1
|
Xiao Y, Wang J, Sun P, Ding T, Li J, Deng Y. Formation and resuscitation of viable but non-culturable (VBNC) yeast in the food industry: A review. Int J Food Microbiol 2025; 426:110901. [PMID: 39243533 DOI: 10.1016/j.ijfoodmicro.2024.110901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The viable but non-culturable (VBNC) state is a survival strategy adopted by microorganisms in response to unfavorable conditions in the environment. VBNC cells are unable to form colonies but still maintain a low level of activity, posing a potential threat to food safety and public health. Therefore, the development of effective strategies to prevent the formation and resuscitation of VBNC cells of microorganisms is a key challenge in food science and microbiology research. However, current research on VBNC cells has primarily focused on bacteria, with relatively limited reports on fungi. This paper provides a comprehensive and systematic review of yeast in the VBNC state, discussing various factors that induce and facilitate resuscitation, along with detection methods and formation and recovery mechanisms. A comprehensive understanding of the induction and resuscitation of yeast in the VBNC state and exploration of its molecular mechanism hold significant implications for food safety and public health. It is imperative to enhance our comprehension of the underlying mechanisms and contributory factors pertaining to VBNC yeast, thereby facilitating the efficient management of the food fermentation process and ensuring the integrity of food quality and safety.
Collapse
Affiliation(s)
- Yang Xiao
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; School of Food Engineering, Qingdao Institute of Technology, Qingdao 266300, China
| | - Jiayang Wang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Key Laboratory of Special Food Processing (Co-construction by Ministry and Province), Ministry of Agriculture Rural Affairs, Qingdao Agricultural University, Qingdao 266109, China; Shandong Technology Innovation Center of Special Food, Qingdao 266109, China
| | - Pengdong Sun
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Key Laboratory of Special Food Processing (Co-construction by Ministry and Province), Ministry of Agriculture Rural Affairs, Qingdao Agricultural University, Qingdao 266109, China; Shandong Technology Innovation Center of Special Food, Qingdao 266109, China; Qingdao Special Food Research Institute, Qingdao 266109, China
| | - Ting Ding
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Key Laboratory of Special Food Processing (Co-construction by Ministry and Province), Ministry of Agriculture Rural Affairs, Qingdao Agricultural University, Qingdao 266109, China; Shandong Technology Innovation Center of Special Food, Qingdao 266109, China; Qingdao Special Food Research Institute, Qingdao 266109, China
| | - Jingyuan Li
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Key Laboratory of Special Food Processing (Co-construction by Ministry and Province), Ministry of Agriculture Rural Affairs, Qingdao Agricultural University, Qingdao 266109, China; Shandong Technology Innovation Center of Special Food, Qingdao 266109, China; Qingdao Special Food Research Institute, Qingdao 266109, China
| | - Yang Deng
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Key Laboratory of Special Food Processing (Co-construction by Ministry and Province), Ministry of Agriculture Rural Affairs, Qingdao Agricultural University, Qingdao 266109, China; Shandong Technology Innovation Center of Special Food, Qingdao 266109, China; Qingdao Special Food Research Institute, Qingdao 266109, China.
| |
Collapse
|
2
|
Li C, Li S, Zhang G, Li Q, Song W, Wang X, Cook JA, van der Stoel M, Wright BW, Altamirano F, Niewold EL, Han J, Kimble G, Zhang P, Luo X, Urra H, May HI, Ferdous A, Sun XN, Deng Y, Ikonen E, Hetz C, Kaufman RJ, Zhang K, Gillette TG, Scherer PE, Hill JA, Chen J, Wang ZV. IRE1α Mediates the Hypertrophic Growth of Cardiomyocytes Through Facilitating the Formation of Initiation Complex to Promote the Translation of TOP-Motif Transcripts. Circulation 2024; 150:1010-1029. [PMID: 38836349 PMCID: PMC11427172 DOI: 10.1161/circulationaha.123.067606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 05/14/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Cardiomyocyte growth is coupled with active protein synthesis, which is one of the basic biological processes in living cells. However, it is unclear whether the unfolded protein response transducers and effectors directly take part in the control of protein synthesis. The connection between critical functions of the unfolded protein response in cellular physiology and requirements of multiple processes for cell growth prompted us to investigate the role of the unfolded protein response in cell growth and underlying molecular mechanisms. METHODS Cardiomyocyte-specific inositol-requiring enzyme 1α (IRE1α) knockout and overexpression mouse models were generated to explore its function in vivo. Neonatal rat ventricular myocytes were isolated and cultured to evaluate the role of IRE1α in cardiomyocyte growth in vitro. Mass spectrometry was conducted to identify novel interacting proteins of IRE1α. Ribosome sequencing and polysome profiling were performed to determine the molecular basis for the function of IRE1α in translational control. RESULTS We show that IRE1α is required for cell growth in neonatal rat ventricular myocytes under prohypertrophy treatment and in HEK293 cells in response to serum stimulation. At the molecular level, IRE1α directly interacts with eIF4G and eIF3, 2 critical components of the translation initiation complex. We demonstrate that IRE1α facilitates the formation of the translation initiation complex around the endoplasmic reticulum and preferentially initiates the translation of transcripts with 5' terminal oligopyrimidine motifs. We then reveal that IRE1α plays an important role in determining the selectivity and translation of these transcripts. We next show that IRE1α stimulates the translation of epidermal growth factor receptor through an unannotated terminal oligopyrimidine motif in its 5' untranslated region. We further demonstrate a physiological role of IRE1α-governed protein translation by showing that IRE1α is essential for cardiomyocyte growth and cardiac functional maintenance under hemodynamic stress in vivo. CONCLUSIONS These studies suggest a noncanonical, essential role of IRE1α in orchestrating protein synthesis, which may have important implications in cardiac hypertrophy in response to pressure overload and general cell growth under other physiological and pathological conditions.
Collapse
Affiliation(s)
- Chao Li
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shiqian Li
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
- Minerva Foundation Institute for Medical Research, Helsinki 00290, Finland
| | - Guangyu Zhang
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Qinfeng Li
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Weidan Song
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaoding Wang
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jane A. Cook
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Miesje van der Stoel
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
- Minerva Foundation Institute for Medical Research, Helsinki 00290, Finland
| | - Bradley W. Wright
- Laboratory of Functional Genomics and Translational Control, Cecil H. and Ida Green Center for Reproductive Biology Sciences, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, the University of Texas Southwestern Medical Center, TX 75390, USA
| | - Francisco Altamirano
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Erica L. Niewold
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jungsoo Han
- Department of Molecular Biology, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Garrett Kimble
- Department of Molecular Biology, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Pengfei Zhang
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Xiang Luo
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hery Urra
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Bellavista, Santiago, Chile
| | - Herman I. May
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anwarul Ferdous
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xue-Nan Sun
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yingfeng Deng
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Elina Ikonen
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
- Minerva Foundation Institute for Medical Research, Helsinki 00290, Finland
| | - Claudio Hetz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago 8380453, Chile
| | - Randal J. Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Thomas G. Gillette
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph A. Hill
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Molecular Biology, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jin Chen
- Laboratory of Functional Genomics and Translational Control, Cecil H. and Ida Green Center for Reproductive Biology Sciences, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, the University of Texas Southwestern Medical Center, TX 75390, USA
| | - Zhao V. Wang
- Division of Cardiology, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
3
|
Moulik S, Karmakar S, Basu A, Ali M, Chatterjee A. Interdependency and differential expression of ERK1 and ERK2 in breast and melanoma cell lines. J Egypt Natl Canc Inst 2024; 36:27. [PMID: 39278984 DOI: 10.1186/s43046-024-00233-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/18/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Regulatory mechanism of ERK1 and ERK2, their mechanisms of action, and how they impact on development, growth, and homeostasis of different organisms have been given much emphasis for long. ERK1 and 2 though are isoforms of ERK mitogen-activated protein kinase but are coded by two different genes MAPK3 and MAPK1 respectively and show differential expressions and interdependency in different cancer cell lines. Our previous investigations substantially stated the effect of ERK1 and ERK2 on different extracellular molecules like MMPs and integrins, responsible for cell growth and differentiation. Here, we aim to study individual roles of ERK1 and ERK2 and their interdependency in progression and invasiveness in various cancer cell lines. METHODS Different cancer cell lines namely B16F10 (melanoma), MCF7, and MDAMB231 (breast cancer) for studying this particular question were used. Methodologies like gelatin zymography, immunoprecipitation, Western blotting, cell invasion assay, wound healing assay, siRNA transfection, and double transfection procedures were followed for our study. RESULTS Our findings suggest compensation for ERK2 deficiency by pERK1, clear ERK2 predominance in MCF7 cell line, ERK1-ERK2 interdependency in MDAMB231 cells with regard to compensating each other, and significant role of both ERK1 and ERK2 in modulation of MMP9. CONCLUSION If summarized, our results prove the contribution of ERK2 in compensating ERK1 loss and vice versa and an evident role of ERK1 in cancer cell invasiveness.
Collapse
Affiliation(s)
- Shuvojit Moulik
- Research and Development Wing, Suraksha Diagnostics Pvt. Ltd., Newtown, Kolkata, West Bengal, India.
| | - Sayantani Karmakar
- Research and Development Wing, Suraksha Diagnostics Pvt. Ltd., Newtown, Kolkata, West Bengal, India
| | - Asmita Basu
- CWF Labs: Transdisciplinary Healthcare & Research, Bolpur, West Bengal, India
| | - Mahammad Ali
- Department of Analytical Chemistry, Jadavpur University, Kolkata, West Bengal, India
| | - Amitava Chatterjee
- Chittaranjan National Cancer Institute (CNCI), Kolkata, West Bengal, India
| |
Collapse
|
4
|
Hossain MA. Targeting the RAS upstream and downstream signaling pathway for cancer treatment. Eur J Pharmacol 2024; 979:176727. [PMID: 38866361 DOI: 10.1016/j.ejphar.2024.176727] [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: 03/08/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Cancer often involves the overactivation of RAS/RAF/MEK/ERK (MAPK) and PI3K-Akt-mTOR pathways due to mutations in genes like RAS, RAF, PTEN, and PIK3CA. Various strategies are employed to address the overactivation of these pathways, among which targeted therapy emerges as a promising approach. Directly targeting specific proteins, leads to encouraging results in cancer treatment. For instance, RTK inhibitors such as imatinib and afatinib selectively target these receptors, hindering ligand binding and reducing signaling initiation. These inhibitors have shown potent efficacy against Non-Small Cell Lung Cancer. Other inhibitors, like lonafarnib targeting Farnesyltransferase and GGTI 2418 targeting geranylgeranyl Transferase, disrupt post-translational modifications of proteins. Additionally, inhibition of proteins like SOS, SH2 domain, and Ras demonstrate promising anti-tumor activity both in vivo and in vitro. Targeting downstream components with RAF inhibitors such as vemurafenib, dabrafenib, and sorafenib, along with MEK inhibitors like trametinib and binimetinib, has shown promising outcomes in treating cancers with BRAF-V600E mutations, including myeloma, colorectal, and thyroid cancers. Furthermore, inhibitors of PI3K (e.g., apitolisib, copanlisib), AKT (e.g., ipatasertib, perifosine), and mTOR (e.g., sirolimus, temsirolimus) exhibit promising efficacy against various cancers such as Invasive Breast Cancer, Lymphoma, Neoplasms, and Hematological malignancies. This review offers an overview of small molecule inhibitors targeting specific proteins within the RAS upstream and downstream signaling pathways in cancer.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
5
|
Sirayapiwat P, Amorim CA, Sereepapong W, Tuntiviriyapun P, Suebthawinkul C, Thuwanut P. Application of fibrin-based biomaterial for human ovarian tissue encapsulation and cryopreservation as alternative approach for fertility preservation. Cryobiology 2024; 117:104955. [PMID: 39236797 DOI: 10.1016/j.cryobiol.2024.104955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024]
Abstract
This study aimed to investigate the effects of fibrin-based hydrogel encapsulation, with or without vascular endothelial growth factor (VEGF), on follicle quality and cell survival signaling pathways after ovarian tissue cryopreservation. Ovarian cortex donated by seven patients (ages 44-47 years old) was divided into four groups: I) fresh control, II) ovarian tissue without encapsulation (non-FT), III) fibrin (10 mg/mL fibrinogen plus 50 IU/mL thrombin; 10FT) encapsulated tissue without VEGF, and IV) encapsulated tissue with 0.1 μg/mL VEGF (10FT-VEGF), followed by a slow freezing process. Evaluation criteria included normal follicle morphology, density, cell proliferation, apoptosis, and metabolism signaling pathways (BAX/BCL-2 ratio, CASPASE-3 and 9, ATP-6 genes, VEGF-A, and ERK-1/2 protein expression levels). Major outcomes revealed that the percentages of morphologically normal follicles and density were significantly decreased by cryopreservation. Ovarian tissue encapsulation using the 10FT formulation (with or without VEGF) could maintain the ERK-signaling cascade, which was comparable to the fresh control. Among the frozen-thawed cohorts, the BAX/BCL-2 ratio, CASPASE-3, CASPASE-9, and ATP-6 expression levels were unfavorable in the non-FT group. However, statistically different results, including VEGF-A expression levels, were not detected. Collectively, our present data demonstrated the first applicable biomaterial matrix for human ovarian tissue encapsulation which might create an optimal intra-ovarian cortex environment during cryopreservation. Further studies to optimize hydrogel polymerization should be expanded, given the potential benefits for cancer patients who wish to preserve fertility through ovarian tissue cryopreservation.
