1
|
Peng S, Zhao Y, Jiang W, Long Y, Hu T, Li M, Hu J, Shen Y. MAPK signaling mediated intestinal inflammation induced by endoplasmic reticulum stress and NOD2. Mol Cell Biochem 2025; 480:3709-3717. [PMID: 39806198 DOI: 10.1007/s11010-025-05212-3] [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: 08/10/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Endoplasmic reticulum (ER) stress is crucially involved in inflammatory bowel disease (IBD), but the mechanisms remain incompletely understood. This study aimed to elucidate how ER stress promotes inflammation in IBD. ER stress marker Grp78 and NOD2 in colon tissues of Crohn's disease (CD) patients and IBD model mice were detected by immunohistochemical analysis. THP-1 cells were exposed to ER stress and the expression of NOD2 and inflammatory cytokines was detected by PCR. We found that ER stress markers Grp78 and NOD2 were upregulated in intestinal tissues of CD patients and in THP-1 cells exposed to ER stress. ER stress inhibitor reduced Grp78 and NOD2 expression in colitis model mice and alleviated colitis. ER stress inducer cooperated with NOD2 ligand MDP to upregulate TNF-α, IL-8 and IL-1β, and activate MAPK signaling in THP-1 cells. Moreover, inhibitors of MAPK signaling led to the downregulation of IL-1β, IL-8 and TNF-α in THP-1 cells stimulated by ER stress inducer and MDP. In conclusion, ER stress upregulates NOD2 and promotes inflammation in IBD, at least partially due to the activation of MAPK pathway.
Collapse
Affiliation(s)
- Siyuan Peng
- Department of Digestive Diseases, Changsha Central Hospital Affiliated to University of South China, No.161 Shaoshan Nanlu, Changsha, Hunan, China
| | - Yan Zhao
- Department of Pathology, Changsha Central Hospital Affiliated to University of South China, No.161 Shaoshan Nanlu, Changsha, Hunan, China
| | - Wang Jiang
- Department of Digestive Diseases, Changsha Central Hospital Affiliated to University of South China, No.161 Shaoshan Nanlu, Changsha, Hunan, China
| | - Yan Long
- Department of Digestive Diseases, Changsha Central Hospital Affiliated to University of South China, No.161 Shaoshan Nanlu, Changsha, Hunan, China
| | - Tian Hu
- Department of Digestive Diseases, Changsha Central Hospital Affiliated to University of South China, No.161 Shaoshan Nanlu, Changsha, Hunan, China
| | - Mengling Li
- Department of Digestive Diseases, Changsha Central Hospital Affiliated to University of South China, No.161 Shaoshan Nanlu, Changsha, Hunan, China
| | - Jinyue Hu
- Medical Research Center, Changsha Central Hospital Affiliated to University of South China, No.161 Shaoshan Nanlu, Changsha, Hunan, China
| | - Yueming Shen
- Department of Digestive Diseases, Changsha Central Hospital Affiliated to University of South China, No.161 Shaoshan Nanlu, Changsha, Hunan, China.
| |
Collapse
|
2
|
Pinto JR, Deepika Bhat K, Bose B, Sudheer Shenoy P. Irisin: muscle's novel player in endoplasmic reticulum stress and disease. Mol Cell Biochem 2025; 480:3605-3619. [PMID: 39984795 DOI: 10.1007/s11010-025-05225-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/02/2025] [Indexed: 02/23/2025]
Abstract
Irisin, an exercise-induced myokine, exhibits elevated levels during physical activity, yet its role in modulating the unfolded protein response (UPR) remains poorly understood. This comprehensive review pioneers an in-depth examination of irisin-mediated endoplasmic reticulum (ER) stress mitigation across various diseases. We provide a nuanced characterization of irisin's molecular profile, biological activity, and significance as a skeletal muscle-derived cytokine analogue. Our discussion elucidates the complex interplay between exercise, irisin signalling, and metabolic outcomes, highlighting key molecular interactions driving salutary effects. Moreover, we delineate the UPR's role as a critical ER stress countermeasure and underscore irisin's pivotal function in alleviating this stress, revealing potential therapeutic avenues for disease management. Exercise-induced release of irisin ameliorates ER stress through AMPK phosphorylation during various diseases (Icon image source: www.flaticon.com ).
Collapse
Affiliation(s)
- Joel Rimson Pinto
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to be University, University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - K Deepika Bhat
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to be University, University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to be University, University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - P Sudheer Shenoy
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to be University, University Road, Deralakatte, Mangalore, Karnataka, 575018, India.
| |
Collapse
|
3
|
de Morais Gomes V, Santos DM, Macedo-da-Silva J, Lazari LC, Machado RRG, Dos Santos AF, Araujo DB, Coutinho JVP, Arini GS, Angeli CB, de Souza EE, Marques RF, Boscardin SB, Wrenger C, Marinho CRF, Oliveira DBL, Durigon EL, Labriola L, Rosa-Fernandes L, Palmisano G. P.1 and P.2 SARS-CoV-2 Brazilian variants activate the unfolded protein response with a time and pathway specificity. J Proteomics 2025; 315:105397. [PMID: 39909104 DOI: 10.1016/j.jprot.2025.105397] [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: 08/15/2024] [Revised: 01/07/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
COVID-19 is a human respiratory syndrome caused by the infection of the SARS-CoV-2 virus that has a high rate of infection and mortality. Viruses modulate the host machinery by altering cellular mechanisms that favor their replication. One of the mechanisms that viruses exploit is the protein folding and processing of post-translational modifications that occur in the endoplasmic reticulum (ER). When ER function is impaired, there is an accumulation of misfolded proteins leading to endoplasmic reticulum stress (ER stress). To maintain homeostasis, cells trigger an adaptive signaling mechanism called the Unfolded Protein Response (UPR) which helps cells deal with stress, but under severe conditions, can activate the apoptotic cell death mechanism. This study elucidated an activation of a diversity of molecular mechanisms by Brazilian variants of SARS-CoV-2 by a time-resolved and large-scale characterization of SARS-CoV-2-infected cells proteomics and immunoblotting. Furthermore, it was shown that pharmacological UPR modulation could reduce viral release by counteracting the different viral activations of its cellular response. Analysis of human clinical specimens and disease outcomes focusing on ER stress reinforces the importance of UPR modulation as a host regulatory mechanism during viral infection and could point to novel therapeutic targets. SIGNIFICANCE: Since the emergence of SARS-CoV-2 and the consequent COVID-19 pandemic, the rapid emergence of variants of this new coronavirus has been a cause for concern since many of them have significantly higher rates of transmissibility and virulence, being called Variants of Concern (VOC). In this work, we studied the VOCs Gamma (P.1) and Zeta (P.2), also known as Brazilian variants. Constant evidence has reported that there are particularities related to each variant of SARS-CoV-2, with different rates of transmissibility, replication and modulation of host biological processes being observed, in addition to the mutations present in the variants. For this reason, this work focused on infections caused by the Brazilian variants of SARS-CoV-2 in different cell lines, in which we were able to observe that the infections caused by the variants induced endoplasmic reticulum stress in the infected cells and activated the UPR pathways, presenting specific modulations of each variant in this pathway. Furthermore, transcriptome analysis of patients revealed a correlation between ER-related genes and COVID-19 progression. Finally, we observed that the use of UPR modulators in host cells decreased viral release of all variants without affecting cell viability. The data presented in this work complement the observations of other studies that aim to understand the pathogenicity of SARS-CoV-2 VOCs and possible new therapeutic strategies, mainly targeting biological processes related to the endoplasmic reticulum.
Collapse
Affiliation(s)
| | - Deivid Martins Santos
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Janaina Macedo-da-Silva
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Lucas C Lazari
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | | | | | - Danielle Bastos Araujo
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil
| | | | - Gabriel Santos Arini
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Brazil
| | - Claudia B Angeli
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Edmarcia E de Souza
- Unit for Drug Discovery, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Rodolfo F Marques
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, ICB, University of São Paulo, Brazil
| | | | - Danielle B L Oliveira
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil
| | - Edison L Durigon
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil; Scientific Platform Pasteur USP, Sao Paulo, Brazil
| | - Leticia Labriola
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Brazil
| | - Livia Rosa-Fernandes
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil; Laboratory of Experimental Immunoparasitology, Department of Parasitology, ICB, University of São Paulo, Brazil; Centre for Motor Neuron Disease Research, Faculty of Medicine, Health & Human Sciences, Macquarie Medical School, Sydney, Australia
| | - Giuseppe Palmisano
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil; School of Natural Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
4
|
Sha N, Zhou B, Hou G, Xi Z, Wang W, Yan M, He J, Zhou Y, Xia Q, Jiang Y, Zhao Q. The protection of UCK2 protein stability by GART maintains pyrimidine salvage synthesis for HCC growth under glucose limitation. Oncogene 2025; 44:1078-1092. [PMID: 39865175 DOI: 10.1038/s41388-025-03274-7] [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: 07/16/2024] [Revised: 12/06/2024] [Accepted: 01/14/2025] [Indexed: 01/28/2025]
Abstract
Overexpression of uridine-cytidine kinase 2 (UCK2), a key enzyme in the pyrimidine salvage pathway, is implicated in human cancer development, while its regulation under nutrient stress remains to be investigated. Here, we show that under glucose limitation, AMPK phosphorylates glycinamide ribonucleotide formyltransferase (GART) at Ser440, and this modification facilitates its interaction with UCK2. Through its binding to UCK2, GART generates tetrahydrofolate (THF) and thus inhibits the activity of integrin-linked kinase associated phosphatase (ILKAP) for removing AKT1-mediated UCK2-Ser254 phosphorylation under glucose limitation, in which dephosphorylation of UCK2-Ser254 tends to cause Trim21-mediated UCK2 polyubiquitination and degradation. In this way, both UCK2 binding ability and THF producing catalytic activity of GART protect protein stability of UCK2 and pyrimidine salvage synthesis, and sustain tumor cell growth under glucose limitation. In addition, UCK2-Ser254 phosphorylation level displays a positive relationship with GART-Ser440 phosphorylation level and its enhancement is correlated with poor prognosis of human hepatocellular carcinoma (HCC) patients. These findings reveal a non-canonical role of GART in regulating pyrimidine salvage synthesis under nutrient stress, and raise the potential for alternative treatments in targeting pyrimidine salvage-dependent tumor growth.
