonstrated. SP600125, an inhibitor of c-Jun N-terminal kinase, was found in our previous study to suppress IAV replication in vitro. In this study, we established a mouse model of H1N1 IAV infection and treated the mice with SP600125 to study its protective effect. The results showed that SP600125 treatment reduced the pulmonary inflammatory response, lung injury, and pulmonary viral load and increased the survival rate of H1N1-infected mice. Our data confirm the crucial role of c-Jun N terminal kinase in H1N1 virus replication and inflammatory responses in vivo. Hence, we speculate that SP600125 has a potential antiviral therapeutic benefit against IAV infection.
Collapse
Affiliation(s)
- Yuling Tang
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Guanghui Yang
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Yuxiang Li
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Ming Wang
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Gebin Li
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China.
| | - Yanxin Hu
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China.
| |
Collapse
|
44
|
Tong J, Zhang Y, Yu P, Liu J, Mei X, Meng J. Protective Effect of Hydrogen Gas on Mouse Hind Limb Ischemia-Reperfusion Injury. J Surg Res 2021; 266:148-159. [PMID: 33992001 DOI: 10.1016/j.jss.2021.03.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 02/14/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND The aim of this study was to investigate the mechanism of hydrogen gas on hind limb IR injury. METHODS Male C57BL/6 mice were randomly divided into three groups: sham group (Sham), ischemia-reperfusion group (IR), IR plus H2 inhalation group (IR + H2). IR was induced by interrupting hind limb blood flow for 3h, followed by 4h of reperfusion, and H2 was administered by inhalation throughout the reperfusion process. Our data show that H2 inhalation could significantly decrease the infarct-affected tissue volume (P < 0.05), attenuate the degree of morphological injury (P < 0.05), and suppress the level of oxidative stress damage (P < 0.05), compared with the IR group. In exploring the underlying mechanisms, we found that hydrogen could markedly mitigate the degree of IR-induced ER stress and apoptosis (P < 0.05). Additionally, hydrogen could markedly inhibit the IR injury by modulating the phosphorylated c-Jun N-terminal kinase (JNK) signaling pathway (P < 0.05). CONCLUSIONS Taken together, these results revealed the protective effect of hydrogen gas on hind limb ischemia reperfusion injury on mice by attenuating oxidative stress, impairing ER stress and apoptosis, and its ability to modulate JNK signaling pathway.
Collapse
Affiliation(s)
- Jian Tong
- Deportment of Orthopedic, Taizhou People's Hospital, NO.366 TaiHu Road, Taizhou, 225300, Jiangsu Province, China
| | - Yu Zhang
- Department of Orthopedic, Jinling Hospital, the first School of Clinical Medicine, Southern Medical University, Nanjing, 210000, Jiangsu Province, China
| | - Pan Yu
- Department of Burn and Plastic Surgery, East Region Military Command General Hospital, School of Medicine, Nanjing University, Nanjing, 210000, Jiangsu Province, China
| | - Jie Liu
- Deportment of Orthopedic, Taizhou People's Hospital, NO.366 TaiHu Road, Taizhou, 225300, Jiangsu Province, China
| | - XiaoLiang Mei
- Deportment of Orthopedic, Taizhou People's Hospital, NO.366 TaiHu Road, Taizhou, 225300, Jiangsu Province, China
| | - Jia Meng
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, China.
| |
Collapse
|
45
|
Anti-inflammatory Effects of Alcohol Are Associated with JNK-STAT3 Downregulation in an In Vitro Inflammation Model in HepG2 Cells. DISEASE MARKERS 2021; 2021:6622701. [PMID: 33791043 PMCID: PMC7997757 DOI: 10.1155/2021/6622701] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/18/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022]
Abstract
Background In several preclinical and in vitro models of acute inflammation, alcohol (ethanol, EtOH) has been described as an immunomodulatory agent. Similarly, in different pathologies, clinical observations have confirmed either pro- or anti-inflammatory effects of EtOH. The liver plays an important role in immunity and alcohol metabolism; therefore, we analysed dose- and time-dependent effects of EtOH on the inflammatory response of human liver cells in an in vitro model of acute inflammation. Methods HepG2 cells were stimulated with IL-1β and subsequently exposed to EtOH in a low or high dose (85 mM, LoD or 170 mM, HiD) for 1 h (acute exposure) or 72 h (prolonged exposure). IL-6 and TNF-α release was determined by ELISA. Cell viability, adhesion of isolated neutrophils to HepG2 monolayers, their ICAM-1 expression, and the activation of stress-induced protein kinase/c-Jun N-terminal kinase (SAPK/JNK) or signal transducer and activator of transcription 3 (STAT3) were analysed. Results In this experimental design, EtOH did not markedly change the cell viability. Acute and prolonged exposure to EtOH significantly reduced dose-independent IL-1β-induced IL-6 and TNF-α release, as well as adhesion capacity to pretreated HepG2 cells. Acute exposure to EtOH significantly decreased the percentage of ICAM-1-expressing cells. IL-1β stimulation notably increased the activation of SAPK/JNK. However, low-dose EtOH exposure reduced this activation considerably, in contradiction to high-dose EtOH exposure. Acute exposure to LoD EtOH significantly diminished the IL-1β-induced STAT3 activation, whereas an acute exposure of cells to either HiD EtOH or in a prolonged setting showed no effects on STAT3 activation. Conclusion EtOH exerts anti-inflammatory potential in this in vitro model of hepatic inflammation. These effects are associated with the reduced activation of JNK/STAT3 by EtOH, particularly in the condition of acute exposure to low-dose EtOH.