Collapse
Affiliation(s)
- Porntip Sirayapiwat
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de La Reproduction (REPR), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Wisan Sereepapong
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Punkavee Tuntiviriyapun
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chanakarn Suebthawinkul
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Paweena Thuwanut
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
6
|
Hajimohammadebrahim-Ketabforoush M, Zali A, Shahmohammadi M, Hamidieh AA. Metformin and its potential influence on cell fate decision between apoptosis and senescence in cancer, with a special emphasis on glioblastoma. Front Oncol 2024; 14:1455492. [PMID: 39267853 PMCID: PMC11390356 DOI: 10.3389/fonc.2024.1455492] [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: 06/26/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Despite reaching enormous achievements in therapeutic approaches worldwide, GBM still remains the most incurable malignancy among various cancers. It emphasizes the necessity of adjuvant therapies from the perspectives of both patients and healthcare providers. Therefore, most emerging studies have focused on various complementary and adjuvant therapies. Among them, metabolic therapy has received special attention, and metformin has been considered as a treatment in various types of cancer, including GBM. It is clearly evident that reaching efficient approaches without a comprehensive evaluation of the key mechanisms is not possible. Among the studied mechanisms, one of the more challenging ones is the effect of metformin on apoptosis and senescence. Moreover, metformin is well known as an insulin sensitizer. However, if insulin signaling is facilitated in the tumor microenvironment, it may result in tumor growth. Therefore, to partially resolve some paradoxical issues, we conducted a narrative review of related studies to address the following questions as comprehensively as possible: 1) Does the improvement of cellular insulin function resulting from metformin have detrimental or beneficial effects on GBM cells? 2) If these effects are detrimental to GBM cells, which is more important: apoptosis or senescence? 3) What determines the cellular decision between apoptosis and senescence?
Collapse
Affiliation(s)
- Melika Hajimohammadebrahim-Ketabforoush
- Student Research Committee, Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Shahmohammadi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Pedroza-Torres A, Romero-Córdoba SL, Montaño S, Peralta-Zaragoza O, Vélez-Uriza DE, Arriaga-Canon C, Guajardo-Barreto X, Bautista-Sánchez D, Sosa-León R, Hernández-González O, Díaz-Chávez J, Alvarez-Gómez RM, Herrera LA. Radio-miRs: a comprehensive view of radioresistance-related microRNAs. Genetics 2024; 227:iyae097. [PMID: 38963803 PMCID: PMC11304977 DOI: 10.1093/genetics/iyae097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/29/2024] [Indexed: 07/06/2024] Open
Abstract
Radiotherapy is a key treatment option for a wide variety of human tumors, employed either alone or alongside with other therapeutic interventions. Radiotherapy uses high-energy particles to destroy tumor cells, blocking their ability to divide and proliferate. The effectiveness of radiotherapy is due to genetic and epigenetic factors that determine how tumor cells respond to ionizing radiation. These factors contribute to the establishment of resistance to radiotherapy, which increases the risk of poor clinical prognosis of patients. Although the mechanisms by which tumor cells induce radioresistance are unclear, evidence points out several contributing factors including the overexpression of DNA repair systems, increased levels of reactive oxygen species, alterations in the tumor microenvironment, and enrichment of cancer stem cell populations. In this context, dysregulation of microRNAs or miRNAs, critical regulators of gene expression, may influence how tumors respond to radiation. There is increasing evidence that miRNAs may act as sensitizers or enhancers of radioresistance, regulating key processes such as the DNA damage response and the cell death signaling pathway. Furthermore, expression and activity of miRNAs have shown informative value in overcoming radiotherapy and long-term radiotoxicity, revealing their potential as biomarkers. In this review, we will discuss the molecular mechanisms associated with the response to radiotherapy and highlight the central role of miRNAs in regulating the molecular mechanisms responsible for cellular radioresistance. We will also review radio-miRs, radiotherapy-related miRNAs, either as sensitizers or enhancers of radioresistance that hold promise as biomarkers or pharmacological targets to sensitize radioresistant cells.
Collapse
Affiliation(s)
- Abraham Pedroza-Torres
- Programa Investigadoras e Investigadores por México, Consejo Nacional de Humanidades, Ciencias y Tecnologías, Mexico City C.P. 03940, Mexico
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Mexico City C.P. 14080, Mexico
| | - Sandra L Romero-Córdoba
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City C.P. 04510, Mexico
- Departamento de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City C.P. 14080, Mexico
| | - Sarita Montaño
- Laboratorio de Bioinformática, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Sinaloa (FCQB-UAS), Culiacán Rosales, Sinaloa C.P. 80030, Mexico
| | - Oscar Peralta-Zaragoza
- Dirección de Infecciones Crónicas y Cáncer, Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos C.P. 62100, Mexico
| | - Dora Emma Vélez-Uriza
- Laboratorio de Traducción y Cáncer, Instituto Nacional de Cancerología, Mexico City C.P. 14080, Mexico
| | - Cristian Arriaga-Canon
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología–Instituto de Investigaciones Biomédicas–Universidad Nacional Autónoma de México (UNAM), Mexico City C.P. 14080, Mexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León C.P. 64710, Mexico
| | - Xiadani Guajardo-Barreto
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología–Instituto de Investigaciones Biomédicas–Universidad Nacional Autónoma de México (UNAM), Mexico City C.P. 14080, Mexico
| | - Diana Bautista-Sánchez
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Rodrigo Sosa-León
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Mexico City C.P. 14080, Mexico
| | - Olivia Hernández-González
- Laboratorio de Microscopia Electrónica, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarraa Ibarra”, Mexico City C.P. 14389, Mexico
| | - José Díaz-Chávez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología–Instituto de Investigaciones Biomédicas–Universidad Nacional Autónoma de México (UNAM), Mexico City C.P. 14080, Mexico
| | - Rosa María Alvarez-Gómez
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Mexico City C.P. 14080, Mexico
| | - Luis A Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología–Instituto de Investigaciones Biomédicas–Universidad Nacional Autónoma de México (UNAM), Mexico City C.P. 14080, Mexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León C.P. 64710, Mexico
| |
Collapse
|
8
|
Hu L, Xiao X, Huang W, Zhou T, Chen W, Zhang C, Ying QL. A novel chemical genetic approach reveals paralog-specific role of ERK1/2 in mouse embryonic stem cell fate control. Front Cell Dev Biol 2024; 12:1415621. [PMID: 39071800 PMCID: PMC11272557 DOI: 10.3389/fcell.2024.1415621] [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: 04/10/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction: Mouse embryonic stem cell (ESC) self-renewal can be maintained through dual inhibition of GSK3 and MEK kinases. MEK has two highly homologous downstream kinases, extracellular signal-regulated kinase 1 and 2 (ERK1/2). However, the exact roles of ERK1/2 in mouse ESC self-renewal and differentiation remain unclear. Methods: We selectively deleted or inhibited ERK1, ERK2, or both using genetic and chemical genetic approaches combined with small molecule inhibitors. The effects of ERK paralog-specific inhibition on mouse ESC self-renewal and differentiation were then assessed. Results: ERK1/2 were found to be dispensable for mouse ESC survival and self-renewal. The inhibition of both ERK paralogs, in conjunction with GSK3 inhibition, was sufficient to maintain mouse ESC self-renewal. In contrast, selective deletion or inhibition of only one ERK paralog did not mimic the effect of MEK inhibition in promoting mouse ESC self-renewal. Regarding ESC differentiation, inhibition of ERK1/2 prevented mesendoderm differentiation. Additionally, selective inhibition of ERK1, but not ERK2, promoted mesendoderm differentiation. Discussion: These findings suggest that ERK1 and ERK2 have both overlapping and distinct roles in regulating ESC self-renewal and differentiation. This study provides new insights into the molecular mechanisms of ERK1/2 in governing ESC maintenance and lineage commitment, potentially informing future strategies for controlling stem cell fate in research and therapeutic applications.
Collapse
Affiliation(s)
- Liang Hu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Xiong Xiao
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Wesley Huang
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Tao Zhou
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Weilu Chen
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Chao Zhang
- Loker Hydrocarbon Research Institute and Department of Chemistry, University of Southern California, Los Angeles, CA, United States
| | - Qi-Long Ying
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
9
|
Alsharoh H, Chiroi P, Isachesku E, Tanasa RA, Pop OL, Pirlog R, Berindan-Neagoe I. Personalizing Therapy Outcomes through Mitogen-Activated Protein Kinase Pathway Inhibition in Non-Small Cell Lung Cancer. Biomedicines 2024; 12:1489. [PMID: 39062063 PMCID: PMC11275062 DOI: 10.3390/biomedicines12071489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/18/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Lung cancer (LC) is a highly invasive malignancy and the leading cause of cancer-related deaths, with non-small cell lung cancer (NSCLC) as its most prevalent histological subtype. Despite all breakthroughs achieved in drug development, the prognosis of NSCLC remains poor. The mitogen-activated protein kinase signaling cascade (MAPKC) is a complex network of interacting molecules that can drive oncogenesis, cancer progression, and drug resistance when dysregulated. Over the past decades, MAPKC components have been used to design MAPKC inhibitors (MAPKCIs), which have shown varying efficacy in treating NSCLC. Thus, recent studies support the potential clinical use of MAPKCIs, especially in combination with other therapeutic approaches. This article provides an overview of the MAPKC and its inhibitors in the clinical management of NSCLC. It addresses the gaps in the current literature on different combinations of selective inhibitors while suggesting two particular therapy approaches to be researched in NSCLC: parallel and aggregate targeting of the MAPKC. This work also provides suggestions that could serve as a potential guideline to aid future research in MAPKCIs to optimize clinical outcomes in NSCLC.
Collapse
Affiliation(s)
- Hasan Alsharoh
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | - Paul Chiroi
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | - Ekaterina Isachesku
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | | | - Ovidiu-Laurean Pop
- Department of Morphology Sciences, University of Oradea, 410087 Oradea, Romania;
| | - Radu Pirlog
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| |
Collapse
|
10
|
Bosso G, Cintra Herpst AC, Laguía O, Adetchessi S, Serrano R, Blasco MA. Differential contribution for ERK1 and ERK2 kinases in BRAF V600E-triggered phenotypes in adult mouse models. Cell Death Differ 2024; 31:804-819. [PMID: 38698060 PMCID: PMC11165013 DOI: 10.1038/s41418-024-01300-x] [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: 09/26/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
The BRAF gene is mutated in a plethora of human cancers. The majority of such molecular lesions result in the expression of a constitutively active BRAF variant (BRAFV600E) which continuously bolsters cell proliferation. Although we recently addressed the early effects triggered by BRAFV600E-activation, the specific contribution of ERK1 and ERK2 in BRAFV600E-driven responses in vivo has never been explored. Here we describe the first murine model suitable for genetically dissecting the ERK1/ERK2 impact in multiple phenotypes induced by ubiquitous BRAFV600E-expression. We unveil that ERK1 is dispensable for BRAFV600E-dependent lifespan shortening and for BRAFV600E-driven tumor growth. We show that BRAFV600E-expression provokes an ERK1-independent lymphocyte depletion which does not rely on p21CIP1-induced cell cycle arrest and is unresponsive to ERK-chemical inhibition. Moreover, we also reveal that ERK1 is dispensable for BRAFV600E-triggered cytotoxicity in lungs and that ERK-chemical inhibition abrogates some of these detrimental effects, such as DNA damage, in Club cells but not in pulmonary lymphocytes. Our data suggest that ERK1/ERK2 contribution to BRAFV600E-driven phenotypes is dynamic and varies dependently on cell type, the biological function, and the level of ERK-pathway activation. Our findings also provide useful insights into the comprehension of BRAFV600E-driven malignancies pathophysiology as well as the consequences in vivo of novel ERK pathway-targeted anti-cancer therapies.
Collapse
Affiliation(s)
- Giuseppe Bosso
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain
| | - Ana Carolina Cintra Herpst
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain
| | - Oscar Laguía
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain
| | - Sarah Adetchessi
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain
| | - Rosa Serrano
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain
| | - Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain.
| |
Collapse
|
11
|
Farder-Gomes CF, Miranda FR, Fernandes KM, Bernardes RC, Sena Bastos DS, Licursi de Oliveira L, Martins GF, Serrão JE. Exposure to low-concentration fipronil impairs survival, behavior, midgut morphology and physiology of Aedes aegypti larvae. CHEMOSPHERE 2024; 358:142240. [PMID: 38705417 DOI: 10.1016/j.chemosphere.2024.142240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/30/2023] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
The Aedes aegypti mosquito is a vector for various arboviruses, including dengue and yellow fever. Insecticides, such as pyrethroids and organophosphates, are widely used to manage and control these insects. However, mosquitoes have developed resistance to these chemicals. Therefore, this study aimed to investigate the effects of the commercial formulation of fipronil (Tuit® Florestal; 80% purity) on the survival, behavior, morphology, and proteins related to signaling pathways of the midgut in A. aegypti larvae under controlled laboratory conditions. Significant reductions in immature survival were observed in all concentrations of fipronil tested. Low insecticide concentration (0.5 ppb) led to decreased locomotor activity in the larvae and caused disorganization of the epithelial tissue in the midgut. Moreover, exposure to the insecticide decreased the activity of detoxifying enzymes such as catalase, superoxide dismutase, and glutathione-S-transferase. On the other hand, the insecticide increased protein oxidation and nitric oxide levels. The detection of LC3, caspase-3, and JNK proteins, related to autophagy and apoptosis, increased after exposure. However, there was a decrease in the positive cells for ERK 1/2. Furthermore, the treatment with fipronil decreased the number of positive cells for the proteins FMRF, Prospero, PH3, Wg, Armadillo, Notch, and Delta, which are related to cell proliferation and differentiation. These findings demonstrate that even at low concentrations, fipronil exerts larvicidal effects on A. aegypti by affecting behavior and enzymatic detoxification, inducing protein oxidation, free radical generation, midgut damage and cell death, and inhibiting cell proliferation and differentiation. Thus, this insecticide may represent a viable alternative for controlling the spread of this vector.