Collapse
Affiliation(s)
- Nannan Sha
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bei Zhou
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guofang Hou
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhifeng Xi
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wang Wang
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Man Yan
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing He
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Zhou
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yuhui Jiang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| | - Qin Zhao
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Liu J, Zuo Z, Ewing M, Cao Q, Cao L, Li Q, Finkel T, Leppla SH, Liu S. ERK pathway reactivation prevents anthrax toxin lethality in mice. Nat Microbiol 2025; 10:1145-1155. [PMID: 40155776 DOI: 10.1038/s41564-025-01977-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: 07/19/2024] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Lethal toxin (LT), the major virulence factor of Bacillus anthracis, proteolytically inactivates MEKs and disables downstream ERK, p38 and JNK pathway signalling leading to tissue damage and mortality. Therapies for LT-induced damage after host cell internalization of the toxin are lacking. Here we constructed MEK variants in which the LT proteolytic site was modified: MEK2(P10V/A11D), MEK3(I27D) and MEK6(I15D). These variants were resistant to proteolysis by LT. Expression in cells enabled sustained activation of ERK and p38 pathways and promoted cell survival upon LT treatment. Survival of LT- or B. anthracis-challenged MEK variant transgenic mice also increased compared with controls. We found that LT-mediated disruption of both ERK and p38 pathway is essential for anthrax pathogenesis. We show that engagement of upstream receptor tyrosine kinases reactivated the LT-disrupted ERK pathway, as did administering a cocktail of EGF, GM-CSF and FGF2 growth factors, which significantly increased survival of LT- or B. anthracis-challenged mice. These findings offer potential towards developing damage-limiting therapeutic strategies for anthrax.
Collapse
Affiliation(s)
- Jie Liu
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zehua Zuo
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Michael Ewing
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Qing Cao
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Liu Cao
- Health Sciences Institute, China Medical University, Shenyang, China
| | - Qi Li
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Toren Finkel
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen H Leppla
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shihui Liu
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Guo L, Lee HK, Oh S, Koirala GR, Kim TI. Smart Bioelectronics for Real-Time Diagnosis and Therapy of Body Organ Functions. ACS Sens 2025. [PMID: 40310273 DOI: 10.1021/acssensors.5c00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Noncommunicable diseases (NCDs) associated with cardiovascular, neurological, and gastrointestinal disorders remain a leading cause of global mortality, sounding the alarm for the urgent need for better diagnostic and therapeutic solutions. Wearable and implantable biointegrated electronics offer a groundbreaking solution, combining real-time, high-resolution monitoring with innovative treatment capabilities tailored to specific organ functions. In this comprehensive review, we focus on the diseases affecting the brain, heart, gastrointestinal organs, bladder, and adrenal gland, along with their associated physiological parameters. Additionally, we provide an overview of the characteristics of these parameters and explore the potential of bioelectronic devices for in situ sensing and therapeutic applications and highlight the recent advancements in their deployment across specific organs. Finally, we analyze the current challenges and prospects of implementing closed-loop feedback control systems in integrated sensor-therapy applications. By emphasizing organ-specific applications and advocating for closed-loop systems, this review highlights the potential of future bioelectronics to address physiological needs and serves as a guide for researchers navigating the interdisciplinary fields of diagnostics, therapeutics, and personalized medicine.
Collapse
Affiliation(s)
- Lili Guo
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Hin Kiu Lee
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Suyoun Oh
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Gyan Raj Koirala
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Tae-Il Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| |
Collapse
|
7
|
Kubatka P, Bojkova B, Nosalova N, Huniadi M, Samuel SM, Sreenesh B, Hrklova G, Kajo K, Hornak S, Cizkova D, Bubnov R, Smokovski I, Büsselberg D, Golubnitschaja O. Targeting the MAPK signaling pathway: implications and prospects of flavonoids in 3P medicine as modulators of cancer cell plasticity and therapeutic resistance in breast cancer patients. EPMA J 2025. [DOI: 10.1007/s13167-025-00407-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/24/2025] [Indexed: 04/21/2025]
Abstract
Abstract
Cancer drug resistance poses a significant challenge in oncology, primarily driven by cancer cell plasticity, which promotes tumor initiation, progression, metastasis, and therapeutic evasion in many different cancers. Breast cancers (BCs) are a prominent example of that, with an estimated 2.3 million new cases and 670,000 BC-related deaths registered worldwide annually. Triple-negative BC is especially challenging for treatments demonstrating particularly aggressive disease course, an early manifestation of metastatic disease, frequent drug-resistant cancer types, and poor individual outcomes. Although chemosensitizing agents have been developed, their clinical utility in oncology remains unproven. The mitogen-activated protein kinase (MAPK) pathway is considered a critical regulator of intracellular and extracellular signaling highly relevant for both — genetic and epigenetic modifications. Dysregulation of the MAPK signaling pathways plays a significant role in conferring chemoresistance in BC. Contextually, targeting the MAPK pathway represents a promising strategy for overcoming drug resistance and enhancing the therapeutic efficacy of anticancer agents in BC treatment. On the other hand, flavonoids, a prominent class of phytochemicals, are key modulators of MAPK signaling. Flavonoids interact with the ERK, JNK, p38, and ERK5 pathways of the MAPK signaling cascade and present a promising avenue for developing novel anti-cancer therapies and re-sensitizing agents for the treatment of BC. Compounds such as quercetin, kaempferol, genistein, luteolin, myricetin, EGCG, baicalein, baicalin, nobiletin, morin, delphinidin, acacetin, isorhamnetin, apigenin, silymarin, among others, have been identified as specific modulators of MAPK signaling, exerting complex downstream effects in BC cells increasing therewith drug efficacy and suppressing tumor growth and aggressivity. These properties reflect mechanisms of great clinical relevance to overcome therapeutic resistance in overall BC management. This article highlights corresponding mechanisms and provides clinically relevant illustrations in the framework of 3P medicine for primary (protection of individuals at high risk against health-to-disease transition) and secondary care (protection against metastatic BC progression). 3PM novelty makes good use of patient phenotyping and stratification, predictive multi-level diagnostics, and application of Artificial Intelligence (AI) tools to the individualized interpretation of big data — all proposed for cost-effective treatments tailored to individualized patient profiles with clear benefits to patients and advanced BC management.
Collapse
|
8
|
Zhang HY, Li KY, Wang YL, Wei CJ, Gao YX, Ren-Zhou, Zhong YB, Yin ZJ, Ren DL. ROS regulates circadian rhythms by modulating Ezh2 interactions with clock proteins. Redox Biol 2025; 81:103526. [PMID: 39952198 PMCID: PMC11875201 DOI: 10.1016/j.redox.2025.103526] [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: 01/10/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025] Open
Abstract
Redox imbalance induced by the accumulation of reactive oxygen species (ROS) accelerates age-related processes, often accompanied by a decrease in circadian rhythm amplitude. However, the underlying mechanisms by which ROS modulate circadian rhythms remain poorly understood. In this study, we found that ROS disrupt circadian rhythms in both zebrafish, as indicated by changes in diurnal behavior and clock gene expression, and in a human cell model. Using weighted gene co-expression network analysis (WGCNA) and machine learning approaches (RF, LASSO, SVM), EZH2 was identified as a key gene involved in regulating circadian rhythms under oxidative stress conditions. To further investigate the role of EZH2, we employed ezh2-/- mutants, Morpholino injection, and overexpression treatment and discovered that EZH2 is crucial in mediating the effect of ROS on circadian rhythms. Furthermore, EZH2 interacts with the CLOCK-BMAL1 complex to regulate the transcription of clock genes, as demonstrated through co-immunoprecipitation (co-IP), chromatin immunoprecipitation (ChIP), and dual-luciferase reporter assays. Our study revealed that ROS disrupt circadian rhythms by regulating the interaction between EZH2 and the CLOCK-BMAL1 complex, shedding light on the molecular mechanisms of circadian rhythm disruption under oxidative stress and suggesting potential targets for age-related and circadian disorders.
Collapse
Affiliation(s)
- Hao-Yi Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Ke-Yun Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Yi-Li Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Chun-Jiao Wei
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Yu-Xuan Gao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Ren-Zhou
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Ying-Bin Zhong
- School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu, 215000, China
| | - Zong-Jun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Da-Long Ren
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China.
| |
Collapse
|
9
|
Zhao L, Wu X, Li Q, Shen Y, Zeng S, Wang J, Liu Q. Inhibition of CCN5 Protects Against Apoptosis and Endoplasmic Reticulum Stress in Bisphenol A-Induced Sertoli Cells via p38/JNK MAPK Signaling Pathway. DNA Cell Biol 2025; 44:174-185. [PMID: 40184555 DOI: 10.1089/dna.2024.0247] [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: 04/06/2025] Open
Abstract
Bisphenol A (BPA) is the most common endocrine disruptor that has toxicity to the reproductive system and male infertility. However, the underlying mechanisms of BPA's toxicity to Sertoli cells remain poorly understood. Cellular communication network factor 5 (CCN5) is reported to regulate cell proliferation, apoptosis, and differentiation. Our study demonstrated a significant elevation of CCN5 expression in the testis of nonobstructive azoospermia patients and TM4 Sertoli cells exposed to BPA. Knockdown of CCN5 reduced apoptotic cells after BPA treatment, as determined by flow cytometry and terminal deoxynucleotidyl transferase dUTP nick end labeling assays. Cells exposed to BPA showed increased expressions of Bax and cleaved poly(ADP-ribose) polymerase, decreased expression of Bcl-2, as well as elevated activities of caspase-3 and caspase-9 in BPA-induced TM4 cells, which were reversed by CCN5 inhibition. Loss of CCN5 declined phosphorylation of protein kinase R-like endoplasmic reticulum kinase and eukaryotic translation initiation factor 2A and decreased activating transcription factor 4 and C/EBP-homologous protein in BPA-treated cells. Furthermore, silencing CCN5 blocked BPA-induced phosphorylation of p38 and c-Jun N-terminal kinase (JNK). Administration of anisomycin, a mitogen-activated protein kinase (MAPK) activator, reversed the effects of CCN5 knockdown on BPA-induced endoplasmic reticulum (ER) stress and apoptosis. Taken together, CCN5 promotes apoptosis and ER stress in Sertoli cells exposed to BPA by activating the p38/JNK MAPK signaling pathway.