Collapse
|
46
|
JNK signaling as a target for anticancer therapy. Pharmacol Rep 2021; 73:405-434. [PMID: 33710509 DOI: 10.1007/s43440-021-00238-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/30/2021] [Accepted: 02/15/2021] [Indexed: 12/15/2022]
Abstract
The JNKs are members of mitogen-activated protein kinases (MAPK) which regulate many physiological processes including inflammatory responses, macrophages, cell proliferation, differentiation, survival, and death. It is increasingly clear that the continuous activation of JNKs has a role in cancer development and progression. Therefore, JNKs represent attractive oncogenic targets for cancer therapy using small molecule kinase inhibitors. Studies showed that the two major JNK proteins JNK1 and JNK2 have opposite functions in different types of cancers, which need more specification in the design of JNK inhibitors. Some of ATP- competitive and ATP non-competitive inhibitors have been developed and widely used in vitro, but this type of inhibitors lack selectivity and inhibits phosphorylation of all JNK substrates and may lead to cellular toxicity. In this review, we summarized and discussed the strategies of JNK binding inhibitors and the role of JNK signaling in the pathogenesis of different solid and hematological malignancies.
Collapse
|
47
|
Lee HS, Jeong GS. Therapeutic effect of kaempferol on atopic dermatitis by attenuation of T cell activity via interaction with multidrug resistance-associated protein 1. Br J Pharmacol 2021; 178:1772-1788. [PMID: 33555623 DOI: 10.1111/bph.15396] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/28/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Kaempferol is a natural flavonoid widely investigated in various fields due to its antioxidant, anti-cancer, and anti-inflammatory activities, but few studies have shown its inhibitory effect on T cell activation. This study examined the therapeutic potential of kaempferol in atopic dermatitis by modulating T cell activation. EXPERIMENTAL APPROACH Effects of kaempferol on T cell activation and the underlying mechanisms were investigated in Jurkat cells and mouse CD4+ T cells. A model of atopic dermatitis in mice was used to determine its therapeutic potential on T cell-mediated conditions in vivo. Western blots, RT-PCR, pulldown assays and ELISA were used, along with histological analysis of skin. KEY RESULTS Pretreatment with kaempferol reduced CD69 expression and production of inflammatory cytokines including IL-2 from activated Jurkat cells and murine CD4+ T cells without cytotoxicity. Pulldown assays revealed that kaempferol physically binds to MRP-1 in T cells, inhibiting the action of MRP-1. In activated T cells, kaempferol suppressed JNK phosphorylation and the TAK1-IKKα mediated NF-κB pathway. Oral administration of kaempferol to mice showed improved manifestation of atopic dermatitis, a T cell-mediated condition. Western blot results showed that, as in the in vitro studies, decreased phosphorylation of JNK was associated with down-regulated MRP-1 activity in vivo, in the kaempferol-treated mice in the atopic dermatitis model. CONCLUSION AND IMPLICATIONS Kaempferol regulates T cell activation by inhibiting MRP-1 activity in activated T cells, thus showing protective effects against T cell mediated disease in vivo.