Collapse
Affiliation(s)
| | - Franciane Rosa Miranda
- Department of General Biology, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | - Kenner Morais Fernandes
- Department of General Biology, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | | | - Daniel Silva Sena Bastos
- Department of General Biology, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | | | - Gustavo Ferreira Martins
- Department of General Biology, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | - José Eduardo Serrão
- Department of General Biology, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| |
Collapse
|
12
|
Grogan L, Shapiro P. Progress in the development of ERK1/2 inhibitors for treating cancer and other diseases. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 100:181-207. [PMID: 39034052 DOI: 10.1016/bs.apha.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
The extracellular signal-regulated kinases-1 and 2 (ERK1/2) are ubiquitous regulators of many cellular functions, including proliferation, differentiation, migration, and cell death. ERK1/2 regulate cell functions by phosphorylating a diverse collection of protein substrates consisting of other kinases, transcription factors, structural proteins, and other regulatory proteins. ERK1/2 regulation of cell functions is tightly regulated through the balance between activating phosphorylation by upstream kinases and inactivating dephosphorylation by phosphatases. Disruption of homeostatic ERK1/2 regulation caused by elevated extracellular signals or mutations in upstream regulatory proteins leads to the constitutive activation of ERK1/2 signaling and uncontrolled cell proliferation observed in many types of cancer. Many inhibitors of upstream kinase regulators of ERK1/2 have been developed and are part of targeted therapeutic options to treat a variety of cancers. However, the efficacy of these drugs in providing sustained patient responses is limited by the development of acquired resistance often involving re-activation of ERK1/2. As such, recent drug discovery efforts have focused on the direct targeting of ERK1/2. Several ATP competitive ERK1/2 inhibitors have been identified and are being tested in cancer clinical trials. One drug, Ulixertinib (BVD-523), has received FDA approval for use in the Expanded Access Program for patients with no other therapeutic options. This review provides an update on ERK1/2 inhibitors in clinical trials, their successes and limitations, and new academic drug discovery efforts to modulate ERK1/2 signaling for treating cancer and other diseases.
Collapse
Affiliation(s)
- Lena Grogan
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Paul Shapiro
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States.
| |
Collapse
|
13
|
Cho N, Kontou G, Smalley JL, Bope C, Dengler J, Montrose K, Deeb TZ, Brandon NJ, Yamamoto T, Davies PA, Giamas G, Moss SJ. The brain-specific kinase LMTK3 regulates neuronal excitability by decreasing KCC2-dependent neuronal Cl - extrusion. iScience 2024; 27:109512. [PMID: 38715938 PMCID: PMC11075064 DOI: 10.1016/j.isci.2024.109512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/20/2023] [Accepted: 03/13/2024] [Indexed: 05/13/2024] Open
Abstract
LMTK3 is a brain-specific transmembrane serine/threonine protein kinase that acts as a scaffold for protein phosphatase-1 (PP1). Although LMKT3 has been identified as a risk factor for autism and epilepsy, its physiological significance is unknown. Here, we demonstrate that LMTK3 copurifies and binds to KCC2, a neuron-specific K+/Cl- transporter. KCC2 activity is essential for Cl--mediated hyperpolarizing GABAAR receptor currents, the unitary events that underpin fast synaptic inhibition. LMTK3 acts to promote the association of KCC2 with PP1 to promote the dephosphorylation of S940 within its C-terminal cytoplasmic domain, a process the diminishes KCC2 activity. Accordingly, acute inhibition of LMTK3 increases KCC2 activity dependent upon S940 and increases neuronal Cl- extrusion. Consistent with this, LMTK3 inhibition reduced intrinsic neuronal excitability and the severity of seizure-like events in vitro. Thus, LMTK3 may have profound effects on neuronal excitability as an endogenous modulator of KCC2 activity.
Collapse
Affiliation(s)
- Noell Cho
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Georgina Kontou
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Joshua L. Smalley
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Christopher Bope
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Jacob Dengler
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Kristopher Montrose
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| | - Tarek Z. Deeb
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | | | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| | - Paul A. Davies
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Georgios Giamas
- Department for Biochemistry and Biomedicine, University of Sussex Brighton, Brighton BN1 9RH, UK
| | - Stephen J. Moss
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1 6BT, UK
| |
Collapse
|
14
|
Jigari Asl F, Khordadmehr M, Baradaran B, Baghbani E, Noorolyai S, Rahmani S, Saberivand A. Restoration of miR-451a-5p/miR-34a-5p could suppress the proliferation and migration of human breast cancer cells through Wnt/β- catenin and ERK/P-ERK signaling pathways. ARCHIVES OF RAZI INSTITUTE 2024; 79:367-377. [PMID: 39463723 PMCID: PMC11512187 DOI: 10.32592/ari.2024.79.2.367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2024]
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs with a length of 21-25 nucleotides and play an essential role in the regulation of cancer initiation, development and progression. Breast cancer (BC) is the most commonly detected malignancy in women and one of the leading causes of death worldwide. In this study, the effects of transfection of microRNA-451a-5p and miR-34a-5p (tumor suppressors), individually and in combination on apoptosis, proliferation and migration of breast cancer cells in vitro were investigated. For this study, malignant breast cancer cells (MDA-MB-231) were transfected with the miR-451a-5p and miR-34a-5p mimics. Subsequently cytotoxicity, apoptosis, proliferation, migration protein and gene expression of caspase-3, caspase-8, MMP9, ROCK, vimentin and c-Myc of the cancer cells were analyzed by MTT, flow cytometry, q-RT-PCR (expression level of caspase-3, caspase-8, MMP9, ROCK, vimentin and c-Myc genes), wound healing and Western blot assays. The results showed that miR-34a-5p and miR-451a-5p could additionally induce apoptosis and cell cycle arrest in the sub-G1phase, suppress proliferation and migration in breast cancer cells, and also decrease the expression of β- catenin and ERK/P-ERK proteins . The present data document that restoration of the tumor suppressor miR-451/miR-34 strongly induces programmed cell death in vitro and apparently inhibits cell proliferation and migration in human breast cancer cells. In summary, miR-451a and miR-34a play an important role in breast cancer cell proliferation and migration via the Wnt/β-catenin and ERK/P-ERK signaling pathways. Therefore, the simultaneous restoration of the presented tumor suppressor miRNAs can be proposed as a valuable and potential therapeutic strategy in the treatment of breast cancer. However, further studies should be useful.
Collapse
Affiliation(s)
- F Jigari Asl
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran
| | - M Khordadmehr
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran
| | - B Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran
| | - E Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran
| | - S Noorolyai
- Immunology Research Center, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran
| | - S Rahmani
- Immunology Research Center, Tabriz University of Medical Sciences, 51666-14761, Tabriz, Iran
| | - A Saberivand
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, 51665-1647, Tabriz, Iran
| |
Collapse
|
15
|
Lee HJ, Kwon YS, Lee JH, Moon YG, Choi J, Hyun M, Tak TK, Kim JH, Heo JD. Pectolinarigenin regulates the tumor-associated proteins in AGS-xenograft BALB/c nude mice. Mol Biol Rep 2024; 51:305. [PMID: 38361124 PMCID: PMC10869406 DOI: 10.1007/s11033-023-09046-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 10/30/2023] [Indexed: 02/17/2024]
Abstract
BACKGROUND Pectolinarigenin (PEC) is a flavone extracted from Cirsium, and because it has anti-inflammatory properties, anti-cancer research is also being conducted. The objective of this work was to find out if PEC is involved in tumor control and which pathways it regulates in vivo and in vitro. METHODS AGS cell lines were xenografted into BALB/c nude mice to create tumors, and PEC was administered intraperitoneally to see if it was involved in tumor control. Once animal testing was completed, tumor proteins were isolated and identified using LC-MS analysis, and gene ontology of the found proteins was performed. RESULTS Body weight and hematological measurements on the xenograft mice model demonstrated that PEC was not harmful to non-cancerous cells. We found 582 proteins in tumor tissue linked to biological reactions such as carcinogenesis and cell death signaling. PEC regulated 6 out of 582 proteins in vivo and in vitro in the same way. CONCLUSION Our findings suggested that PEC therapy may inhibit tumor development in gastric cancer (GC), and proteomic research gives fundamental information about proteins that may have great promise as new therapeutic targets in GC.
Collapse
Affiliation(s)
- Ho Jeong Lee
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Young Sang Kwon
- Environmental Safety Assessment Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Ju Hong Lee
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Yeon Gyu Moon
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Jungil Choi
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Moonjung Hyun
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Tae Kil Tak
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Je-Hein Kim
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea
| | - Jeong Doo Heo
- Gyeongnam Bio-Health Research Support Center, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju, 52834, Republic of Korea.
| |
Collapse
|
16
|
Saleem S. Targeting MAPK signaling: A promising approach for treating inflammatory lung disease. Pathol Res Pract 2024; 254:155122. [PMID: 38246034 DOI: 10.1016/j.prp.2024.155122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024]
Abstract
The extracellular signals that initiate intracellular reactions are dispatched by the mitogen-activated protein kinases (MAPKs), which oversee a multitude of cellular activities. p38, Extracellular signal-regulated kinase (ERK), and c-Jun NH2-terminal kinase (JNK) are members of the vertebrate family of MAPKs, and each MAPK signaling pathway consists of a MAPK kinase (MAP3K), a MAPK kinase (MAP2K), and a MAPK. These signaling pathways orchestrate numerous cellular processes, including cell growth, survival, differentiation, and apoptosis. The emergence of various inflammatory respiratory diseases in humans has been linked to the dysregulation of MAPK signaling pathways. Conditions such as asthma, lung cancer, pulmonary fibrosis, and COPD are among the prevalent respiratory ailments where MAPK plays a pivotal role. Additionally, MAPK is implicated in infectious diseases, including COVID-19, pneumonia, and tuberculosis. COPD, asthma, emphysema, chronic bronchitis, and other inflammatory lung disorders highlight the significance of MAPK as a potential target for therapeutic development. Further studies are needed to delve into the molecular mechanisms by which the MAPK signaling pathway contributes to inflammatory lung disorders, representing an area that demands continued research.
Collapse
Affiliation(s)
- Shakir Saleem
- Department of Public Health, College of Health Sciences, Saudi Electronic University, P.O. Box 93499, Riyadh 11673, Saudi Arabia.
| |
Collapse
|
17
|
Patel AS, Ludwinski FE, Mondragon A, Nuthall K, Saha P, Lyons O, Squadrito ML, Siow R, De Palma M, Smith A, Modarai B. HTATIP2 regulates arteriogenic activity in monocytes from patients with limb ischemia. JCI Insight 2023; 8:e131419. [PMID: 37847559 PMCID: PMC10807724 DOI: 10.1172/jci.insight.131419] [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: 07/01/2019] [Accepted: 10/12/2023] [Indexed: 10/18/2023] Open
Abstract
Use of autologous cells isolated from elderly patients with multiple comorbidities may account for the modest efficacy of cell therapy in patients with chronic limb threatening ischemia (CLTI). We aimed to determine whether proarteriogenic monocyte/macrophages (Mo/MΦs) from patients with CLTI were functionally impaired and to demonstrate the mechanisms related to any impairment. Proarteriogenic Mo/MΦs isolated from patients with CLTI were found to have an impaired capacity to promote neovascularization in vitro and in vivo compared with those isolated from healthy controls. This was associated with increased expression of human HIV-1 TAT interactive protein-2 (HTATIP2), a transcription factor known to suppress angiogenesis/arteriogenesis. Silencing HTATIP2 restored the functional capacity of CLTI Mo/MΦs, which was associated with increased expression of arteriogenic regulators Neuropilin-1 and Angiopoietin-1, and their ability to enhance angiogenic (endothelial tubule formation) and arteriogenic (smooth muscle proliferation) processes in vitro. In support of the translational relevance of our findings, silencing HTATIP2 in proarteriogenic Mo/MΦs isolated from patients with CLTI rescued their capacity to enhance limb perfusion in the ischemic hindlimb by effecting greater angiogenesis and arteriogenesis. Ex vivo modulation of HTATIP2 may offer a strategy for rescuing the functional impairment of pro-angio/arteriogenic Mo/MΦs prior to autologous delivery and increase the likelihood of clinical efficacy.