Collapse
Affiliation(s)
- Lijiang Zhao
- Tianjin Medical University, Tianjin, China
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Xueqing Wu
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Qiang Li
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Yan Shen
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Sheng Zeng
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
| | - Jinbao Wang
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
10
|
Himani, Kaur C, Kumar R, Mishra R, Singh G. Targeting TGF-β: a promising strategy for cancer therapy. Med Oncol 2025; 42:142. [PMID: 40155496 DOI: 10.1007/s12032-025-02667-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Transforming growth factor β (TGF-β) has important role in regulating the cellular processes including cell growth, differentiation, and migration. TGF-β exerts its effect by binding with transcellular membranes and kinases. Our findings demonstrate that TGF- β possess dual role as tumor suppressor and tumor promoter in different stages of cancer. TGF-β emerged as a promising anticancer agent that exhibits the apoptosis by acting on the suppressor of mothers against decapentaplegic (SMAD) and non-SMAD pathways. In this review we are focusing on the different types of TGF- β inhibitors active against skin cancer, breast cancer, colorectal cancer, lung cancer and ovarian cancer. TGF-β inhibitors includes ligand traps, monoclonal antibodies and receptor kinase inhibitors. In recent studies, TGF- β inhibitors have also been used in combination therapies in the treatment of cancer. The TGF-β has important role in vaccine therapy, Chemo and Radio Resistance in Cancer. TGF-β inhibitors present the novel therapeutic approach for the cancer therapy, highlighting the mechanism of action involved, clinical trials, challenges and exploring therapeutic opportunities. This will help to develop the novel TGF-β inhibitors as anticancer agents as well as help to resolve the problem of drug resistance by developing new drugs as anticancer agents.
Collapse
Affiliation(s)
- Himani
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Charanjit Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rajesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rakhi Mishra
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Gurvinder Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.
| |
Collapse
|
11
|
Poirier A, Utecht T, Villot R, Gélinas Y, Mouchiroud M, Kordahi M, Kolnohuz A, Pasteur C, Roy J, Beaulieu MJ, Orain M, Samson N, Blanchet MR, Joubert P, Laplante M. ZNF768 loss amplifies p53 action and reduces lung tumorigenesis in mice. Oncogene 2025:10.1038/s41388-025-03352-w. [PMID: 40133474 DOI: 10.1038/s41388-025-03352-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
Cell proliferation is a fundamental process required for organismal development, growth, and maintenance. Failure to control this process leads to several diseases, including cancer. Zinc finger protein 768 (ZNF768) is an emerging transcription factor that plays key roles in driving proliferation. In addition to controlling a gene network supporting cell division, ZNF768 physically interacts and inhibits the activity of the tumor suppressor p53. Although the importance of ZNF768 in promoting cell proliferation has been well demonstrated in vitro, the physiological and pathological roles of ZNF768 in vivo are still unknown. Here, we report the generation and characterization of a ZNF768 null mouse model. ZNF768 null mice are viable but show a growth defect early in life. Mouse embryonic fibroblasts (MEFs) isolated from ZNF768 null embryos exhibit higher p53 levels, premature senescence, and higher sensitivity to genotoxic stress. In line with these findings, ZNF768 null mice showed increased radiosensitivity. This effect was associated not only with higher expression of a subset of p53 target genes, but also with alterations in genes regulating transmembrane receptor signaling, cell adhesion, and growth. Because ZNF768 levels are elevated in tumors, we tested the impact of ZNF768 loss on cancer development in mice. Here, we show that ZNF768 deletion was sufficient to repress lung tumor development in a KRASG12D-induced cancer mouse model. Overall, our findings establish ZNF768 as an important protein controlling cell proliferation that could potentially be targeted to reduce tumorigenesis.
Collapse
Affiliation(s)
- Audrey Poirier
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada
| | - Timon Utecht
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Romain Villot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Yves Gélinas
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Mathilde Mouchiroud
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Manal Kordahi
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Alona Kolnohuz
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada
| | - Coline Pasteur
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Joanny Roy
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Marie-Josée Beaulieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Michèle Orain
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Nolwenn Samson
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Marie-Renée Blanchet
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Philippe Joubert
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Mathieu Laplante
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada.
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada.
- Faculté de médecine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
12
|
Xu L, Liu B, Xiao L, Zhang Z, Niu H, Zhao D, Sun S, Guo H. Comparative toxicity mechanisms of sulfoxaflor and lambda-cyhalothrin against Apolygus lucorum from enzymatic and transcriptomic perspectives: Efficient application of insecticides. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2025; 208:106306. [PMID: 40015898 DOI: 10.1016/j.pestbp.2025.106306] [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: 12/13/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 03/01/2025]
Abstract
The pyrethroid insecticide lambda-cyhalothrin is threatened by insecticide resistance and has been registered to control Apolygus lucorum. The sulfoximine insecticide sulfoxaflor as an excellent candidate is recommended for its management. Previous studies have mainly focused on identifying resistance genes and their sublethal effects on the biological characteristics of these two insecticides in this pest. However, the toxicity mechanism differences of lambda-cyhalothrin and sulfoxaflor exposures are largely unknown. The LD10 and LD30 values were measured with significant difference as 0.15, 0.46, 33.58, and 73.60 ng/insect for sulfoxaflor and lambda-cyhalothrin, respectively, indicating differences in the insecticide type. Exposure to sublethal sulfoxaflor resulted in a higher total number of differentially expressed genes (DEGs) (550 and 995 DEGs) than exposure to sublethal lambda-cyhalothrin (101 and 112 DEGs). Moreover, enrichment analysis showed that more metabolic and signaling pathways were involved in the toxicity of sulfoxaflor than that of lambda-cyhalothrin, and enzyme activities in the enriched pathways were induced by sulfoxaflor and inhibited by lambda-cyhalothrin. For transcriptome validation, DEGs encoding detoxification-related genes were identified and validated by quantitative real-time PCR (qRT-PCR). These results indicate that sulfoxaflor is more toxic than lambda-cyhalothrin due to different modes of action. Our findings not only first provide insight into the toxicity mechanism differences of lambda-cyhalothrin and sulfoxaflor action and detoxification in A. lucorum at molecular and biochemical levels but also offer data and techniques for registering candidate sulfoxaflor and efficient application of insecticides in the field.
Collapse
Affiliation(s)
- Lu Xu
- Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; Key Laboratory of Integrated Pest Management on Crops in East China, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
| | - Baosheng Liu
- Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Liubin Xiao
- Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Zhichun Zhang
- Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Hongtao Niu
- Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Dongxiao Zhao
- Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Shuai Sun
- Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Huifang Guo
- Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| |
Collapse
|
13
|
Yin M, Zheng X, Shi L. Targeting p38 MAPK: A potential bridge between ER stress and age-related bone loss. Cell Signal 2025; 127:111549. [PMID: 39638139 DOI: 10.1016/j.cellsig.2024.111549] [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: 08/26/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
The endoplasmic reticulum (ER) is crucial in the development of numerous age-related bone disorders. Notably, ER stress can precipitate bone loss by orchestrating inflammatory responses, apoptosis, and autophagy through the activation of the p38 MAPK pathway. Age-related bone loss diseases pose a significant burden on society and healthcare as the global population ages. This review provides a comprehensive analysis of recent research advancements, delving into the critical role of ER stress-activated p38 MAPK in inflammation, apoptosis, and autophagy, as well as its impact on bone formation and bone resorption. This review elucidates the molecular mechanisms underlying the involvement of ER stress-activated p38 MAPK in osteoporosis, rheumatoid arthritis, periodontitis, and osteoarthritis and discusses the therapeutic potential of targeting p38 MAPK. Furthermore, this review provides a scientific foundation for new therapeutic strategies by highlighting prospective research directions.
Collapse
Affiliation(s)
- Meng Yin
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xin Zheng
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Shi
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
14
|
Chandrakar P, Nelson CS, Podestà MA, Cavazzoni CB, Gempler M, Lee JM, Richardson S, Zhang H, Samarpita S, Ciofani M, Chatila T, Kuchroo VK, Sage PT. Progressively differentiated T FH13 cells are stabilized by JunB to mediate allergen germinal center responses. Nat Immunol 2025; 26:473-483. [PMID: 39891019 DOI: 10.1038/s41590-025-02077-y] [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: 04/29/2024] [Accepted: 01/06/2025] [Indexed: 02/03/2025]
Abstract
Allergic diseases are common and affect a large proportion of the population. Interleukin-13 (IL-13)-expressing follicular helper T (TFH13) cells are a newly identified population of TFH cells that have been associated with high-affinity IgE responses. However, the origins, developmental signals, transcriptional programming and precise functions of TFH13 cells are unknown. Here, we examined the developmental signals for TFH13 cells and found a direct and progressive differentiation pathway marked by the production of IL-21. These two pathways differed in kinetics and extrinsic requirements. However, both pathways converged, forming transcriptionally similar TFH13 cells that express the transcription factor JunB as a critical stabilizing factor. Using an intersectional genetics-based TFH13-diphtheria toxin receptor model to perturb these cells, we found that TFH13 cells were essential to drive broad germinal center responses and allergen-specific IgG and IgE. Moreover, we found that IL-21 is a broad positive regulator of allergen germinal center B cells and synergizes with IL-13 produced by TFH13 cells to amplify allergic responses. Thus, TFH13 cells orchestrate multiple features of allergic inflammation.