Collapse
Affiliation(s)
- Hyun-Su Lee
- College of Pharmacy, Keimyung University, Daegu, Korea
| | | |
Collapse
|
48
|
Wang Y, Tang N, Mao M, Zhou Y, Wu Y, Li J, Zhang W, Peng C, Chen X, Li J. Fine particulate matter (PM2.5) promotes IgE-mediated mast cell activation through ROS/Gadd45b/JNK axis. J Dermatol Sci 2021; 102:47-57. [PMID: 33676788 DOI: 10.1016/j.jdermsci.2021.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/08/2021] [Accepted: 02/14/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Mast cells play an important role in allergic responses and persistently exposure to environmental fine particulate matter (PM2.5) exacerbates allergic diseases,but the details remained elucidative. OBJECTIVES To investigate the effect of PM2.5 on IgE-mediated mast cell responses through an IgE-mediated mouse model and mast cell activation. METHODS The β-hexosaminidase release and a BALB/c model of passive cutaneous anaphylaxis (PCA) was used to test IgE-mediated mast cells activation in vitro and in vivo. RNA-Seq technique was conducted to study the gene expression profile. Reactive oxygen species (ROS) production was measured by flow-cytometry. RT-PCR,WB and ELISA were performed to examine targeting molecules expression. RESULTS PM2.5 facilitated IgE-mediated degranulation and increased cytokines expression in mast cells. Meanwhile, the Evan's blue extravasation as well as serum cytokines in mice was increased after treatment with PM2.5. Furthermore, PM2.5 treatment dramatically increased the expression of Gadd45b which is an oxidative stress molecule that directly activates down-stream pathway, such as MEKK4/JNK. PM2.5 treatment activated MEKK4, JNK1/2 but not ERK1/2 and p38. Meanwhile, Knockdown of Gadd45b significantly attenuated PM2.5-mediated JNK1/2 activation and expression of cytokines. In addition, a JNK1/2-specific inhibitor SP600125 blocked IgE-mediated mast cell activation and cytokine release in PCA model mice. Moreover, PM2.5 treatment increased the ROS level and ROS inhibitor dramatically blocked the PM2.5-induced ROS production and reversed the PM2.5-mediated gene expression in the mitochondrial respiratory chain. CONCLUSIONS PM2.5 regulates ROS production through Gadd45b/MEKK4/JNK pathway, facilitating IgE-mediated mast cell activation.
Collapse
Affiliation(s)
- Ying Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Ni Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Manyun Mao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Youyou Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Yingfang Wu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Juan Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Wei Zhang
- Institute of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Jie Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China; Hunan Engineering Research Center of Skin Health and Disease, Changsha, China.
| |
Collapse
|
49
|
Ludwig S, Hrincius ER, Boergeling Y. The Two Sides of the Same Coin-Influenza Virus and Intracellular Signal Transduction. Cold Spring Harb Perspect Med 2021; 11:a038513. [PMID: 31871235 PMCID: PMC7778220 DOI: 10.1101/cshperspect.a038513] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cells respond to extracellular agents by activation of intracellular signaling pathways. Viruses can be regarded as such agents, leading to a firework of signaling inside the cell, primarily induced by pathogen-associated molecular patterns (PAMPs) that provoke safeguard mechanisms to defend from the invader. In the constant arms race between pathogen and cellular defense, viruses not only have evolved mechanisms to suppress or misuse supposedly antiviral signaling processes for their own benefit but also actively induce signaling to promote replication. This creates viral dependencies that may be exploited for novel strategies of antiviral intervention. Here, we will summarize the current knowledge of activation and function of influenza virus-induced signaling pathways with a focus on nuclear factor (NF)-κB signaling, mitogen-activated protein kinase cascades, and the phosphatidylinositol-3-kinase pathway. We will discuss the opportunities and drawbacks of targeting these signaling pathways for antiviral intervention.
Collapse
Affiliation(s)
- Stephan Ludwig
- Institute of Virology Muenster, University of Muenster, 48149 Muenster, Germany
| | - Eike R Hrincius
- Institute of Virology Muenster, University of Muenster, 48149 Muenster, Germany
| | - Yvonne Boergeling
- Institute of Virology Muenster, University of Muenster, 48149 Muenster, Germany
| |
Collapse
|
50
|
Tauffenberger A, Magistretti PJ. Reactive Oxygen Species: Beyond Their Reactive Behavior. Neurochem Res 2021; 46:77-87. [PMID: 33439432 PMCID: PMC7829243 DOI: 10.1007/s11064-020-03208-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 11/02/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022]
Abstract
Cellular homeostasis plays a critical role in how an organism will develop and age. Disruption of this fragile equilibrium is often associated with health degradation and ultimately, death. Reactive oxygen species (ROS) have been closely associated with health decline and neurological disorders, such as Alzheimer's disease or Parkinson's disease. ROS were first identified as by-products of the cellular activity, mainly mitochondrial respiration, and their high reactivity is linked to a disruption of macromolecules such as proteins, lipids and DNA. More recent research suggests more complex function of ROS, reaching far beyond the cellular dysfunction. ROS are active actors in most of the signaling cascades involved in cell development, proliferation and survival, constituting important second messengers. In the brain, their impact on neurons and astrocytes has been associated with synaptic plasticity and neuron survival. This review provides an overview of ROS function in cell signaling in the context of aging and degeneration in the brain and guarding the fragile balance between health and disease.
Collapse
Affiliation(s)
- Arnaud Tauffenberger
- King Abdullah University of Science and Technology, Thuwal, 23955, Kingdom of Saudi Arabia.
| | - Pierre J Magistretti
- King Abdullah University of Science and Technology, Thuwal, 23955, Kingdom of Saudi Arabia.
| |
Collapse
|