Collapse
Affiliation(s)
- Ashish S. Patel
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, King’s BHF Centre of Research Excellence, King’s College London, United Kingdom
| | - Francesca E. Ludwinski
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, King’s BHF Centre of Research Excellence, King’s College London, United Kingdom
| | - Angeles Mondragon
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, King’s BHF Centre of Research Excellence, King’s College London, United Kingdom
| | - Katherine Nuthall
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, King’s BHF Centre of Research Excellence, King’s College London, United Kingdom
| | - Prakash Saha
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, King’s BHF Centre of Research Excellence, King’s College London, United Kingdom
| | - Oliver Lyons
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, King’s BHF Centre of Research Excellence, King’s College London, United Kingdom
| | - Mario Leonardo Squadrito
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Richard Siow
- Department of Vascular Biology and Inflammation, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, King’s BHF Centre of Research Excellence, King’s College London, United Kingdom
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alberto Smith
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, King’s BHF Centre of Research Excellence, King’s College London, United Kingdom
| | - Bijan Modarai
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, King’s BHF Centre of Research Excellence, King’s College London, United Kingdom
| |
Collapse
|
18
|
Bunch H, Kim D, Naganuma M, Nakagawa R, Cong A, Jeong J, Ehara H, Vu H, Chang JH, Schellenberg MJ, Sekine SI. ERK2-topoisomerase II regulatory axis is important for gene activation in immediate early genes. Nat Commun 2023; 14:8341. [PMID: 38097570 PMCID: PMC10721843 DOI: 10.1038/s41467-023-44089-y] [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: 09/14/2022] [Accepted: 11/29/2023] [Indexed: 12/17/2023] Open
Abstract
The function of the mitogen-activated protein kinase signaling pathway is required for the activation of immediate early genes (IEGs), including EGR1 and FOS, for cell growth and proliferation. Recent studies have identified topoisomerase II (TOP2) as one of the important regulators of the transcriptional activation of IEGs. However, the mechanism underlying transcriptional regulation involving TOP2 in IEG activation has remained unknown. Here, we demonstrate that ERK2, but not ERK1, is important for IEG transcriptional activation and report a critical ELK1 binding sequence for ERK2 function at the EGR1 gene. Our data indicate that both ERK1 and ERK2 extensively phosphorylate the C-terminal domain of TOP2B at mutual and distinctive residues. Although both ERK1 and ERK2 enhance the catalytic rate of TOP2B required to relax positive DNA supercoiling, ERK2 delays TOP2B catalysis of negative DNA supercoiling. In addition, ERK1 may relax DNA supercoiling by itself. ERK2 catalytic inhibition or knock-down interferes with transcription and deregulates TOP2B in IEGs. Furthermore, we present the first cryo-EM structure of the human cell-purified TOP2B and etoposide together with the EGR1 transcriptional start site (-30 to +20) that has the strongest affinity to TOP2B within -423 to +332. The structure shows TOP2B-mediated breakage and dramatic bending of the DNA. Transcription is activated by etoposide, while it is inhibited by ICRF193 at EGR1 and FOS, suggesting that TOP2B-mediated DNA break to favor transcriptional activation. Taken together, this study suggests that activated ERK2 phosphorylates TOP2B to regulate TOP2-DNA interactions and favor transcriptional activation in IEGs. We propose that TOP2B association, catalysis, and dissociation on its substrate DNA are important processes for regulating transcription and that ERK2-mediated TOP2B phosphorylation may be key for the catalysis and dissociation steps.
Collapse
Affiliation(s)
- Heeyoun Bunch
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
- School of Applied Biosciences, College of Agriculture & Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Deukyeong Kim
- School of Applied Biosciences, College of Agriculture & Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Masahiro Naganuma
- Laboratory for Transcription Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Reiko Nakagawa
- RIKEN BDR Laboratory for Phyloinformatics, Hyogo, 650-0047, Japan
| | - Anh Cong
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jaehyeon Jeong
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Haruhiko Ehara
- Laboratory for Transcription Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Hongha Vu
- Department of Biology Education, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jeong Ho Chang
- Department of Biology Education, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Matthew J Schellenberg
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Shun-Ichi Sekine
- Laboratory for Transcription Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| |
Collapse
|
19
|
Gross J, Herrera-Marschitz M. Potential Key Proteins, Molecular Networks, and Pathways in Perinatal Hypoxia. Neurotox Res 2023; 41:571-588. [PMID: 37651081 DOI: 10.1007/s12640-023-00663-2] [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: 07/06/2022] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023]
Abstract
Perinatal hypoxia is a common risk factor for CNS development. Using bioinformatics databases, a list of 129 genes involved in perinatal hypoxia was selected from the literature and analyzed with respect to proteins important for biological processes influencing the brain development. Functional enrichment analysis using the DAVID database was performed to identify relevant Gene Ontology (GO) biological processes like response to hypoxia, inflammatory response, positive and negative regulation of apoptosis, and positive and negative regulation of cell proliferation. The selected GO processes contain 17-30 proteins and show an enrichment of 6.3-14.3-fold. The STRING protein-protein interaction network and the Cytoscape data analyzer were used to identify interacting proteins playing a significant role in these processes. The two top protein pairs referring to the proteins with highest degrees and the corresponding proteins connected by high score edges exert opposite or regulatory functions and are essential for the balance between damaging, repairing, protective, or epigenetic processes. The GO response to hypoxia is characterized by the high score protein-protein interaction pairs CASP3/FAS promoting apoptosis and by the protective acting BDNF/MECP2 protein pair. Core components of the GO processes positive and negative regulation of apoptosis are the proteins CASP3/FAS/AKT/eNOS/RPS6KB1 involved in several signal pathways. The GO processes cell proliferation are characterized by the high-score protein-protein interaction pairs MYC/ MAPK1, JUN/MAPK1, IL6/IL1B, and JUN/HDAC1. The study provides new insights into the pathophysiology of perinatal hypoxia and is of importance for future investigations, diagnostics, and therapy of perinatal hypoxia.
Collapse
Affiliation(s)
- Johann Gross
- Tinnitus Center, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin and Leibniz Society of Sciences Berlin, 10117, Berlin, Germany.
| | - Mario Herrera-Marschitz
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Santiago 7, Chile
| |
Collapse
|
20
|
Kim DK, Huh JW, Yu H, Lee Y, Jin Y, Ha UH. Pseudomonas aeruginosa-Derived DnaJ Induces the Expression of IL-1β by Engaging the Interplay of p38 and ERK Signaling Pathways in Macrophages. Int J Mol Sci 2023; 24:15957. [PMID: 37958940 PMCID: PMC10648868 DOI: 10.3390/ijms242115957] [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: 09/26/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
As members of pathogen-associated molecular patterns, bacterial heat shock proteins (HSPs) are widely recognized for their role in initiating innate immune responses. This study aimed to examine the impact of DnaJ, a homolog of HSP40 derived from Pseudomonas aeruginosa (P. aeruginosa), on the regulation of IL-1β expression in macrophages. We demonstrated that DnaJ modulates macrophages to secrete IL-1β by activating NF-κB and MAPK signaling pathways. Specifically, ERK was identified as a positive mediator for IL-1β expression, while p38 acted as a negative mediator. These results suggest that the reciprocal actions of these two crucial MAPKs play a vital role in controlling IL-1β expression. Additionally, the reciprocal actions of MAPKs were found to regulate the activation of inflammasome-related molecules, including vimentin, NLRP3, caspase-1, and GSDMD. Furthermore, our investigation explored the involvement of CD91/CD40 in ERK signaling-mediated IL-1β production from DnaJ-treated macrophages. These findings emphasize the importance of understanding the signaling mechanisms underlying IL-1β induction and suggest the potential utility of DnaJ as an adjuvant for stimulating inflammasome activation.
Collapse
Affiliation(s)
- Dae-Kyum Kim
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (D.-K.K.); (J.-W.H.); (H.Y.); (Y.L.)
- Interdisciplinary Graduate Program for Artificial Intelligence Smart Convergence Technology, Korea University, Sejong 30019, Republic of Korea
| | - Jin-Won Huh
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (D.-K.K.); (J.-W.H.); (H.Y.); (Y.L.)
| | - Hyeonseung Yu
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (D.-K.K.); (J.-W.H.); (H.Y.); (Y.L.)
| | - Yeji Lee
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (D.-K.K.); (J.-W.H.); (H.Y.); (Y.L.)
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, Nankai University, Tianjin 300071, China;
| | - Un-Hwan Ha
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (D.-K.K.); (J.-W.H.); (H.Y.); (Y.L.)
- Interdisciplinary Graduate Program for Artificial Intelligence Smart Convergence Technology, Korea University, Sejong 30019, Republic of Korea
| |
Collapse
|
21
|
Petrosino M, Novak L, Pasquo A, Turina P, Capriotti E, Minicozzi V, Consalvi V, Chiaraluce R. The complex impact of cancer-related missense mutations on the stability and on the biophysical and biochemical properties of MAPK1 and MAPK3 somatic variants. Hum Genomics 2023; 17:95. [PMID: 37891694 PMCID: PMC10612357 DOI: 10.1186/s40246-023-00544-x] [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: 07/28/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Mitogen-activated protein kinases 1 and 3 (MAPK1 and MAPK3), also called extracellular regulated kinases (ERK2 and ERK1), are serine/threonine kinase activated downstream by the Ras/Raf/MEK/ERK signal transduction cascade that regulates a variety of cellular processes. A dysregulation of MAPK cascade is frequently associated to missense mutations on its protein components and may be related to many pathologies, including cancer. In this study we selected from COSMIC database a set of MAPK1 and MAPK3 somatic variants found in cancer tissues carrying missense mutations distributed all over the MAPK1 and MAPK3 sequences. The proteins were expressed as pure recombinant proteins, and their biochemical and biophysical properties have been studied in comparison with the wild type. The missense mutations lead to changes in the tertiary arrangements of all the variants. The thermodynamic stability of the wild type and variants has been investigated in the non-phosphorylated and in the phosphorylated form. Significant differences in the thermal stabilities of most of the variants have been observed, as well as changes in the catalytic efficiencies. The energetics of the catalytic reaction is affected for all the variants for both the MAPK proteins. The stability changes and the variation in the enzyme catalysis observed for most of MAPK1/3 variants suggest that a local change in a residue, distant from the catalytic site, may have long-distance effects that reflect globally on enzyme stability and functions.
Collapse
Affiliation(s)
- Maria Petrosino
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Leonore Novak
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alessandra Pasquo
- ENEA CR Frascati, Diagnostics and Metrology Laboratory FSN-TECFIS-DIM, Frascati, Italy
| | - Paola Turina
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Emidio Capriotti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Velia Minicozzi
- Department of Physics, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Valerio Consalvi
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Roberta Chiaraluce
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland.
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
22
|
Martin-Vega A, Cobb MH. Navigating the ERK1/2 MAPK Cascade. Biomolecules 2023; 13:1555. [PMID: 37892237 PMCID: PMC10605237 DOI: 10.3390/biom13101555] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
The RAS-ERK pathway is a fundamental signaling cascade crucial for many biological processes including proliferation, cell cycle control, growth, and survival; common across all cell types. Notably, ERK1/2 are implicated in specific processes in a context-dependent manner as in stem cells and pancreatic β-cells. Alterations in the different components of this cascade result in dysregulation of the effector kinases ERK1/2 which communicate with hundreds of substrates. Aberrant activation of the pathway contributes to a range of disorders, including cancer. This review provides an overview of the structure, activation, regulation, and mutational frequency of the different tiers of the cascade; with a particular focus on ERK1/2. We highlight the importance of scaffold proteins that contribute to kinase localization and coordinate interaction dynamics of the kinases with substrates, activators, and inhibitors. Additionally, we explore innovative therapeutic approaches emphasizing promising avenues in this field.
Collapse
Affiliation(s)
- Ana Martin-Vega
- Department of Pharmacology, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA;
| | - Melanie H. Cobb
- Department of Pharmacology, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA;
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA
| |
Collapse
|
23
|
Zhang Q, Lei X, Wang F, He X, Liu L, Hou Y, Liu Y, Jin F, Chen C, Li B. ERK1-mediated immunomodulation of mesenchymal stem cells ameliorates inflammatory disorders. iScience 2023; 26:107868. [PMID: 37790278 PMCID: PMC10543658 DOI: 10.1016/j.isci.2023.107868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/03/2023] [Accepted: 09/06/2023] [Indexed: 10/05/2023] Open
Abstract
Immune system disorders, especially T cell disorders, are important therapeutic targets of mesenchymal stem cells (MSCs) in many autoimmune diseases (ADs). Although extracellular regulated protein kinases (ERKs) play a role in MSC therapy by promoting T cell apoptosis, the mechanism remains unclear. Our findings indicate that ERK1-/- bone marrow MSCs (BMMSCs), but not ERK2-/- BMMSCs, failed to promote T cell apoptosis due to incapacity to activate the ETS2/AURKA/NF-κB/Fas/MCP-1 cascade. Moreover, ERK1-/- BMMSCs were unable to upregulate regulatory T cells and suppress T helper 17 cells. Licochalcone A (LA), which promotes ERK pathway activation, enhanced the therapeutic efficacy of MSC therapy in ulcerative colitis and collagen-induced arthritis mice. Our findings suggest that ERK1, but not ERK2, plays a crucial role in regulating T cells in MSCs. LA-treated MSCs provide a strategy to improve the efficacy of MSC-based treatments for ADs.