Collapse
Affiliation(s)
- Pragya Chandrakar
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cody S Nelson
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Manuel A Podestà
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Unit of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cecilia B Cavazzoni
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maya Gempler
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeong-Mi Lee
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sierra Richardson
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hengcheng Zhang
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Snigdha Samarpita
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Ciofani
- Department of Integrative Immunology, Duke University Medical Center, Durham, NC, USA
| | - Talal Chatila
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vijay K Kuchroo
- Gene Lay Institute of Immunology and Inflammatory Diseases, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Peter T Sage
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Moshood AY, Abdulraheem MI, Li L, Zhang Y, Raghavan V, Hu J. Deciphering nutrient stress in plants: integrative insight from metabolomics and proteomics. Funct Integr Genomics 2025; 25:38. [PMID: 39955391 DOI: 10.1007/s10142-025-01551-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: 12/24/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
To comprehend the responses and resilience of plants under unfavorable environmental conditions, it is crucial to study the metabolomics and proteomic insights into nutrient stress. Nutrient stress substantially challenges agriculture, impacting plant growth, development, and productivity due to a lack or imbalance of essential nutrients, which can happen due to poor soil quality, limited nutrient availability, or unfavorable climatic conditions. Although there has been significant progress in the study of plant nutrient stress using metabolomics and proteomics, several challenges and research gaps still need to be addressed, such as the standardized experimental protocols, data integration strategies, and bioinformatic tools are necessary for comparative analysis and interpretation of omics data. Hence, this review explores the theoretical frameworks of metabolomics and proteomics as powerful tools to decode plant responses to nutrient stress, addressing critical knowledge gaps in the field. This review highlights the advantages of integrative analyses, combining metabolomics, proteomics, and transcriptomics, to uncover the molecular networks governing nutrient stress resilience. Key findings underscore the potential of these techniques to enhance breeding strategies and genetic engineering efforts aimed at developing nutrient-efficient crops. Through metabolomics and proteomic analyses, novel molecular components and regulatory networks have been revealed as responsive to nutrient stress, and this breakthrough has the potential to bolster plant resilience and optimize nutrient utilization. Understanding the synergistic roles of metabolites and proteins in nutrient stress resilience has profound implications for crop improvement and agricultural sustainability. Future research should focus on refining integrative methodologies and exploring their applications across diverse plant species and environmental conditions, paving the way for innovative solutions to nutrient stress challenges.
Collapse
Affiliation(s)
- Abiodun Yusuff Moshood
- Department of Electrical Engineering, Henan Agricultural University, Zhengzhou, 450002, China
- Henan International Joint Laboratory of Laser Technology in Agriculture Science, Zhengzhou, 450002, China
| | - Mukhtar Iderawumi Abdulraheem
- Department of Electrical Engineering, Henan Agricultural University, Zhengzhou, 450002, China.
- Henan International Joint Laboratory of Laser Technology in Agriculture Science, Zhengzhou, 450002, China.
- Department of Agricultural Science, Oyo State College of Education, Lanlate, 202001, Nigeria.
| | - Linze Li
- Department of Electrical Engineering, Henan Agricultural University, Zhengzhou, 450002, China
- Henan International Joint Laboratory of Laser Technology in Agriculture Science, Zhengzhou, 450002, China
| | - Yanyan Zhang
- Department of Electrical Engineering, Henan Agricultural University, Zhengzhou, 450002, China
| | - Vijaya Raghavan
- Department of Bioresource Engineering, Faculty of Agriculture and Environmental Studies, McGill University, Sainte- Anne-de-Bellevue, QC, H9X 3V9, Canada
| | - Jiandong Hu
- Henan International Joint Laboratory of Laser Technology in Agriculture Science, Zhengzhou, 450002, China.
- Department of Agricultural Science, Oyo State College of Education, Lanlate, 202001, Nigeria.
| |
Collapse
|
16
|
Yu P, Xiao L, Hu K, Ling J, Chen Y, Liang R, Liu X, Zhang D, Liu Y, Weng T, Jiang H, Zhang J, Wang W. Comprehensive exploration of programmed cell death landscape in lung adenocarcinoma combining multi-omic analysis and experimental verification. Sci Rep 2025; 15:5364. [PMID: 39948103 PMCID: PMC11825851 DOI: 10.1038/s41598-025-87982-w] [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: 11/02/2024] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
The mortality and therapeutic failure in lung adenocarcinoma (LUAD) are mainly resulted from the wide metastasis and chemotherapy resistance. Up to now, accurate and stable predictive prognostic indicator for revealing the progress and novel therapeutic strategies of LUAD is infrequent, nonetheless. Diversified programmed cell death (PCD) has been widely confirmed that participated in the occurrence and development of various malignant tumors, respectively. In this research, we integrated fourteen types of PCD, bulk multi-omic data from TCGA-LUAD and other cohorts in gene expression omnibus (GEO) and clinical LUAD patients to develop our analysis. Consequently, pivotal fourteen PCD genes, especially CAMP, CDK5R1, CTSW, DAPK2, GAB2, GAPDH, GATA2, HGF, MAPT, NAPSA, NUPR1, PIK3CG, PLA2G3, and SLC7A11, were utilized to establish the prognostic signature, namely cell death index (CDI). The validation in several external cohorts indicated that CDI can be regarded as a potential risk factor of LUAD patients. Combined with other common clinical information, a nomogram with potential predictive ability was constructed. Besides, according to the CDI signature, the tumor microenvironment (TME) and sensitivity to some potential chemotherapeutic drugs were further and deeply explored. Notably, verification and functional experiments further demonstrated the remarkable correlation between CDI and unfold protein response. Given all the above, a novel CDI gene signature was indicated to predict the prognosis and exploit precision therapeutic strategies of LUAD patients.
Collapse
Affiliation(s)
- Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Leyang Xiao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medical College, Nanchang University, Nanchang, China
| | - Kaibo Hu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medical College, Nanchang University, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ruiqi Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xinyu Liu
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Pok Fu Lam, Hong Kong
| | - Yuzhen Liu
- Department of Thoracic Surgery, Jiangxi Provincial Chest Hospital, Nanchang, China
| | - Tongchun Weng
- Department of Thoracic Surgery, Jiangxi Provincial Chest Hospital, Nanchang, China
| | - Hongfa Jiang
- Department of Thoracic Surgery, Jiangxi Provincial Chest Hospital, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wuming Wang
- Department of Thoracic Surgery, Jiangxi Provincial Chest Hospital, Nanchang, China.
| |
Collapse
|
17
|
Sharma P, Salunke A, Pandya N, Shah H, Pandya P, Parikh P. De novo Transcriptomic analysis to unveil the deltamethrin induced resistance mechanisms in Callosobruchus chinensis (L.). Sci Rep 2025; 15:5163. [PMID: 39939732 PMCID: PMC11822196 DOI: 10.1038/s41598-025-89466-3] [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: 07/16/2024] [Accepted: 02/04/2025] [Indexed: 02/14/2025] Open
Abstract
The use of synthetic insecticides has been crucial in the management of insect pests however the extensive use of insecticides can result in the development of resistance. Callosobruchus chinensis is a highly destructive pest of stored grains, it's a major feeder and infests a range of stored grains that are vital to both global food security and human nutrition. We extensively investigated gene expression changes of adults in response to deltamethrin to decipher the mechanism behind the insecticide resistance. The analysis of gene expression revealed 25,343 unigenes with a mean length of 1,435 bp. All the expressed genes were identified, and analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment. Exposure to deltamethrin (4.6 ppm) causes 320 differentially expressed genes (DEGs), of which 280 down-regulated and 50 up-regulated. The transcriptome analysis revealed that DEGs were found to be enriched in pathways related to xenobiotics metabolism, signal transduction, cellular processes, organismal systems and information processing. The quantitative real-time PCR was used to validate the DEGs encoding metabolic detoxification. To the best of our knowledge, these results offer the first toxicity mechanisms enabling a more comprehensive comprehension of the action and detoxification of deltamethrin in C. chinensis.
Collapse
Affiliation(s)
- Pankaj Sharma
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India, 390002
| | - Ankita Salunke
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India, 390002
| | - Nishi Pandya
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India, 390002
| | - Hetvi Shah
- Department of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India, 391410
| | - Parth Pandya
- Department of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India, 391410.
| | - Pragna Parikh
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India, 390002.
| |
Collapse
|
18
|
Gibbs JR, Mei C, Wunderlich Z. Beyond the heat shock pathway: Heat stress responses in Drosophila development. Dev Biol 2025; 518:53-60. [PMID: 39557149 PMCID: PMC11703687 DOI: 10.1016/j.ydbio.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
Heat stress has broad effects on an organism and is an inevitable part of life. Embryos face a particular challenge when faced with heat stress - the intricate molecular processes that pattern the embryo can all be affected by heat, and the embryo lacks some of the strategies that adults can use to manage or avoid heat stress. We use Drosophila melanogaster as a model, as insects are capable of developing normally under a wide range of temperatures and are exposed to daily temperature swings as they develop. Research has focused on the heat shock pathway and the transcription of heat shock proteins as the main response to heat and heat damage. This review explores embryonic heat responses beyond the heat shock pathway. We examine the effects of heat from a biochemical standpoint, as well as highlighting other mechanisms of heat stress regulation, such as miRNA activity or other signaling pathways. We discuss how different elements of the heat stress response must be coordinated across the embryo to enable development under a wide range of temperatures. Studying heat stress in Drosophila melanogaster can be a powerful lens into how developmental systems ensure robustness to environmental factors.
Collapse
Affiliation(s)
- Julia R Gibbs
- Department of Biology, Biological Design Center, Boston University, Boston, MA, 02215, USA
| | - Christian Mei
- Department of Biology, Biological Design Center, Boston University, Boston, MA, 02215, USA
| | - Zeba Wunderlich
- Department of Biology, Biological Design Center, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
19
|
Jochim BE, Topalidou I, Lehrbach NJ. Protein sequence editing defines distinct and overlapping functions of SKN-1A/Nrf1 and SKN-1C/Nrf2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635299. [PMID: 39975340 PMCID: PMC11838306 DOI: 10.1101/2025.01.29.635299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The Nrf/NFE2L family of transcription factors regulates redox balance, xenobiotic detoxification, metabolism, proteostasis, and aging. Nrf1/NFE2L1 is primarily responsible for stress-responsive upregulation of proteasome subunit genes and is essential for adaptation to proteotoxic stress. Nrf2/NFE2L2 is mainly involved in activating oxidative stress responses and promoting xenobiotic detoxification. Nrf1 and Nrf2 contain very similar DNA binding domains and can drive similar transcriptional responses. In C. elegans, a single gene, skn-1, encodes distinct protein isoforms, SKN-1A and SKN-1C, that function analogously to mammalian Nrf1 and Nrf2, respectively, and share an identical DNA binding domain. Thus, the extent to which SKN-1A/Nrf1 and SKN-1C/Nrf2 functions are distinct or overlapping has been unclear. Regulation of the proteasome by SKN-1A/Nrf1 requires post-translational conversion of N-glycosylated asparagine residues to aspartate by the PNG-1/NGLY1 peptide:N-glycanase, a process we term 'sequence editing'. Here, we reveal the consequences of sequence editing for the transcriptomic output of activated SKN-1A. We confirm that activation of proteasome subunit genes is strictly dependent on sequence editing. In addition, we find that sequence edited SKN-1A can also activate genes linked to redox homeostasis and xenobiotic detoxification that are also regulated by SKN-1C, but the extent of these genes' activation is antagonized by sequence editing. Using mutant alleles that selectively inactivate either SKN-1A or SKN-1C, we show that both isoforms promote optimal oxidative stress resistance, acting as effectors for distinct signaling pathways. These findings suggest that sequence editing governs SKN-1/Nrf functions by tuning the SKN-1A/Nrf1 regulated transcriptome.