Collapse
Affiliation(s)
- Qing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Xiao Lei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Fang Wang
- Department of Blood Purification, General Hospital of Central Theater Command of PLA, 68 Huangpu Road, Wuhan, Hubei 430010, China
| | - Xiaoning He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Lu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yuxia Hou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Yuan Liu
- The Affiliated Northwest Women’s and Children’s Hospital of Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi 710061, China
| | - Fang Jin
- Shannxi Clinical Research Center for Oral Diseases & Department of Orthodontics, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, the Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| |
Collapse
|
24
|
Kim C. Extracellular Signal-Regulated Kinases Play Essential but Contrasting Roles in Osteoclast Differentiation. Int J Mol Sci 2023; 24:15342. [PMID: 37895023 PMCID: PMC10607827 DOI: 10.3390/ijms242015342] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Bone homeostasis is regulated by the balanced actions of osteoblasts that form the bone and osteoclasts (OCs) that resorb the bone. Bone-resorbing OCs are differentiated from hematopoietic monocyte/macrophage lineage cells, whereas osteoblasts are derived from mesenchymal progenitors. OC differentiation is induced by two key cytokines, macrophage colony-stimulating factor (M-CSF), a factor essential for the proliferation and survival of the OCs, and receptor activator of nuclear factor kappa-B ligand (RANKL), a factor for responsible for the differentiation of the OCs. Mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinases (ERKs), p38, and c-Jun N-terminal kinases, play an essential role in regulating the proliferation, differentiation, and function of OCs. ERKs have been known to play a critical role in the differentiation and activation of OCs. In most cases, ERKs positively regulate OC differentiation and function. However, several reports present conflicting conclusions. Interestingly, the inhibition of OC differentiation by ERK1/2 is observed only in OCs differentiated from RAW 264.7 cells. Therefore, in this review, we summarize the current understanding of the conflicting actions of ERK1/2 in OC differentiation.
Collapse
Affiliation(s)
- Chaekyun Kim
- BK21 Program in Biomedical Science & Engineering, Laboratory for Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
25
|
Azizi SA, Qiu T, Brookes NE, Dickinson BC. Regulation of ERK2 activity by dynamic S-acylation. Cell Rep 2023; 42:113135. [PMID: 37715953 PMCID: PMC10591828 DOI: 10.1016/j.celrep.2023.113135] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/28/2023] [Accepted: 08/30/2023] [Indexed: 09/18/2023] Open
Abstract
Extracellular signal-regulated kinases (ERK1/2) are key effector proteins of the mitogen-activated protein kinase pathway, choreographing essential processes of cellular physiology. Here, we discover that ERK1/2 are subject to S-acylation, a reversible lipid modification of cysteine residues, at C271/C254. The levels of ERK1/2 S-acylation are modulated by epidermal growth factor (EGF) signaling, mirroring its phosphorylation dynamics, and acylation-deficient ERK2 displays altered phosphorylation patterns. We show that ERK1/2 S-acylation is mediated by "writer" protein acyl transferases (PATs) and "eraser" acyl protein thioesterases (APTs) and that chemical inhibition of either lipid addition or removal alters ERK1/2's EGF-triggered transcriptional program. Finally, in a mouse model of metabolic syndrome, we find that ERK1/2 lipidation levels correlate with alterations in ERK1/2 lipidation writer/eraser expression, solidifying a link between ERK1/2 activity, ERK1/2 lipidation, and organismal health. This study describes how lipidation regulates ERK1/2 and offers insight into the role of dynamic S-acylation in cell signaling more broadly.
Collapse
Affiliation(s)
- Saara-Anne Azizi
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Medical Scientist Training Program, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Tian Qiu
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Noah E Brookes
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Bryan C Dickinson
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
26
|
Tserunyan V, Finley SD. A systems and computational biology perspective on advancing CAR therapy. Semin Cancer Biol 2023; 94:34-49. [PMID: 37263529 PMCID: PMC10529846 DOI: 10.1016/j.semcancer.2023.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 04/24/2023] [Accepted: 05/28/2023] [Indexed: 06/03/2023]
Abstract
In the recent decades, chimeric antigen receptor (CAR) therapy signaled a new revolutionary approach to cancer treatment. This method seeks to engineer immune cells expressing an artificially designed receptor, which would endue those cells with the ability to recognize and eliminate tumor cells. While some CAR therapies received FDA approval and others are subject to clinical trials, many aspects of their workings remain elusive. Techniques of systems and computational biology have been frequently employed to explain the operating principles of CAR therapy and suggest further design improvements. In this review, we sought to provide a comprehensive account of those efforts. Specifically, we discuss various computational models of CAR therapy ranging in scale from organismal to molecular. Then, we describe the molecular and functional properties of costimulatory domains frequently incorporated in CAR structure. Finally, we describe the signaling cascades by which those costimulatory domains elicit cellular response against the target. We hope that this comprehensive summary of computational and experimental studies will further motivate the use of systems approaches in advancing CAR therapy.
Collapse
Affiliation(s)
- Vardges Tserunyan
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Stacey D Finley
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA; Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
27
|
Buscà R, Onesto C, Egensperger M, Pouysségur J, Pagès G, Lenormand P. N-terminal alanine-rich (NTAR) sequences drive precise start codon selection resulting in elevated translation of multiple proteins including ERK1/2. Nucleic Acids Res 2023; 51:7714-7735. [PMID: 37414542 PMCID: PMC10450180 DOI: 10.1093/nar/gkad528] [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: 09/08/2022] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
We report the discovery of N-terminal alanine-rich sequences, which we term NTARs, that act in concert with their native 5'-untranslated regions to promote selection of the proper start codon. NTARs also facilitate efficient translation initiation while limiting the production of non-functional polypeptides through leaky scanning. We first identified NTARs in the ERK1/2 kinases, which are among the most important signaling molecules in mammals. Analysis of the human proteome reveals that hundreds of proteins possess NTARs, with housekeeping proteins showing a particularly high prevalence. Our data indicate that several of these NTARs act in a manner similar to those found in the ERKs and suggest a mechanism involving some or all of the following features: alanine richness, codon rarity, a repeated amino acid stretch and a nearby second AUG. These features may help slow down the leading ribosome, causing trailing pre-initiation complexes (PICs) to pause near the native AUG, thereby facilitating accurate translation initiation. Amplification of erk genes is frequently observed in cancer, and we show that NTAR-dependent ERK protein levels are a rate-limiting step for signal output. Thus, NTAR-mediated control of translation may reflect a cellular need to precisely control translation of key transcripts such as potential oncogenes. By preventing translation in alternative reading frames, NTAR sequences may be useful in synthetic biology applications, e.g. translation from RNA vaccines.
Collapse
Affiliation(s)
- Roser Buscà
- Université Côte d’Azur (UCA), CNRS UMR 7284 and INSERM U 1081, Institute for Research on Cancer and Aging Nice (IRCAN), 28 Avenue de Valombrose, 06107 Nice, France
- Centre Antoine Lacassagne, Nice, France
| | - Cercina Onesto
- Université Côte d’Azur (UCA), CNRS UMR 7284 and INSERM U 1081, Institute for Research on Cancer and Aging Nice (IRCAN), 28 Avenue de Valombrose, 06107 Nice, France
- Centre Antoine Lacassagne, Nice, France
- Polytech’Nice Sophia, Bioengineering Department, Sophia-Antipolis, France
| | - Mylène Egensperger
- Université Côte d’Azur (UCA), CNRS UMR 7284 and INSERM U 1081, Institute for Research on Cancer and Aging Nice (IRCAN), 28 Avenue de Valombrose, 06107 Nice, France
- Centre Antoine Lacassagne, Nice, France
| | - Jacques Pouysségur
- Université Côte d’Azur (UCA), CNRS UMR 7284 and INSERM U 1081, Institute for Research on Cancer and Aging Nice (IRCAN), 28 Avenue de Valombrose, 06107 Nice, France
- Centre Antoine Lacassagne, Nice, France
- Centre Scientifique de Monaco, Biomedical Department, Principality of Monaco
| | - Gilles Pagès
- Université Côte d’Azur (UCA), CNRS UMR 7284 and INSERM U 1081, Institute for Research on Cancer and Aging Nice (IRCAN), 28 Avenue de Valombrose, 06107 Nice, France
- Centre Antoine Lacassagne, Nice, France
- Centre Scientifique de Monaco, Biomedical Department, Principality of Monaco
| | - Philippe Lenormand
- Université Côte d’Azur (UCA), CNRS UMR 7284 and INSERM U 1081, Institute for Research on Cancer and Aging Nice (IRCAN), 28 Avenue de Valombrose, 06107 Nice, France
- Centre Antoine Lacassagne, Nice, France
| |
Collapse
|
28
|
Bardwell L, Thorner J. Mitogen-activated protein kinase (MAPK) cascades-A yeast perspective. Enzymes 2023; 54:137-170. [PMID: 37945169 DOI: 10.1016/bs.enz.2023.07.001] [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] [Indexed: 11/12/2023]
Abstract
Discovery of the class of protein kinase now dubbed a mitogen (or messenger)-activated protein kinase (MAPK) is an illustrative example of how disparate lines of investigation can converge and reveal an enzyme family universally conserved among eukaryotes, from single-celled microbes to humans. Moreover, elucidation of the circuitry controlling MAPK function defined a now overarching principle in enzyme regulation-the concept of an activation cascade mediated by sequential phosphorylation events. Particularly ground-breaking for this field of exploration were the contributions of genetic approaches conducted using several model organisms, but especially the budding yeast Saccharomyces cerevisiae. Notably, examination of how haploid yeast cells respond to their secreted peptide mating pheromones was crucial in pinpointing genes encoding MAPKs and their upstream activators. Fully contemporaneous biochemical analysis of the activities elicited upon stimulation of mammalian cells by insulin and other growth- and differentiation-inducing factors lead eventually to the demonstration that components homologous to those in yeast were involved. Continued studies of these pathways in yeast were integral to other foundational discoveries in MAPK signaling, including the roles of tethering, scaffolding and docking interactions.
Collapse
Affiliation(s)
- Lee Bardwell
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| | - Jeremy Thorner
- Division of Biochemistry, Biophysics and Structural Biology, Department of Molecular and Cell Biology, College of Letters and Science, University of California, Berkeley, Berkeley, CA, United States.
| |
Collapse
|
29
|
Romano A, Freudenthal R, Feld M. Molecular insights from the crab Neohelice memory model. Front Mol Neurosci 2023; 16:1214061. [PMID: 37415833 PMCID: PMC10321408 DOI: 10.3389/fnmol.2023.1214061] [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: 04/28/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023] Open
Abstract
Memory acquisition, formation and maintenance depend on synaptic post-translational machinery and regulation of gene expression triggered by several transduction pathways. In turns, these processes lead to stabilization of synaptic modifications in neurons in the activated circuits. In order to study the molecular mechanisms involved in acquisition and memory, we have taken advantage of the context-signal associative learning and, more recently, the place preference task, of the crab Neohelice granulata. In this model organism, we studied several molecular processes, including activation of extracellular signal-regulated kinase (ERK) and the nuclear factor kappa light chain enhancer of activated B cells (NF-κB) transcription factor, involvement of synaptic proteins such as NMDA receptors and neuroepigenetic regulation of gene expression. All these studies allowed description of key plasticity mechanisms involved in memory, including consolidation, reconsolidation and extinction. This article is aimed at review the most salient findings obtained over decades of research in this memory model.
Collapse
Affiliation(s)
- Arturo Romano
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología, Biología Molecular y Celular “Dr. Hector Maldonado” (FBMC), Buenos Aires, Argentina
- Biología Molecular y Neurociencias (IFIBYNE), Instituto de Fisiología, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Ramiro Freudenthal
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología, Biología Molecular y Celular “Dr. Hector Maldonado” (FBMC), Buenos Aires, Argentina
- Biotecnología y Biología Traslacional (IB3), Facultad de Ciencias Exactas y Naturales, Instituto de Biociencias, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariana Feld
- Biología Molecular y Neurociencias (IFIBYNE), Instituto de Fisiología, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| |
Collapse
|
30
|
Berek K, Bauer A, Rudzki D, Auer M, Barket R, Zinganell A, Lerch M, Hofer L, Grams A, Poskaite P, Wurth S, Berger T, Di Pauli F, Deisenhammer F, Hegen H, Reindl M. Immune profiling in multiple sclerosis: a single-center study of 65 cytokines, chemokines, and related molecules in cerebrospinal fluid and serum. Front Immunol 2023; 14:1200146. [PMID: 37383229 PMCID: PMC10294231 DOI: 10.3389/fimmu.2023.1200146] [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: 04/04/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction The understanding of the pathophysiology of multiple sclerosis (MS) has evolved alongside the characterization of cytokines and chemokines in cerebrospinal fluid (CSF) and serum. However, the complex interplay of pro- and anti-inflammatory cytokines and chemokines in different body fluids in people with MS (pwMS) and their association with disease progression is still not well understood and needs further investigation. Therefore, the aim of this study was to profile a total of 65 cytokines, chemokines, and related molecules in paired serum and CSF samples of pwMS at disease onset. Methods Multiplex bead-based assays were performed and baseline routine laboratory diagnostics, magnetic resonance imaging (MRI), and clinical characteristics were assessed. Of 44 participants included, 40 had a relapsing-remitting disease course and four a primary progressive MS. Results There were 29 cytokines and chemokines that were significantly higher in CSF and 15 in serum. Statistically significant associations with moderate effect sizes were found for 34 of 65 analytes with sex, age, CSF, and MRI parameters and disease progression. Discussion In conclusion, this study provides data on the distribution of 65 different cytokines, chemokines, and related molecules in CSF and serum in newly diagnosed pwMS.