Collapse
|
20
|
Zhang S, Wang N, Gao Z, Gao J, Wang X, Xie H, Wang CY, Zhang S. Reductive stress: The key pathway in metabolic disorders induced by overnutrition. J Adv Res 2025:S2090-1232(25)00031-1. [PMID: 39805424 DOI: 10.1016/j.jare.2025.01.012] [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: 09/01/2024] [Revised: 12/04/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The balance of redox states is crucial for maintaining physiological homeostasis. For decades, the focus has been mainly on the concept of oxidative stress, which is involved in the mechanism of almost all diseases. However, robust evidence has highlighted that reductive stress, the other side of the redox spectrum, plays a pivotal role in the development of various diseases, particularly those related to metabolism and cardiovascular health. AIM OF REVIEW In this review, we present an extensive array of evidence for the occurrence of reductive stress and its significant implications mainly in metabolic and cardiovascular diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW Reductive stress is defined as a shift in the cellular redox balance towards a more reduced state, characterized by an excess of endogenous reductants (such as NADH, NADPH, and GSH) over their oxidized counterparts (NAD+, NADP+, and GSSG). While oxidative stress has been the predominant mechanism studied in obesity, metabolic disorders, and cardiovascular diseases, growing evidence underscores the critical role of reductive stress. This review discusses how reductive stress contributes to metabolic and cardiovascular pathologies, emphasizing its effects on key cellular processes. For example, excessive NADH accumulation can disrupt mitochondrial function by impairing the electron transport chain, leading to decreased ATP production and increased production of reactive oxygen species. In the endoplasmic reticulum (ER), an excess of reductive equivalents hampers protein folding, triggering ER stress and activating the unfolded protein response, which can lead to insulin resistance and compromised cellular homeostasis. Furthermore, we explore how excessive antioxidant supplementation can exacerbate reductive stress by further shifting the redox balance, potentially undermining the beneficial effects of exercise, impairing cardiovascular health, and aggravating metabolic disorders, particularly in obese individuals. This growing body of evidence calls for a reevaluation of the role of reductive stress in disease pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- Shiyi Zhang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhichao Gao
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Gao
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohui Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Xie
- Institute of Translational Medicine, Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Cong-Yi Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shu Zhang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
21
|
Kawataki S, Kubota Y, Katayama K, Imoto S, Takekawa M. GADD45β-MTK1 signaling axis mediates oncogenic stress-induced activation of the p38 and JNK pathways. Cancer Sci 2025; 116:128-142. [PMID: 39526327 PMCID: PMC11711059 DOI: 10.1111/cas.16389] [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: 09/23/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
The ERK pathway governs essential biological processes such as cell proliferation and survival, and its hyperactivation by various oncogenes ultimately drives carcinogenesis. However, normal mammalian cells typically recognize aberrant ERK signaling as oncogenic stress and respond by inducing cell cycle arrest or apoptosis through activation of the p38 and JNK pathways. Despite the critical role of this response in preventing carcinogenesis, the precise molecular mechanisms underlying oncogene-induced, ERK-dependent activation of p38/JNK and its tumor-suppressive effects remain unclear. Here, we demonstrate that MAP three kinase 1 (MTK1), a stress-responsive MAPKKK, serves as a key mediator of p38/JNK activation induced by oncogenic ERK signaling. Mechanistically, aberrant ERK signaling induces sustained expression of the transcription factor early growth response protein 1 (EGR1), which promotes the production of the MTK1 activator GADD45β, leading to persistent activation of MTK1-p38/JNK signaling. Gene knockout and transcriptome analyses revealed that this GADD45β/MTK1-mediated cross-talk between the ERK and p38/JNK pathways preferentially upregulates a specific set of genes involved in apoptosis and the immune response. Notably, the expression of EGR1, GADD45β, and MTK1 is frequently downregulated in many cancers with high ERK activity, resulting in the disruption of the tumor-suppressive ERK-p38/JNK cross-talk. Restoring GADD45β expression in cancer cells reactivates p38/JNK signaling and suppresses tumorigenesis. Our findings delineate a molecular mechanism by which normal cells sense and respond to oncogenic stress to prevent abnormal growth, and highlight the significance of its dysregulation in cancer.
Collapse
Affiliation(s)
- Saeko Kawataki
- Division of Cell Signaling and Molecular Medicine, Institute of Medical ScienceThe University of TokyoTokyoJapan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoChibaJapan
| | - Yuji Kubota
- Division of Cell Signaling and Molecular Medicine, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Kotoe Katayama
- Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Seiya Imoto
- Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical ScienceThe University of TokyoTokyoJapan
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Mutsuhiro Takekawa
- Division of Cell Signaling and Molecular Medicine, Institute of Medical ScienceThe University of TokyoTokyoJapan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoChibaJapan
| |
Collapse
|
22
|
Gupta M, Dwivedi V, Kumar S, Patel A, Niazi P, Yadav VK. Lead toxicity in plants: mechanistic insights into toxicity, physiological responses of plants and mitigation strategies. PLANT SIGNALING & BEHAVIOR 2024; 19:2365576. [PMID: 38899525 PMCID: PMC11195469 DOI: 10.1080/15592324.2024.2365576] [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: 02/28/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Soil toxicity is a major environmental issue that leads to numerous harmful effects on plants and human beings. Every year a huge amount of Pb is dumped into the environment either from natural sources or anthropogenically. Being a heavy metal it is highly toxic and non-biodegradable but remains in the environment for a long time. It is considered a neurotoxic and exerts harmful effects on living beings. In the present review article, investigators have emphasized the side effects of Pb on the plants. Further, the authors have focused on the various sources of Pb in the environment. Investigators have emphasized the various responses including molecular, biochemical, and morphological of plants to the toxic levels of Pb. Further emphasis was given to the effect of elevated levels of Pb on the microbial population in the rhizospheres. Further, emphasized the various remediation strategies for the Pb removal from the soil and water sources.
Collapse
Affiliation(s)
- Minoti Gupta
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Chandigarh, Punjab, India
| | - Vinay Dwivedi
- Amity Institute of Biotechnology, Amity University, Gwalior, Madhya Pradesh, India
| | - Swatantar Kumar
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Chandigarh, Punjab, India
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, Gujarat, India
| | - Parwiz Niazi
- Department of Biology, Faculty of Education, Kandahar University, Kandahar, Afghanistan
- Department of Plant Protection, Faculty of Agriculture, EGE University, İzmir, Turkey
| | - Virendra Kumar Yadav
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, Gujarat, India
| |
Collapse
|
23
|
Mazurek M, Tobiasz-Salach R, Stadnik B, Migut D. Silicon-Mitigated Effect on Zinc-Induced Stress Conditions: Epigenetic, Morphological, and Physiological Screening of Barley Plants. Int J Mol Sci 2024; 26:104. [PMID: 39795961 PMCID: PMC11720124 DOI: 10.3390/ijms26010104] [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: 11/30/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Plants are increasingly exposed to stress-induced factors, including heavy metals. Zinc, although it is a microelement, at high concentrations can be phytotoxic to plants by limiting their growth and development. The presented research confirmed the inhibition effect of Zn on morphological and physiological parameters in barley plants. However, the effect was Zn dose dependent (50 µM, 100 µM, and 200 µM), as well as part of the plants (above ground or roots). To mitigate the negative effects of Zn, plants were sprayed with 0.1% silicon. Silicon was proven to have a positive effect on mitigating the inhibitory effects of Zn-induced stress. In most cases, an increase in both morphological (length, elongation, fresh and dry weights, and weather content) and physiological (relative chlorophyll content and fluorescence) parameters was observed. This occurrence was dependent on the Zn dose. Epigenetic analyses confirmed differences in the DNA methylation level, both between plants subjected to stress at different strengths (50 µM, 100 µM, and 200 µM Zn) and between plants sprayed with Si or not. The differences indicate that silicon affects the epigenome of barley plants, thereby modifying the response of plants to stress factors. This modification may be the basis for plants to acquire resistance as "epigenetic memory".
Collapse
Affiliation(s)
- Marzena Mazurek
- Department of Physiology and Plant Biotechnology, University of Rzeszow, Ćwiklińskiej 2, 35-601 Rzeszow, Poland
| | - Renata Tobiasz-Salach
- Department of Crop Production, University of Rzeszow, Zelwerowicza 4, 35-601 Rzeszow, Poland; (R.T.-S.); (B.S.); (D.M.)
| | - Barbara Stadnik
- Department of Crop Production, University of Rzeszow, Zelwerowicza 4, 35-601 Rzeszow, Poland; (R.T.-S.); (B.S.); (D.M.)
- Doctoral School, University of Rzeszow, Rejtana 16C, 35-959 Rzeszow, Poland
| | - Dagmara Migut
- Department of Crop Production, University of Rzeszow, Zelwerowicza 4, 35-601 Rzeszow, Poland; (R.T.-S.); (B.S.); (D.M.)
| |
Collapse
|
24
|
Liu W, Zhang Q, Guo S, Wang H. The role of microRNAs regulation of endoplasmic reticulum stress in ischemia-reperfusion injury: A review. Int J Biol Macromol 2024; 283:137566. [PMID: 39542287 DOI: 10.1016/j.ijbiomac.2024.137566] [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: 07/26/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
The endoplasmic reticulum (ER) is an important organelle in eukaryotic cells, responsible for a range of biological functions such as the secretion, modification and folding of proteins, maintaining Ca2+ homeostasis and the synthesis of steroids/lipids, secreted proteins and membrane proteins. When cells are affected by internal or external factors, including abnormal energy metabolism, disrupted Ca2+ balance, altered glycosylation, drug toxicity, and so on, the unfolded or misfolded proteins accumulate in the ER, leading to the unfolded protein response (UPR) and ER stress. The abnormal ER stress has been reported to be involved in various pathological processes. MicroRNAs (miRNAs) are non-coding RNAs with the length of approximately 19-25 nucleotides. They control the expression of multiple genes through posttranscriptional gene silencing in eukaryotes or some viruses. Increasing evidence indicates that miRNAs are involved in various cellular functions and biological processes, such as cell proliferation and differentiation, growth and development, and metabolic homeostasis. Hence, miRNAs participate in multiple pathological processes. Recently, many studies have shown that miRNAs play an important role by regulating ER stress in ischemia-reperfusion (I/R) injury, but the relevant mechanisms are not fully understood. In this review, we reviewed the current understanding of ER stress, as well as the biogenesis and function of miRNAs, and focused on the role of miRNAs regulation of ER stress in I/R injury, with the aim of providing new targets for the treatment of I/R injury.
Collapse
Affiliation(s)
- Wanying Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Qi Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
25
|
Ejiohuo O, Bajia D, Pawlak J, Szczepankiewicz A. Asoprisnil as a Novel Ligand Interacting with Stress-Associated Glucocorticoid Receptor. Biomedicines 2024; 12:2745. [PMID: 39767652 PMCID: PMC11726916 DOI: 10.3390/biomedicines12122745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Background/objective: The glucocorticoid receptor (GR) is critical in regulating cortisol production during stress. This makes it a key target for treating conditions associated with hypothalamic-pituitary-adrenal (HPA) axis dysregulation, such as mental disorders. This study explores novel ligands beyond mifepristone for their potential to modulate GR with improved efficacy and safety. By investigating these interactions, we seek to identify new pharmacotherapeutic options for stress-related mental illness. Methods: The ligands asoprisnil, campestanol, and stellasterol were selected based on structural similarities to mifepristone (reference ligand) and evaluated for pharmacological and ADME (absorption, distribution, metabolism, and excretion) properties using the SwissADME database. Molecular docking with AutoDock 4.2.6 and molecular dynamics simulations were performed to investigate ligand-protein interactions with the human glucocorticoid receptor, and binding free energies were calculated using MMPBSA. Results: Pharmacokinetic analysis revealed that asoprisnil exhibited high gastrointestinal absorption and obeyed Lipinski's rule, while mifepristone crossed the blood-brain barrier. Toxicological predictions showed that mifepristone was active for neurotoxicity and immunotoxicity, while asoprisnil, campestanol, and stellasterol displayed lower toxicity profiles. Asoprisnil demonstrated the highest stability in molecular dynamics simulations, with the highest negative binding energy of -62.35 kcal/mol, when compared to mifepristone, campestanol, and stellasterol, with binding energies of -57.08 kcal/mol, -49.99 kcal/mol, and -46.69 kcal/mol, respectively. Conclusion: This makes asoprisnil a potentially favourable therapeutic candidate compared to mifepristone. However, further validation of asoprisnil's interaction, efficacy, and safety in stress-related mental disorders through experimental studies and clinical trials is needed.
Collapse
Affiliation(s)
- Ovinuchi Ejiohuo
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland;
- Molecular and Cell Biology Unit, Poznan University of Medical Sciences, 60-572 Poznan, Poland;
| | - Donald Bajia
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland;
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Joanna Pawlak
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | | |
Collapse
|
26
|
Bastari G, Solar Fernandez V, Muzzi M, Moreno S, Marino M, Fiocchetti M. Neuroglobin-enriched secretome provides neuroprotection against hydrogen peroxide and mitochondrial toxin-induced cellular stress. Cell Stress 2024; 8:99-111. [PMID: 39600400 PMCID: PMC11589466 DOI: 10.15698/cst2024.11.300] [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: 02/09/2024] [Revised: 09/30/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Aberrant response to physiological cell stress is part of the mechanisms underlying the development of diverse human diseases, including neuropathologies. Neuroglobin (NGB), an intracellular monomeric globin, has gained attention for its role in endogenous stress response pathways in neuroprotection. To date, evidence supports the concept of NGB as an inducible protein, triggered by physiological and pathological stimuli via transcriptional and/or post-transcriptional mechanisms, offering cell-autonomous neuroprotective functions under various cellular stresses. Notably, recent evidence suggests the extracellular occurrence of NGB. We aimed to explore whether NGB redistribution in the cell microenvironment may serve in transmitting resilience capability in a model with neuronal characteristics. Results obtained in SH-SY5Y demonstrated that intracellular NGB upregulation is associated with the promotion of the extracellular release of the globin. Additionally, cell secretome from NGB-overexpressing cells, characterized by globin accumulation, exhibits protective effects against oxidative stress and mitochondrial toxicity, as evidenced by reduced apoptosis and preserved mitochondrial structure. These findings shed light on the potential significance of extracellular NGB as part of a common cell response to physiological and stress conditions and as a factor promoting cell resilience. Furthermore, the potential for neuroprotection of extracellular NGB paves the way for future therapeutic opportunities.
Collapse
Affiliation(s)
- Giovanna Bastari
- Department of Science, Section Biomedical Sciences and Technology, University Roma Tre, V.le G. MarconiRome, 00146Italy
| | - Virginia Solar Fernandez
- Department of Science, Section Biomedical Sciences and Technology, University Roma Tre, V.le G. MarconiRome, 00146Italy
- Present address: Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC, Arturo Duperier, 4, 28029 Madrid, Spain; Department of Biochemistry, School of Medicine, Universidad Autonóma de Madrid, 28029 Madrid, Spain
| | - Maurizio Muzzi
- Department of Science, Section Molecular, Cellular, Environmental and Evolutionary Biology, LIMERome, 00146Italy
- Department of Bioscience and Territory, University of MolisePesche, 86090Italy
| | - Sandra Moreno
- Department of Science, Section Molecular, Cellular, Environmental and Evolutionary Biology, LIMERome, 00146Italy
- IRCCS Fondazione Santa LuciaRome, 00179Italy
| | - Maria Marino
- Department of Science, Section Biomedical Sciences and Technology, University Roma Tre, V.le G. MarconiRome, 00146Italy
- IRCCS Fondazione Santa LuciaRome, 00179Italy
| | - Marco Fiocchetti
- Department of Science, Section Biomedical Sciences and Technology, University Roma Tre, V.le G. MarconiRome, 00146Italy
- IRCCS Fondazione Santa LuciaRome, 00179Italy
| |
Collapse
|
27
|
De Leon-Oliva D, Boaru DL, Minaya-Bravo AM, De Castro-Martinez P, Fraile-Martinez O, Garcia-Montero C, Cobo-Prieto D, Barrena-Blázquez S, Lopez-Gonzalez L, Albillos A, Alvarez-Mon M, Saez MA, Diaz-Pedrero R, Ortega MA. Improving understanding of ferroptosis: Molecular mechanisms, connection with cellular senescence and implications for aging. Heliyon 2024; 10:e39684. [PMID: 39553553 PMCID: PMC11564042 DOI: 10.1016/j.heliyon.2024.e39684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
In the face of cell damage, cells can initiate a response ranging from survival to death, the balance being crucial for tissue homeostasis and overall health. Cell death, in both accidental and regulated forms, plays a fundamental role in maintaining tissue homeostasis. Among the regulated mechanisms of cell death, ferroptosis has garnered attention for its iron-dependent phospholipid (PL) peroxidation and its implications in aging and age-related disorders, as well as for its therapeutic relevance. In this review, we provide an overview of the mechanisms, regulation, and physiological and pathological roles of ferroptosis. We present new insights into the relationship between ferroptosis, cellular senescence and aging, emphasizing how alterations in ferroptosis pathways contribute to aging-related tissue dysfunction. In addition, we examine the therapeutic potential of ferroptosis in aging-related diseases, offering innovative insights into future interventions aimed at mitigating the effects of aging and promoting longevity.
Collapse
Affiliation(s)
- Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Ana M. Minaya-Bravo
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - David Cobo-Prieto
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Immune System Diseases-Rheumatology Service, Central University Hospital of Defence-UAH Madrid, 28801, Alcala de Henares, Spain
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala de Henares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Agustín Albillos
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Gastroenterology and Hepatology Service, Ramón y Cajal University Hospital, University of Alcalá, IRYCIS, Network Biomedical Research Center for Liver and Digestive Diseases (CIBERehd), Carlos III Health Institute, Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806, Alcala de Henares, Spain
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, 28801, Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala de Henares, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| |
Collapse
|
28
|
Li L, Chang J, Xu Z, Chu L, Zhang J, Xing Q, Bao Z. Functional allocation of Mitogen-activated protein kinases (MAPKs) unveils thermotolerance in scallop Argopecten irradians irradians. MARINE ENVIRONMENTAL RESEARCH 2024; 202:106750. [PMID: 39293275 DOI: 10.1016/j.marenvres.2024.106750] [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: 08/13/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Global warming has significantly impacted agriculture, particularly in animal husbandry and aquaculture industry. Rising ocean temperatures due to global warming are severely affecting shellfish production, necessitating an understanding of how shellfish cope with thermal stress. The mitogen-activated protein kinases (MAPK) signaling pathway plays a crucial role in cell growth, differentiation, adaptation to environmental stress, inflammatory response, and managing high temperature stress. To investigate the function of MAPKs in bay scallops, a comparative genomics and bioinformatics approach identified three MAPK genes: AiERK, Aip38, and AiJNK. Structural and phylogenetic analyses of these proteins were conducted to determine their evolutionary relationships. Spatiotemporal expression patterns were examined at different developmental stages and in various tissues of healthy adult scallops. Additionally, the expression regulation of these genes was studied in selected tissues (hemocyte, gill, heart, mantle) following exposure to high temperatures (32 °C) for different durations (0 h, 6 h, 12 h, 24 h, 3 d, 6 d, 10 d). The spatiotemporal expressions of AiMAPKs were ubiquitous, with significant increases in AiERK expression observed at the umbo larval stage (3.09-fold), while Aip38 and AiJNK were identified as potential maternal effect genes. In adult scallops, different gene expression patterns of AiMAPKs were observed across eight tissues, with high expressions in the foot and gill, and lower expressions in the striated muscle. Following high temperature stress, AiMAPKs expressions in the gill and mantle were mainly up-regulated, while in the hemocyte, they were primarily down-regulated. These findings indicate time- and tissue-dependent expression patterns with functional allocation in response to different thermal durations. This study enhances our understanding of the function and evolution of AiMAPKs genes in shellfish and provides a theoretical basis for elucidating the energy regulation mechanism of bay scallops in response to high temperature stress.