Collapse
Affiliation(s)
- Klaus Berek
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Angelika Bauer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- VASCage Research Centre on Vascular Ageing and Stroke, Innsbruck, Austria
| | - Dagmar Rudzki
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- VASCage Research Centre on Vascular Ageing and Stroke, Innsbruck, Austria
| | - Michael Auer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Robert Barket
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anne Zinganell
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Lerch
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Livia Hofer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Astrid Grams
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Paulina Poskaite
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Wurth
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Franziska Di Pauli
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Harald Hegen
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Reindl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
31
|
Jang S, Yun C, Kim B, Kang S, Lee J, Jeong S, Cho Y, Kim SH, Lee CM, Moon C, Kim JS. Immunohistochemical analysis of extracellular signal-regulated kinase expression in mature and immature bulls' testes and epididymides. VET MED-CZECH 2023; 68:231-237. [PMID: 37982001 PMCID: PMC10581511 DOI: 10.17221/34/2023-vetmed] [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: 03/18/2023] [Accepted: 06/13/2023] [Indexed: 11/21/2023] Open
Abstract
Extracellular signal-regulated kinase (ERK) has been implicated in mammalian testicular and epididymal development. This study aimed to investigate ERK expression in the immature and mature testes and epididymides of bulls. We evaluated ERK expression using immunoblot analysis and immunohistochemistry. Immunoblot analysis revealed that immature bull testes and epididymides had higher phosphorylated ERK (pERK) expression than mature bull testes and epididymides. pERK immunoreactivity was higher in immature epididymides than in immature testes. pERK was localised mostly in spermatogonia, undifferentiated sustentacular (Sertoli) cells, and interstitial (Leydig) cells in immature testes, as well as in some spermatocytes and spermatids in mature testes. In immature epididymides, the body and tail had higher pERK expression than the head, whereas pERK was broadly distributed throughout the stereocilia, basal cells, and connective tissues. pERK distribution in the head of mature epididymides was similar to that in immature epididymides, whereas few connective tissue cells were expressed in the body and tail of mature epididymides. Collectively, these results suggest that ERK is expressed in the testis and epididymis of immature and mature bulls with varying intensities, and the role of ERK in male reproductive organs may include the specific function of its development.
Collapse
Affiliation(s)
- Sungwoong Jang
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
- Sungwoong Jang and Changjin Yun contributed equally to this work
| | - Changjin Yun
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
- Sungwoong Jang and Changjin Yun contributed equally to this work
| | - Bohye Kim
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
| | - Sohi Kang
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
| | - Jeongmin Lee
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
| | - Sohee Jeong
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
| | - Yongho Cho
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
| | - Sung-Ho Kim
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
| | - Chang-Min Lee
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
| | - Changjong Moon
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
| | - Joong-Sun Kim
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Buk-gu, Gwangju, Republic of Korea
| |
Collapse
|
32
|
Yellon DM, Beikoghli Kalkhoran S, Davidson SM. The RISK pathway leading to mitochondria and cardioprotection: how everything started. Basic Res Cardiol 2023; 118:22. [PMID: 37233787 PMCID: PMC10220132 DOI: 10.1007/s00395-023-00992-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/27/2023]
Abstract
Ischaemic heart disease, which often manifests clinically as myocardial infarction (MI), remains a major cause of mortality worldwide. Despite the development of effective pre-clinical cardioprotective therapies, clinical translation has been disappointing. Nevertheless, the 'reperfusion injury salvage kinase' (RISK) pathway appears to be a promising target for cardioprotection. This pathway is crucial for the induction of cardioprotection by numerous pharmacological and non-pharmacological interventions, such as ischaemic conditioning. An important component of the cardioprotective effects of the RISK pathway involves the prevention of mitochondrial permeability transition pore (MPTP) opening and subsequent cardiac cell death. Here, we will review the historical perspective of the RISK pathway and focus on its interaction with mitochondria in the setting of cardioprotection.
Collapse
Affiliation(s)
- Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
| | | | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| |
Collapse
|
33
|
Ma J, Hu Z, Yue H, Luo Y, Wang C, Wu X, Gu Y, Wang L. GRM2 Regulates Functional Integration of Adult-Born DGCs by Paradoxically Modulating MEK/ERK1/2 Pathway. J Neurosci 2023; 43:2822-2836. [PMID: 36878727 PMCID: PMC10124958 DOI: 10.1523/jneurosci.1886-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/01/2023] [Accepted: 02/19/2023] [Indexed: 03/08/2023] Open
Abstract
Metabotropic glutamate receptor 2 (GRM2) is highly expressed in hippocampal dentate granule cells (DGCs), regulating synaptic transmission and hippocampal functions. Newborn DGCs are continuously generated throughout life and express GRM2 when they are mature. However, it remained unclear whether and how GRM2 regulates the development and integration of these newborn neurons. We discovered that the expression of GRM2 in adult-born DGCs increased with neuronal development in mice of both sexes. Lack of GRM2 caused developmental defects of DGCs and impaired hippocampus-dependent cognitive functions. Intriguingly, our data showed that knockdown of Grm2 resulted in decreased b/c-Raf kinases and paradoxically led to an excessive activation of MEK/ERK1/2 pathway. Inhibition of MEK ameliorated the developmental defects caused by Grm2 knockdown. Together, our results indicate that GRM2 is necessary for the development and functional integration of newborn DGCs in the adult hippocampus through regulating the phosphorylation and activation state of MEK/ERK1/2 pathway.SIGNIFICANCE STATEMENT Metabotropic glutamate receptor 2 (GRM2) is highly expressed in mature dentate granule cells (DGCs) in the hippocampus. It remains unclear whether GRM2 is required for the development and integration of adult-born DGCs. We provided in vivo and in vitro evidence to show that GRM2 regulates the development of adult-born DGCs and their integration into existing hippocampal circuits. Lack of GRM2 in a cohort of newborn DGCs impaired object-to-location memory in mice. Moreover, we revealed that GRM2 knockdown paradoxically upregulated MEK/ERK1/2 pathway by suppressing b/c-Raf in developing neurons, which is likely a common mechanism underlying the regulation of the development of neurons expressing GRM2. Thus, Raf/MEK/ERK1/2 pathway could be a potential target for brain diseases related to GRM2 abnormality.
Collapse
Affiliation(s)
- Jiao Ma
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, 310027 Hangzhou, People's Republic of China
- Center of Stem Cell and Regenerative Medicine, and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058 Hangzhou, People's Republic of China
| | - Zhechun Hu
- School of Brain Science and Brain Medicine, Zhejiang University, 310058 Hangzhou, People's Republic of China
| | - Huimin Yue
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, 310027 Hangzhou, People's Republic of China
- School of Brain Science and Brain Medicine, Zhejiang University, 310058 Hangzhou, People's Republic of China
| | - Yujian Luo
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, 310027 Hangzhou, People's Republic of China
- School of Brain Science and Brain Medicine, Zhejiang University, 310058 Hangzhou, People's Republic of China
| | - Chao Wang
- School of Brain Science and Brain Medicine, Zhejiang University, 310058 Hangzhou, People's Republic of China
| | - Xuan Wu
- Center of Stem Cell and Regenerative Medicine, and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058 Hangzhou, People's Republic of China
| | - Yan Gu
- Center of Stem Cell and Regenerative Medicine, and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058 Hangzhou, People's Republic of China
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University, 310058 Hangzhou, People's Republic of China
| | - Lang Wang
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, 310027 Hangzhou, People's Republic of China
- School of Brain Science and Brain Medicine, Zhejiang University, 310058 Hangzhou, People's Republic of China
| |
Collapse
|
34
|
Jeruzalska E, Mazur AJ. The Role of non-muscle actin paralogs in cell cycle progression and proliferation. Eur J Cell Biol 2023; 102:151315. [PMID: 37099935 DOI: 10.1016/j.ejcb.2023.151315] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023] Open
Abstract
Uncontrolled cell proliferation leads to several pathologies, including cancer. Thus, this process must be tightly regulated. The cell cycle accounts for cell proliferation, and its progression is coordinated with changes in cell shape, for which cytoskeleton reorganization is responsible. Rearrangement of the cytoskeleton allows for its participation in the precise division of genetic material and cytokinesis. One of the main cytoskeletal components is filamentous actin-based structures. Mammalian cells have at least six actin paralogs, four of which are muscle-specific, while two, named β- and γ-actin, are abundantly present in all types of cells. This review summarizes the findings that establish the role of non-muscle actin paralogs in regulating cell cycle progression and proliferation. We discuss studies showing that the level of a given non-muscle actin paralog in a cell influences the cell's ability to progress through the cell cycle and, thus, proliferation. Moreover, we elaborate on the non-muscle actins' role in regulating gene transcription, interactions of actin paralogs with proteins involved in controlling cell proliferation, and the contribution of non-muscle actins to different structures in a dividing cell. The data cited in this review show that non-muscle actins regulate the cell cycle and proliferation through varying mechanisms. We point to the need for further studies addressing these mechanisms.
Collapse
Affiliation(s)
- Estera Jeruzalska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Poland
| | - Antonina J Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Poland.
| |
Collapse
|
35
|
Korovina I, Vehlow A, Temme A, Cordes N. Targeting integrin α2 as potential strategy for radiochemosensitization of glioblastoma. Neuro Oncol 2023; 25:648-661. [PMID: 36219689 PMCID: PMC10076950 DOI: 10.1093/neuonc/noac237] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a fast-growing primary brain tumor characterized by high invasiveness and resistance. This results in poor patient survival. Resistance is caused by many factors, including cell-extracellular matrix (ECM) interactions. Here, we addressed the role of adhesion protein integrin α2, which we identified in a high-throughput screen for novel potential targets in GBM cells treated with standard therapy consisting of temozolomide (TMZ) and radiation. METHODS In our study, we used a range of primary/stem-like and established GBM cell models in vitro and in vivo. To identify regulatory mechanisms, we employed high-throughput kinome profiling, Western blotting, immunofluorescence staining, reporter, and activity assays. RESULTS Our data showed that integrin α2 is overexpressed in GBM compared to normal brain and, that its deletion causes radiochemosensitization. Similarly, invasion and adhesion were significantly reduced in TMZ-irradiated GBM cell models. Furthermore, we found that integrin α2-knockdown impairs the proliferation of GBM cells without affecting DNA damage repair. At the mechanistic level, we found that integrin α2 affects the activity of activating transcription factor 1 (ATF1) and modulates the expression of extracellular signal-regulated kinase 1 (ERK1) regulated by extracellular signals. Finally, we demonstrated that integrin α2-deficiency inhibits tumor growth and thereby prolongs the survival of mice with orthotopically growing GBM xenografts. CONCLUSIONS Taken together our data suggest that integrin α2 may be a promising target to overcome GBM resistance to radio- and chemotherapy. Thus, it would be worth evaluating how efficient and safe the adjuvant use of integrin α2 inhibitors is to standard radio(chemo)therapy in GBM.
Collapse
Affiliation(s)
- Irina Korovina
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology–OncoRay, Dresden, Germany
| | - Anne Vehlow
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Achim Temme
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurosurgery, Section Experimental Neurosurgery and Tumor Immunology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nils Cordes
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology–OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
36
|
Kim MJ, Eom YB. Applicable Forensic Biomarker for Drowning Diagnosis: Extracellular Signal-Regulated Kinase 2 (ERK2). Int J Legal Med 2023:10.1007/s00414-023-02992-5. [PMID: 36973587 DOI: 10.1007/s00414-023-02992-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Drowning is a common cause of accidental death worldwide, and it continues to be a serious public health problem. However, diagnosing drowning is a challenging task in forensic investigation because it is difficult to prove actual drowning and other submerged deaths with the autopsy techniques that are currently in use. Here, we show biomarkers that may be helpful for the diagnosis of drowning. We divided the experimental animals into four groups (drowning, postmortem submersion, hypoxia, and control) to evaluate the expression patterns of extracellular signal-regulated kinase 1/2 (ERK1/2). On gene expression analysis, only ERK2 was found to be significantly increased in the drowning groups compared to the other cases. In the immunoblot analysis, phosphorylated ERK2 (p-ERK2) was found to be upregulated in the drowning groups. Immunohistochemical staining also showed that p-ERK in alveolar cells revealed a granular pattern in the drowning groups. However, the expression pattern of ERK2 over time after drowning differed between the freshwater and seawater drowning groups. Taken together, these results indicate that ERK2 may be useful for distinguishing between drowning and postmortem submersion if the postmortem interval (PMI) of drowning is short. Conversely, if the PMI is long from the time that death occurs until the discovery of dead bodies, it is possibly more helpful for identifying between freshwater and seawater drowning.
Collapse
Affiliation(s)
- Min-Jeong Kim
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea
| | - Yong-Bin Eom
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
| |
Collapse
|
37
|
Erdenebaatar P, Gunarta IK, Suzuki R, Odongoo R, Fujii T, Fukunaga R, Kanemaki MT, Yoshioka K. Redundant roles of extra-cellular signal-regulated kinase (ERK) 1 and 2 in the G1-S transition and etoposide-induced G2/M checkpoint in HCT116 cells. Drug Discov Ther 2023; 17:10-17. [PMID: 36642508 DOI: 10.5582/ddt.2022.01120] [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: 01/15/2023]
Abstract
The extracellular signal-regulated kinase (ERK) 1 and 2 intracellular signaling pathways play key roles in a variety of cellular processes, such as proliferation and differentiation. Dysregulation of ERK1/2 signaling has been implicated in many diseases, including cancer. Although ERK1/2 signaling pathways have been extensively studied, controversy remains as to whether ERK1 and ERK2 have specific or redundant functions. In this study, we examined the functional roles of ERK1 and ERK2 in cell proliferation and cell cycle progression using an auxin-inducible degron system combined with gene knockout technology. We found that ERK1/2 double depletion, but not ERK1 or ERK2 depletion, substantially inhibited the proliferation of HCT116 cells during G1-S transition. We further demonstrated that ERK1/2-double-depleted cells were much more tolerant to etoposide-induced G2/M arrest than ERK1 or ERK2 single-knockout cells. Together, these results strongly suggest the functional redundancy of ERK1 and ERK2 in both the G1-S transition under physiological conditions and the DNA damage-induced G2/M checkpoint. Our findings substantially advance understanding of the ERK1/2 pathways, which could have strong implications for future pharmacological developments.