Collapse
Affiliation(s)
- Linshu Li
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Jiaxi Chang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Zhaosong Xu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Longfei Chu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Junhao Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Qiang Xing
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| | - Zhenmin Bao
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| |
Collapse
|
29
|
Xu Y, Shen Y, Zhang C, Zheng L, Ji F, Chen J, Cheng S, Zheng Y. Exploring the Effect of Fidgetin-Like 1 on Colorectal Cancer Through Tissue Chip and In Vitro Experiments. Balkan Med J 2024; 41:491-498. [PMID: 39319820 PMCID: PMC11589218 DOI: 10.4274/balkanmedj.galenos.2024.2024-7-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024] Open
Abstract
Background Fidgetin-like 1 (FIGNL1) is extensively overexpressed in a variety of cancers. It facilitates non‑small cell lung cancer tumor cell proliferation and hepatocellular carcinoma formation due to abnormal DNA repair. Clinically relevant data indicates that its high expression is linked with the poor prognosis of patients with renal clear-cell carcinoma, low-grade gliomas, and hepatocellular carcinoma. Nevertheless, the scope of FIGNL1’s involvement in cancer, particularly colorectal cancer (CRC), remains unclear. Aims To investigate the function of FIGNL1 in CRC. Study Design Cell culture study. Methods The TCGA database and immunohistochemistry analysis were employed to investigate FIGNL1 expression in CRC tissue. A cell viability assay was performed using the Cell Counting Kit-8. The cell migration and invasion were evaluated using the transwell assay. Small interfering RNA (siRNA) transfection was conducted to knockdown FIGNL1 expression. Infection with FIGNL1 overexpression lentivirus was performed to promote FIGNL1 overexpression. The STRING database was employed for predicting protein interaction. Results FIGNL1 was substantially upregulated in human CRC tissues and was associated with TNM stages and lymph node metastasis in patients. The inhibition of CRC cell proliferation, migration, and invasion in Caco-2 cells was achieved by silencing FIGNL1 using siRNA. Additional investigations suggested that FIGNL1 overexpression could promote CRC cell proliferation, migration, and invasion via P38 signaling pathway activation in Colo-205 cells. Subsequent experiments demonstrated that FIGNL1-mediated P38 phosphorylation was contingent upon SPIDR interaction. Conclusion These results implied that FIGNL1 was a potential anticancer drug target, which also offered a novel strategy for future CRC treatment.
Collapse
Affiliation(s)
- Yunxing Xu
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Yucheng Shen
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Chen Zhang
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Liangfeng Zheng
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Feiyue Ji
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Jin Chen
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Shouliang Cheng
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| | - Yu Zheng
- Clinic of Central Laboratory, Hai’an City People’s Hospital of Jiangsu Province, Hai’an Hospital Affiliated to Nantong University, Nantong, China
| |
Collapse
|
30
|
Lu J, Yuan H, Liu S, Liu Y, Qin Z, Han W, Zhang R. Gene coexpression network analysis reveals the genes and pathways in pectoralis major muscle and liver associated with wooden breast in broilers. Poult Sci 2024; 103:104056. [PMID: 39094498 PMCID: PMC11342257 DOI: 10.1016/j.psj.2024.104056] [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: 04/08/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 08/04/2024] Open
Abstract
Wooden breast (WB) is a myopathy mainly affecting pectoralis major (PM) muscle in modern commercial broiler chickens, causing enormous economic losses in the poultry industry. Recent studies have observed hepatic and PM muscle injury in broilers affected by WB, but the relationships between WB and the 2 tissues are mostly unclear. In the current study, the RNA-seq raw data of PM muscle and liver were downloaded from GSE144000, and we constructed the gene coexpression networks of PM muscle and liver to explore the relationships between WB and the 2 tissues using the weighted gene coexpression network analysis (WGCNA) method. Six and 2 gene coexpression modules were significantly correlated with WB in the PM muscle and liver networks, respectively. TGF-beta signaling, Toll-like receptor signaling and mTOR signaling pathways were significantly enriched in the genes within the 6 gene modules of PM muscle network. Meanwhile, mTOR signaling pathway was significantly enriched in the genes within the 2 gene modules of liver network. In the consensus gene coexpression network across the 2 tissues, salmon module (r = -0.5 and p = 0.05) was significantly negatively correlated with WB, in which Toll-like receptor signaling, apoptosis, and autophagy pathways were significantly enriched. The genes related with the 3 pathways, myeloid differentiation primary response 88 (MYD88), interferon regulatory factor 7 (IRF7), mitogen-activated protein kinase 14 (MAPK14), FBJ murine osteosarcoma viral oncogene homolog (FOS), jun proto-oncogene (JUN), caspase-10, unc-51 like autophagy activating kinase 2 (ULK2) and serine/threonine kinase 11 (LKB1), were identified in salmon module. In this current study, we found that the signaling pathways related with cell inflammation, apoptosis and autophagy might influence WB across 2 tissues in broilers.
Collapse
Affiliation(s)
- Jun Lu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Hui Yuan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China.
| | - Shengnan Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Yuan Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Ziwen Qin
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Wenpeng Han
- Department of Biotechnology, Jieyang Polytechnic, Jieyang City 522000, Guangdong Province, China
| | - Runxiang Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| |
Collapse
|
31
|
Tripathi S, Kharkwal G, Mishra R, Singh G. Nuclear factor erythroid 2-related factor 2 (Nrf2) signaling in heavy metals-induced oxidative stress. Heliyon 2024; 10:e37545. [PMID: 39309893 PMCID: PMC11416300 DOI: 10.1016/j.heliyon.2024.e37545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Organisms encounter reactive oxidants through intrinsic metabolism and environmental exposure to toxicants. Reactive oxygen and nitrogen species (ROS, RNS) are generally considered detrimental because they induce oxidative stress. In order to combat oxidative stress, a potential modulator of cellular defense nuclear factor erythroid 2-related factor 2 (Nrf2) and its endogenous inhibitor Kelch-like ECH-associated protein 1 (Keap1) operate as a common, genetically preserved intrinsic defense system. There has been a significant increase in the amount of harmful metalloids and metals that individuals are exposed to through their food, water, and air, primarily due to human activities. Many studies have looked at the connection between the emergence of different ailments in humans and ecological exposure to metalloids, i.e., arsenic (As) and metals viz., chromium (Cr), mercury (Hg), cadmium (Cd), cobalt (Co), and lead (Pb). It is known that they can produce ROS in several organs by both direct and indirect means. Studies suggest that Nrf2 signaling is a crucial mechanism in maintaining antioxidant balance and can have two roles, depending on the particular biological setting. From one perspective, Nrf2 is an essential defense mechanism against metal-induced toxicity. Still, it may also operate as a catalyst for metal-induced carcinogenesis in situations involving protracted exposure and persistent activation. Therefore, this review aims to provide an overview of the antioxidant defense mechanism of Nrf2-Keap1 signaling and the interrelation between Nrf2 signaling and the toxic elements.
Collapse
Affiliation(s)
- Swapnil Tripathi
- Toxicology Department, ICMR-National Institute of Occupational Health, Ahmedabad-380016, India
- Department of Biochemistry & Forensic Science, Gujarat University, Ahmedabad - 380009, India
| | - Gitika Kharkwal
- Toxicology Department, ICMR-National Institute of Occupational Health, Ahmedabad-380016, India
| | - Rajeev Mishra
- Department of Life Sciences & Biotechnology, Chhatrapati Shahu Ji Maharaj University Kanpur - 208024, India
| | - Gyanendra Singh
- Toxicology Department, ICMR-National Institute of Occupational Health, Ahmedabad-380016, India
| |
Collapse
|
32
|
Mo YY, Han YX, Xu SN, Jiang HL, Wu HX, Cai JM, Li L, Bu YH, Xiao F, Liang HD, Wen Y, Liu YZ, Yin YL, Zhou HD. Adipose Tissue Plasticity: A Comprehensive Definition and Multidimensional Insight. Biomolecules 2024; 14:1223. [PMID: 39456156 PMCID: PMC11505740 DOI: 10.3390/biom14101223] [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: 08/18/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Adipose tissue is composed of adipocytes, stromal vascular fraction, nerves, surrounding immune cells, and the extracellular matrix. Under various physiological or pathological conditions, adipose tissue shifts cellular composition, lipid storage, and organelle dynamics to respond to the stress; this remodeling is called "adipose tissue plasticity". Adipose tissue plasticity includes changes in the size, species, number, lipid storage capacity, and differentiation function of adipocytes, as well as alterations in the distribution and cellular composition of adipose tissue. This plasticity has a major role in growth, obesity, organismal protection, and internal environmental homeostasis. Moreover, certain thresholds exist for this plasticity with significant individualized differences. Here, we comprehensively elaborate on the specific connotation of adipose tissue plasticity and the relationship between this plasticity and the development of many diseases. Meanwhile, we summarize possible strategies for treating obesity in response to adipose tissue plasticity, intending to provide new insights into the dynamic changes in adipose tissue and contribute new ideas to relevant clinical problems.
Collapse
Affiliation(s)
- Yu-Yao Mo
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Xin Han
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Shi-Na Xu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hong-Li Jiang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hui-Xuan Wu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Jun-Min Cai
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Long Li
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yan-Hong Bu
- Department of Blood Transfusion, The Second Xiangya Hospital, Central South University, Changsha 410012, China;
| | - Fen Xiao
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Han-Dan Liang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Ying Wen
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Ze Liu
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China;
| | - Yu-Long Yin
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| |
Collapse
|
33
|
Skeens E, Maschietto F, Manjula R, Shillingford S, Lolis EJ, Batista VS, Bennett AM, Lisi GP. Dynamic and structural insights into allosteric regulation on MKP5 a dual-specificity phosphatase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611520. [PMID: 39282375 PMCID: PMC11398491 DOI: 10.1101/2024.09.05.611520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Dual-specificity mitogen-activated protein kinase (MAPK) phosphatases (MKPs) directly dephosphorylate and inactivate the MAPKs. Although the catalytic mechanism of dephosphorylation of the MAPKs by the MKPs is established, a complete molecular picture of the regulatory interplay between the MAPKs and MKPs still remains to be fully explored. Here, we sought to define the molecular mechanism of MKP5 regulation through an allosteric site within its catalytic domain. We demonstrate using crystallographic and NMR spectroscopy approaches that residue Y435 is required to maintain the structural integrity of the allosteric pocket. Along with molecular dynamics simulations, these data provide insight into how changes in the allosteric pocket propagate conformational flexibility in the surrounding loops to reorganize catalytically crucial residues in the active site. Furthermore, Y435 contributes to the interaction with p38 MAPK and JNK, thereby promoting dephosphorylation. Collectively, these results highlight the role of Y435 in the allosteric site as a novel mode of MKP5 regulation by p38 MAPK and JNK.