Collapse
Affiliation(s)
- Purev Erdenebaatar
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - I Ketut Gunarta
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Ryusuke Suzuki
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Ravdandorj Odongoo
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Toshihiro Fujii
- Department of Biochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Rikiro Fukunaga
- Department of Biochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Masato T Kanemaki
- Department of Chromosome Science, National Institute of Genetics, Research Organization of Information and Systems (ROIS), Mishima, Japan.,Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Japan
| | - Katsuji Yoshioka
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
38
|
Li W, Ke C, Yang C, Li J, Chen Q, Xia Z, Xu J. LncRNA DICER1-AS1 promotes colorectal cancer progression by activating the MAPK/ERK signaling pathway through sponging miR-650. Cancer Med 2023; 12:8351-8366. [PMID: 36708020 PMCID: PMC10134332 DOI: 10.1002/cam4.5550] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a disease with high morbidity and mortality rates globally. Long noncoding RNAs (lncRNAs) play a fundamental role in tumor progression, and increasing attention has been paid to their role in CRC. This study aimed to determine the function of lncRNA DICER1 antisense RNA 1 (DICER1-AS1) in CRC and confirm its potential regulatory mechanisms in CRC. METHODS The publicly available dataset was used to assess DICER1-AS1 function and expression in CRC. RT-qPCR or western blot assays were performed to verify DICER1-AS1, miR-650, and mitogen-activated protein kinase 1 (MAPK1) expression in CRC cells or tissues. To determine the function of DICER1-AS1, we performed CCK-8, colony formation, transwell, cell cycle, and in vivo animal assays. Using RNA sequence analysis, luciferase reporter assays, and bioinformatics analysis, the connection between DICER1-AS1, MAPK1, and miR-650 was investigated. RESULTS DICER1-AS1 was significantly upregulated in CRC tissue compared to normal colon tissue. High DICER1-AS1 expression suggested a poor prognosis in CRC patients. Functionally, upregulation of DICER1-AS1 effectively promoted CRC proliferation, migration, and invasion ex vivo and tumor progression in vivo. Mechanistically, DICER1-AS1 functions as a competitive endogenous RNA (ceRNA) that sponges miR-650 to upregulate MAPK1, promotes ERK1/2 phosphorylation, and sequentially activates the MAPK/ERK signaling pathway. CONCLUSION Our investigations found that upregulation of DICER1-AS1 activates the MAPK/ERK signaling pathway by sponging miR-650 to promote CRC progression, revealing a possible clinically significant biomarker and therapeutic target.
Collapse
Affiliation(s)
- Wenfei Li
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Chuanfeng Ke
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cuiyan Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jieyao Li
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qikui Chen
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhongsheng Xia
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jihao Xu
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
39
|
Yoshioka S, Ikeda T, Fukuchi S, Kawai Y, Ohta K, Murakami H, Ogo N, Muraoka D, Takikawa O, Asai A. Identification and Characterization of a Novel Dual Inhibitor of
Indoleamine 2,3-dioxygenase 1 and Tryptophan 2,3-dioxygenase. Int J Tryptophan Res 2022; 15:11786469221138456. [PMCID: PMC9716449 DOI: 10.1177/11786469221138456] [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: 07/07/2022] [Accepted: 10/21/2022] [Indexed: 12/05/2022] Open
Abstract
Kynurenine (Kyn), a metabolite of tryptophan (Trp), is a key regulator of mammal
immune responses such as cancer immune tolerance. Indoleamine-2,3-dioxygenase
(IDO) and tryptophan-2,3-dioxygenase (TDO) are main enzymes regulating the first
and rate-limiting step of the Kyn pathway. To identify new small molecule
inhibitors of TDO, we selected A172 glioblastoma cell line constitutively
expressed TDO. Characterization of this cell line using kinase inhibitor library
resulted in identification of MEK/ERK pathway-dependent TDO expression. After
knowing the properties for TDO expression, we further proceeded to screen
chemical library for TDO inhibitors. We previously determined that
S-benzylisothiourea derivatives are enzymatic inhibitors of indoleamine
2,3-dioxygenase 1 (IDO1) and suggested that the isothiourea moiety could be an
important pharmacophore for binding to heme. Based on this premise, we screened
an in-house library composed of various isothiourea derivatives and identified a
bisisothiourea derivative, PVZB3001, as an inhibitor of TDO. Interestingly,
PVZB3001 also inhibited the enzymatic activity of IDO1 in both cell-based and
cell-free assays but did not inhibit other heme enzymes. Molecular docking
studies suggested the importance of isothiourea moieties at the ortho position
of the phenyl ring for the inhibition of catalytic activity. PVZB3001 showed
competitive inhibition against TDO, and this was supported by the docking
simulation. PVZB3001 recovered natural killer (NK) cell viability and functions
by inhibiting Kyn accumulation in conditioned medium of both IDO1- and
TDO-expressing cells. Furthermore, oral administration of IDO1-overexpressing
tumor-bearing mice with PVZB3001 significantly inhibited tumor growth. Thus, we
identified a novel selective dual inhibitor of IDO1 and TDO using the Kyn
production assay with a glioblastoma cell line. This inhibitor could be a useful
pharmacological tool for modulating the Kyn pathway in a variety of experimental
systems.
Collapse
Affiliation(s)
- Saeko Yoshioka
- Center for Drug Discovery, Graduate
School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Tomonori Ikeda
- Center for Drug Discovery, Graduate
School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Sogo Fukuchi
- Center for Drug Discovery, Graduate
School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yurika Kawai
- Center for Drug Discovery, Graduate
School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Katsumi Ohta
- Center for Drug Discovery, Graduate
School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Hisashi Murakami
- Center for Drug Discovery, Graduate
School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Naohisa Ogo
- Center for Drug Discovery, Graduate
School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Daisuke Muraoka
- Department of Oncology, Nagasaki
University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Osamu Takikawa
- National Institute for Longevity
Sciences, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Akira Asai
- Center for Drug Discovery, Graduate
School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan,Akira Asai, Graduate School of
Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka
422-8526, Japan.
| |
Collapse
|
40
|
From Nucleus to Organs: Insights of Aryl Hydrocarbon Receptor Molecular Mechanisms. Int J Mol Sci 2022; 23:ijms232314919. [PMID: 36499247 PMCID: PMC9738205 DOI: 10.3390/ijms232314919] [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: 10/25/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a markedly established regulator of a plethora of cellular and molecular processes. Its initial role in the detoxification of xenobiotic compounds has been partially overshadowed by its involvement in homeostatic and organ physiology processes. In fact, the discovery of its ability to bind specific target regulatory sequences has allowed for the understanding of how AHR modulates such processes. Thereby, AHR presents functions in transcriptional regulation, chromatin architecture modifications and participation in different key signaling pathways. Interestingly, such fields of influence end up affecting organ and tissue homeostasis, including regenerative response both to endogenous and exogenous stimuli. Therefore, from classical spheres such as canonical transcriptional regulation in embryonic development, cell migration, differentiation or tumor progression to modern approaches in epigenetics, senescence, immune system or microbiome, this review covers all aspects derived from the balance between regulation/deregulation of AHR and its physio-pathological consequences.
Collapse
|
41
|
Casella M, Lori G, Coppola L, La Rocca C, Tait S. BDE-47, -99, -209 and Their Ternary Mixture Disrupt Glucose and Lipid Metabolism of Hepg2 Cells at Dietary Relevant Concentrations: Mechanistic Insight through Integrated Transcriptomics and Proteomics Analysis. Int J Mol Sci 2022; 23:ijms232214465. [PMID: 36430946 PMCID: PMC9697228 DOI: 10.3390/ijms232214465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are persistent organic chemicals implied as flame retardants. Humans are mainly exposed to BDE-47, -99, and -209 congeners by diet. PBDEs are metabolic disruptors with the liver as the main target organ. To investigate their mode of action at a human-relevant concentration, we exposed HepG2 cells to these congeners and their mixture at 1 nM, analyzing their transcriptomic and proteomic profiles. KEGG pathways and GSEA Hallmarks enrichment analyses evidenced that BDE-47 disrupted the glucose metabolism and hypoxia pathway; all the congeners and the MIX affected lipid metabolism and signaling Hallmarks regulating metabolism as mTORC1 and PI3K/AKT/MTOR. These results were confirmed by glucose secretion depletion and increased lipid accumulation, especially in BDE-47 and -209 treated cells. These congeners also affected the EGFR/MAPK signaling; further, BDE-47 enriched the estrogen pathway. Interestingly, BDE-209 and the MIX increased ERα gene expression, whereas all the congeners and the MIX induced ERβ and PPARα. We also found that PBDEs modulated several lncRNAs and that HNRNAP1 represented a central hub in all the four interaction networks. Overall, the PBDEs investigated affected glucose and lipid metabolism with different underlying modes of action, as highlighted by the integrated omics analysis, at a dietary relevant concentration. These results may support the mechanism-based risk assessment of these compounds in relation to liver metabolism disruption.
Collapse
Affiliation(s)
- Marialuisa Casella
- Core Facilities, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Gabriele Lori
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
- Science Department, Università Degli Studi di Roma Tre, Viale Guglielmo Marconi 446, 00146 Rome, Italy
| | - Lucia Coppola
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Cinzia La Rocca
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Sabrina Tait
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
- Correspondence: ; Tel.: +39-06-49902839
| |
Collapse
|
42
|
Lucas RM, Luo L, Stow JL. ERK1/2 in immune signalling. Biochem Soc Trans 2022; 50:1341-1352. [PMID: 36281999 PMCID: PMC9704528 DOI: 10.1042/bst20220271] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 07/30/2023]
Abstract
Extracellular signal-related kinases 1 and 2 (ERK1/2) are the final components of the mitogen-activated protein kinase (MAPK) phosphorylation cascade, an integral module in a diverse array of signalling pathways for shaping cell behaviour and fate. More recently, studies have shown that ERK1/2 plays an essential role downstream of immune receptors to elicit inflammatory gene expression in response to infection and cell or tissue damage. Much of this work has studied ERK1/2 activation in Toll-like receptor (TLR) pathways, providing mechanistic insights into its recruitment, compartmentalisation and activation in cells of the innate immune system. In this review, we summarise the typical activation of ERK1/2 in growth factor receptor pathways before discussing its known roles in immune cell signalling with a focus downstream of TLRs. We examine emerging research uncovering evidence of dysfunctional ERK1/2 signalling in inflammatory diseases and discuss the potential therapeutic benefit of targeting ERK1/2 pathways in inflammation.
Collapse
Affiliation(s)
- Richard M. Lucas
- Institute for Molecular Bioscience (IMB) and Centre for Inflammation and Disease Research, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Lin Luo
- Institute for Molecular Bioscience (IMB) and Centre for Inflammation and Disease Research, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Jennifer L. Stow
- Institute for Molecular Bioscience (IMB) and Centre for Inflammation and Disease Research, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
43
|
Okuyama S, Yoshimura M, Amakura Y, Nakajima M, Furukawa Y. Activation of Extracellular Signal-Regulated Kinase 2 and cAMP Response Element-Binding Protein in Cultured Neurons by the Macrocyclic Ellagitannin Oenothein B. NEUROSCI 2022; 3:387-394. [PMID: 39483426 PMCID: PMC11523700 DOI: 10.3390/neurosci3030028] [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: 05/22/2022] [Accepted: 07/04/2022] [Indexed: 11/03/2024] Open
Abstract
(1) Background: The findings of our recent in vivo study indicated that the oral administration of oenothein B, a unique macrocyclic ellagitannin, activated extracellular signal-regulated kinase (ERK) 2 and cAMP response element-binding protein (CREB) in the mouse brain. A large hydrophilic oenothein B is unable to reach the brain, suggesting that any metabolite(s) of oenothein B might function in the brain. (2) Results: The addition of oenothein B to the culture medium of rat cortical neurons induced the prompt and significant activation of ERK2 and CREB. (3) Conclusions: The activation of ERK2 and CREB is crucial for synaptic transmission and learning/memory formation in the brain. The present results suggest oenothein B exerts neurotrophic/neuroprotective effects in the brain through the modulation of neuronal signaling pathways, if it reaches the brain.
Collapse
Affiliation(s)
- Satoshi Okuyama
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama 790-8578, Ehime, Japan; (S.O.); (M.N.)
| | - Morio Yoshimura
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama 790-8578, Ehime, Japan; (M.Y.); (Y.A.)
| | - Yoshiaki Amakura
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama 790-8578, Ehime, Japan; (M.Y.); (Y.A.)
| | - Mitsunari Nakajima
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama 790-8578, Ehime, Japan; (S.O.); (M.N.)
| | - Yoshiko Furukawa
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, 4-2 Bunkyo-cho, Matsuyama 790-8578, Ehime, Japan; (S.O.); (M.N.)
| |
Collapse
|
44
|
Rasl J, Grusanovic J, Klimova Z, Caslavsky J, Grousl T, Novotny J, Kolar M, Vomastek T. ERK2 signaling regulates cell-cell adhesion of epithelial cells and enhances growth factor-induced cell scattering. Cell Signal 2022; 99:110431. [PMID: 35933033 DOI: 10.1016/j.cellsig.2022.110431] [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: 03/29/2022] [Revised: 07/13/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
Abstract
The ERK signaling pathway, consisting of core protein kinases Raf, MEK and effector kinases ERK1/2, regulates various biological outcomes such as cell proliferation, differentiation, apoptosis, or cell migration. Signal transduction through the ERK signaling pathway is tightly controlled at all levels of the pathway. However, it is not well understood whether ERK pathway signaling can be modulated by the abundance of ERK pathway core kinases. In this study, we investigated the effects of low-level overexpression of the ERK2 isoform on the phenotype and scattering of cuboidal MDCK epithelial cells growing in discrete multicellular clusters. We show that ERK2 overexpression reduced the vertical size of lateral membranes that contain cell-cell adhesion complexes. Consequently, ERK2 overexpressing cells were unable to develop cuboidal shape, remained flat with increased spread area and intercellular adhesive contacts were present only on the basal side. Interestingly, ERK2 overexpression was not sufficient to increase phosphorylation of multiple downstream targets including transcription factors and induce global changes in gene expression, namely to increase the expression of pro-migratory transcription factor Fra1. However, ERK2 overexpression enhanced HGF/SF-induced cell scattering as these cells scattered more rapidly and to a greater extent than parental cells. Our results suggest that an increase in ERK2 expression primarily reduces cell-cell cohesion and that weakened intercellular adhesion synergizes with upstream signaling in the conversion of the multicellular epithelium into single migrating cells. This mechanism may be clinically relevant as the analysis of clinical data revealed that in one type of cancer, pancreatic adenocarcinoma, ERK2 overexpression correlates with a worse prognosis.
Collapse
Affiliation(s)
- Jan Rasl
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic; Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Josipa Grusanovic
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
| | - Zuzana Klimova
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
| | - Josef Caslavsky
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
| | - Tomas Grousl
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
| | - Jiri Novotny
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic; Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology, 160 00 Prague, Czech Republic
| | - Michal Kolar
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Tomas Vomastek
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic.
| |
Collapse
|
45
|
The role of protein kinases as key drivers of metabolic dysfunction-associated fatty liver disease progression: New insights and future directions. Life Sci 2022; 305:120732. [PMID: 35760093 DOI: 10.1016/j.lfs.2022.120732] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 02/07/2023]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD), proposed in 2020 is a novel term for non-alcoholic fatty liver disease (NAFLD) which was coined for the first time in 1980. It is a leading cause of the most chronic liver disease and hepatic failure all over the world, and unfortunately, with no licensed drugs for treatment yet. The progress of the disease is driven by the triggered inflammatory process, oxidative stress, and insulin resistance in many pathways, starting with simple hepatic steatosis to non-alcoholic steatohepatitis, fibrosis, cirrhosis, and liver cancer. Protein kinases (PKs), such as MAPK, ErbB, PKC, PI3K/Akt, and mTOR, govern most of the pathological pathways by acting on various downstream key points in MAFLD and regulating both hepatic gluco- lipo-neogenesis and inflammation. Therefore, modulating the function of those potential protein kinases that are effectively involved in MAFLD might be a promising therapeutic approach for tackling this disease. In the current review, we have discussed the key role of protein kinases in the pathogenesis of MAFLD and performed a protein-protein interaction (PPI) network among the main proteins of each kinase pathway with MAFLD-related proteins to predict the most likely targets of the PKs in MAFLD. Moreover, we have reported the experimental, pre-clinical, and clinical data for the most recent investigated molecules that are activating p38-MAPK and AMPK proteins and inhibiting the other PKs to improve MAFLD condition by regulating oxidation and inflammation signalling.
Collapse
|
46
|
Extracellular Signal-Regulated Kinase 1 Alone Is Dispensable for Hyperoxia-Mediated Alveolar and Pulmonary Vascular Simplification in Neonatal Mice. Antioxidants (Basel) 2022; 11:antiox11061130. [PMID: 35740027 PMCID: PMC9219973 DOI: 10.3390/antiox11061130] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 02/04/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a morbid lung disease distinguished by lung alveolar and vascular simplification. Hyperoxia, an important BPD causative factor, increases extracellular signal-regulated kinases (ERK)-1/2 expression, whereas decreased lung endothelial cell ERK2 expression reduces angiogenesis and potentiates hyperoxia-mediated BPD in mice. However, ERK1′s role in experimental BPD is unclear. Thus, we hypothesized that hyperoxia-induced experimental BPD would be more severe in global ERK1-knockout (ERK1-/-) mice than their wild-type (ERK1+/+ mice) littermates. We determined the extent of lung development, ERK1/2 expression, inflammation, and oxidative stress in ERK1-/- and ERK1+/+ mice exposed to normoxia (FiO2 21%) or hyperoxia (FiO2 70%). We also quantified the extent of angiogenesis and hydrogen peroxide (H2O2) production in hyperoxia-exposed neonatal human pulmonary microvascular endothelial cells (HPMECs) with normal and decreased ERK1 signaling. Compared with ERK1+/+ mice, ERK1-/- mice displayed increased pulmonary ERK2 activation upon hyperoxia exposure. However, the extent of hyperoxia-induced inflammation, oxidative stress, and interrupted lung development was similar in ERK1-/- and ERK1+/+ mice. ERK1 knockdown in HPMECs increased ERK2 activation at baseline, but did not affect in vitro angiogenesis and hyperoxia-induced H2O2 production. Thus, we conclude ERK1 is dispensable for hyperoxia-induced experimental BPD due to compensatory ERK2 activation.
Collapse
|
47
|
Yang X, Zeng T, Liu Z, He W, Hu M, Tang T, Chen L, Xing L. Long noncoding RNA GK-IT1 promotes esophageal squamous cell carcinoma by regulating MAPK1 phosphorylation. Cancer Med 2022; 11:4555-4574. [PMID: 35608100 PMCID: PMC9741976 DOI: 10.1002/cam4.4795] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/13/2022] [Accepted: 03/30/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are implicated in the oncogenesis and metastasis of multiple human cancers. Nonetheless, the precise molecular mechanisms underlying the oncogenic role of lncRNA in esophageal squamous cell carcinoma (ESCC) remains to be clarified. METHODS The expression of GK intronic transcript 1 (GK-IT1) was analyzed using ESCC RNA-seq data from The Cancer Genome Atlas database. Quantitative real-time PCR was used to measure the expression of GK-IT1 in ESCC clinical samples and cells. The correlation between GK-IT1 expression and clinicopathological variables was examined using chi-squared tests. Kaplan-Meier survival and Cox regression analyses were employed to generate the survival curve and assess the prognostic value of GK-IT1. Functional experiments were utilized to explore the role of GK-IT1 in promoting cell migration, invasion, proliferation, and suppressing apoptosis and autophagy in ESCC. To understand the mechanism, an RNA pulldown assay, RNA immunoprecipitation, agarose gel electrophoresis, immunofluorescence, and co-immunoprecipitation assays were used. RESULTS In this study we identified an unreported lncRNA, termed GK-IT1 that was aberrantly overexpressed in ESCC tissues and cells. GK-IT1 was closely associated with advanced clinical stage, and it was an independent prognostic indicator of ESCC. Functional assays verified that GK-IT1 significantly promoted ESCC proliferation, invasion, and migration, and suppressed ESCC apoptosis and autophagy. Furthermore, tumorigenesis experiments in nude mice indicated that GK-IT1 promoted ESCC tumor growth and metastasis. Mechanistically, GK-IT1 competitively bound to mitogen-activated protein kinase 1 (MAPK1) to prevent the interaction between dual specificity phosphatase 6 (DUSP6) and MAPK1, thereby controlling the phosphorylation of MAPK1 and promoting ESCC progression. CONCLUSION Our study revealed that GK-IT1 competed with DUSP6 to attenuate the interaction between DUSP6 and MAPK1, leading to activation of the ERK/MAPK pathway, thereby promoting progression of ESCC. Our research indicated that GK-IT1 served as a novel potential target for the diagnosis and treatment of ESCC.
Collapse
Affiliation(s)
- Xin Yang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Tianyang Zeng
- Department of Thoracic SurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Ziyang Liu
- Department of Thoracic SurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Wanlun He
- The Frist People's HospitalChongqing Liang Jiang New AreaChongqingChina
| | - Mengting Hu
- Department of Cell Biology and GeneticsChongqing Medical UniversityChongqingChina
| | - Ti Tang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Li Chen
- Department of Thoracic SurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Lei Xing
- Department of Endocrine and Breast SurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
48
|
A Long-Lasting PARP1-Activation Mediates Signal-Induced Gene Expression. Cells 2022; 11:cells11091576. [PMID: 35563882 PMCID: PMC9101275 DOI: 10.3390/cells11091576] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/26/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
This overview presents recent evidence for a long-lasting PARP1 activation by a variety of signal transduction mechanisms, mediating signal-induced gene expression and chromatin remodeling. This mode of PARP1 activation has been reported in a variety of cell types, under physiological conditions. In this mechanism, PARP1 is not transiently activated by binding to DNA breaks. Moreover, damaged DNA interfered with this long-lasting PARP1 activation.
Collapse
|
49
|
Cingolani F, Liu Y, Shen Y, Wen J, Farris AB, Czaja MJ. Redundant Functions of ERK1 and ERK2 Maintain Mouse Liver Homeostasis Through Down-Regulation of Bile Acid Synthesis. Hepatol Commun 2022; 6:980-994. [PMID: 34936222 PMCID: PMC9035584 DOI: 10.1002/hep4.1867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/22/2021] [Accepted: 11/03/2021] [Indexed: 11/07/2022] Open
Abstract
Activation of extracellular signal-regulated kinase (ERK) 1/2 promotes hepatocyte proliferation in response to growth stimuli, but whether constitutive hepatocyte ERK1/2 signaling functions in liver physiology is unknown. To examine the role of ERK1/2 in hepatic homeostasis, the effects of a knockout of Erk1 and/or Erk2 in mouse liver were examined. The livers of mice with a global Erk1 knockout or a tamoxifen-inducible, hepatocyte-specific Erk2 knockout were normal. In contrast, Erk1/2 double-knockout mice developed hepatomegaly and hepatitis by serum transaminases, histology, terminal deoxynucleotide transferase-mediated deoxyuridine triphosphate nick end-labeling, and assays of hepatic inflammation. Liver injury was associated with biochemical evidence of cholestasis with increased serum and hepatic bile acids and led to hepatic fibrosis and mortality. RNA sequencing and polymerase chain reaction analysis of double-knockout mouse livers revealed that the rate-limiting bile acid synthesis gene Cyp7a1 (cholesterol 7α-hydroxylase) was up-regulated in concert with decreased expression of the transcriptional repressor short heterodimer partner. Elevated bile acids were the mechanism of liver injury, as bile acid reduction by SC-435, an inhibitor of the ileal apical sodium-dependent bile acid transporter, prevented liver injury. Conclusion: Constitutive ERK1 and ERK2 signaling has a redundant but critical physiological function in the down-regulation of hepatic bile acid synthesis to maintain normal liver homeostasis.
Collapse
Affiliation(s)
- Francesca Cingolani
- Division of Digestive DiseasesDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Yunshan Liu
- Division of Digestive DiseasesDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Yang Shen
- Division of Digestive DiseasesDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Jing Wen
- Division of Digestive DiseasesDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Alton B. Farris
- Department of Pathology & Laboratory MedicineEmory University School of MedicineAtlantaGAUSA
| | - Mark J. Czaja
- Division of Digestive DiseasesDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| |
Collapse
|
50
|
Pan X, Pei J, Wang A, Shuai W, Feng L, Bu F, Zhu Y, Zhang L, Wang G, Ouyang L. Development of small molecule extracellular signal-regulated kinases (ERKs) inhibitors for cancer therapy. Acta Pharm Sin B 2022; 12:2171-2192. [PMID: 35646548 PMCID: PMC9136582 DOI: 10.1016/j.apsb.2021.12.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 01/09/2023] Open
Abstract
The mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway is widely activated by a variety of extracellular stimuli, and its dysregulation is associated with the proliferation, invasion, and migration of cancer cells. ERK1/2 is located at the distal end of this pathway and rarely undergoes mutations, making it an attractive target for anticancer drug development. Currently, an increasing number of ERK1/2 inhibitors have been designed and synthesized for antitumor therapy, among which representative compounds have entered clinical trials. When ERK1/2 signal transduction is eliminated, ERK5 may provide a bypass route to rescue proliferation, and weaken the potency of ERK1/2 inhibitors. Therefore, drug research targeting ERK5 or based on the compensatory mechanism of ERK5 for ERK1/2 opens up a new way for oncotherapy. This review provides an overview of the physiological and biological functions of ERKs, focuses on the structure-activity relationships of small molecule inhibitors targeting ERKs, with a view to providing guidance for future drug design and optimization, and discusses the potential therapeutic strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Xiaoli Pan
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Junping Pei
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Aoxue Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Wen Shuai
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Lu Feng
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Faqian Bu
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Yumeng Zhu
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|