Collapse
Affiliation(s)
- Erin Skeens
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | | | - Ramu Manjula
- Department of Pharmacology, Yale School of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Shanelle Shillingford
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
- Department of Pharmacology, Yale School of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Elias J. Lolis
- Department of Pharmacology, Yale School of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Victor S. Batista
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
| | - Anton M. Bennett
- Department of Pharmacology, Yale School of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| | - George P. Lisi
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
34
|
Wang B, Ma Y, Zhang Y, Yin X. Therapeutic potential of ASK1 activators in cancer treatment: Current insights and future directions. Biomed Pharmacother 2024; 178:117214. [PMID: 39079264 DOI: 10.1016/j.biopha.2024.117214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/18/2024] [Accepted: 07/26/2024] [Indexed: 08/25/2024] Open
Abstract
Apoptosis signal-regulated kinase 1 (ASK1) is a member of the mitogen-activated protein kinase kinase (MAP3K) family, whose activation and regulation are intricately associated with apoptosis. ASK1 is activated in response to oxidative stress, among other stimuli, subsequently triggering downstream JNK, p38 MAPK, and mitochondria-dependent apoptotic signaling, which participate in the initiation of tumor cell apoptosis induced by various stimuli. Research has shown that ASK1 plays a crucial role in the apoptosis of lung cancer, breast cancer, and liver cancer cells. Currently, the investigation of effective ASK1 activators is a hot topic in research on tumor cell apoptosis. Synthetic compounds such as human β-defensin, triazolothiazide derivatives and heat shock protein 27 inhibitors; natural compounds such as quercetin, Laminarina japonica polysaccharide-1 peptide and theabrownin; and nanomedicines such as cerium oxide nanoparticles, magnetite FeO nanoparticles and silver nanoparticles can activate ASK1 and induce apoptosis in various tumor cells. This review extensively investigates the roles and activation mechanisms of ASK1, explores its impact on a variety of apoptotic signaling pathways, and discusses the potential therapeutic applications of various ASK1 activators in cancer treatment. In addition, this paper provides an in-depth discussion of the future development of this field and proposes a promising method for further research and clinical progress.
Collapse
Affiliation(s)
- Bo Wang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun 130103, China
| | - Ying Ma
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun 130103, China
| | - Yue Zhang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun 130103, China.
| | - Xunzhe Yin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| |
Collapse
|
35
|
Brockmöller S, Seeger T, Worek F, Rothmiller S. Cell-Sonar, a Novel Method for Intracellular Tracking of Secretory Pathways. Cells 2024; 13:1449. [PMID: 39273021 PMCID: PMC11394445 DOI: 10.3390/cells13171449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Intracellular tracking is commonly used in trafficking research. Until today, the respective techniques have remained complex, and complicated, mostly transgenic target protein changes are necessary, often requiring expensive equipment and expert knowledge. METHODS We present a novel method, which we term "cell-sonar", that enables the user to track expression changes of specific protein markers that serve as points of interaction. Our study provides comparable analyses of expression changes of these marker proteins by in-cell Western analyses in two otherwise isogenic cell lines that only differ in the overexpression of the tracked target protein. Using the overexpressed human adult muscle-type nicotinic acetylcholine receptor as an example, we demonstrate that cell-sonar can cover multiple intracellular compartments such as the endoplasmic reticulum, the pathway between it and the Golgi apparatus, and the endocytic pathway. RESULTS We provide evidence for receptor maturation in the Golgi and storage in recycling endosomes, rather than the fate of increased insertion into the plasma membrane. Additionally, we demonstrate with the implementation of nicotine that the receptor's destiny is exasperated up to secondary degradation. CONCLUSIONS Cell-sonar is an affordable, easy-to-implement, and cheap method that can be adapted to a broad variety of proteins and cellular pathways of interest to researchers.
Collapse
Affiliation(s)
- Sabrina Brockmöller
- Bundeswehr Institute of Pharmacology and Toxicology, 80937 Munich, Bavaria, Germany
| | | | | | | |
Collapse
|
36
|
Zhao Y, Li R, Wang W, Zhang H, Zhang Q, Jiang J, Wang Y, Li Y, Guan F, Nie Y. O-GlcNAc signaling: Implications for stress-induced adaptive response pathway in the tumor microenvironment. Cancer Lett 2024; 598:217101. [PMID: 38969156 DOI: 10.1016/j.canlet.2024.217101] [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: 04/16/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
The tumor microenvironment (TME) consists of tumor cells, non-tumor cells, extracellular matrix, and signaling molecules, which can contribute to tumor initiation, progression, and therapy resistance. In response to starvation, hypoxia, and drug treatments, tumor cells undergo a variety of deleterious endogenous stresses, such as hypoxia, DNA damage, and oxidative stress. In this context, to survive the difficult situation, tumor cells evolve multiple conserved adaptive responses, including metabolic reprogramming, DNA damage checkpoints, homologous recombination, up-regulated antioxidant pathways, and activated unfolded protein responses. In the last decades, the protein O-GlcNAcylation has emerged as a crucial causative link between glucose metabolism and tumor progression. Here, we discuss the relevant pathways that regulate the above responses. These pathways are adaptive adjustments induced by endogenous stresses in cells. In addition, we systematically discuss the role of O-GlcNAcylation-regulated stress-induced adaptive response pathways (SARPs) in TME remodeling, tumor progression, and treatment resistance. We also emphasize targeting O-GlcNAcylation through compounds that modulate OGT or OGA activity to inhibit tumor progression. It seems that targeting O-GlcNAcylated proteins to intervene in TME may be a novel approach to improve tumor prognosis.
Collapse
Affiliation(s)
- Yu Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Renlong Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Weizhen Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Haohao Zhang
- Department of Digestive Surgery, Honghui Hospital, Xi'an Jiaotong University, 710054, Xi'an, Shaanxi, China
| | - Qiujin Zhang
- Second Clinical Medicine College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jialu Jiang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Ying Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Yan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Feng Guan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China.
| | - Yongzhan Nie
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China; State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
37
|
Fukushima N, Miyamoto Y, Yamauchi J. Knockdown of Rab9 Recovers Defective Morphological Differentiation Induced by Chemical ER Stress Inducer or PMD-Associated PLP1 Mutant Protein in FBD-102b Cells. PATHOPHYSIOLOGY 2024; 31:420-435. [PMID: 39311306 PMCID: PMC11417737 DOI: 10.3390/pathophysiology31030032] [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: 05/29/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/26/2024] Open
Abstract
Small GTP-binding proteins of the Rab family regulate intracellular vesicle trafficking across many aspects of the transport system. Among these, Rab9 is recognized for its role in controlling the transport system not only around the trans-Golgi network but also around the late endosome. However, the specific functions across different cell types and tissues remain unclear. Here, for the first time, we report that Rab9 negatively regulates morphological changes in the FBD-102b cell line, an oligodendroglial precursor cell line undergoing morphological differentiation. The knockdown of Rab9 led to an increase in cell shape alterations characterized by widespread membrane extensions. These changes were accompanied by increased expression levels of oligodendroglial cell differentiation and myelination marker proteins. Notably, the knockdown of Rab9 was capable of recovering defective cell morphological changes induced by tunicamycin, an inducer of endoplasmic reticulum (ER) stress, which is one of the major causes of oligodendroglial cell diseases such as Pelizaeus-Merzbacher disease (PMD, currently known as hypomyelinating leukodystrophy type 1 [HLD1]). In addition, Rab9 knockdown recovered levels of ER stress marker proteins and differentiation markers. Similar results were obtained in the cases of dithiothreitol (DTT), another chemical ER stress inducer, as well as HLD1-associated proteolipid protein 1 (PLP1) mutant protein. These results indicate a unique role for Rab9 in oligodendroglial cell morphological changes, suggesting its potential as a therapeutic target for mitigating diseases such as HLD1 at the molecular and cellular levels.
Collapse
Affiliation(s)
- Nana Fukushima
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (N.F.); (Y.M.)
| | - Yuki Miyamoto
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (N.F.); (Y.M.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (N.F.); (Y.M.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
38
|
Silva GN, Brandão VGA, Perez MV, Blum K, Lewandrowski KU, Fiorelli RKA. Neuroinflammatory Approach to Surgical Trauma: Biomarkers and Mechanisms of Immune and Neuroendocrine Responses. J Pers Med 2024; 14:829. [PMID: 39202020 PMCID: PMC11355628 DOI: 10.3390/jpm14080829] [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] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
The severity and invasiveness of clinical outcomes from organic responses to trauma are influenced by individual, surgical, and anesthetic factors. A stress response elicits neuroendocrine and immune reactions that may lead to multi-organ dysfunction. The degree of neuroinflammatory reflex activation from trauma can increase pro-inflammatory cytokine production, leading to endothelial dysfunction, glycocalyx damage, neutrophil activation, and multisystem tissue destruction. A shift in patient treatment towards a neuroinflammatory perspective has prompted a new evaluation protocol for surgical patients, required to understand surgical pathogenesis and its link to chosen anesthetic-surgical methods. The goal of this study is to summarize and disseminate the present knowledge about the mechanisms involved in immune and neuroendocrine responses, focusing on video laparoscopic surgeries. This article outlines various measures cited in the literature aimed at reducing the burden of surgical trauma. It reviews anesthetic drugs, anesthetic techniques, and intensive care procedures that are known to have immunomodulatory effects. The results show a preference for more sensitive inflammatory mediators to tissue trauma serving as care tools, indicators for prognosis, and therapeutic outcomes.
Collapse
Affiliation(s)
- Gustavo N. Silva
- Department of Anesthesiology, Gaffrée e Guinle Universitary Hospital (EBSERH), Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro 22290-240, RJ, Brazil;
| | - Virna G. A. Brandão
- Department of Anesthesiology, Gaffrée e Guinle Universitary Hospital (EBSERH), Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro 22290-240, RJ, Brazil;
| | - Marcelo V. Perez
- Department of Surgery and Anesthesia, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, SP, Brazil;
| | - Kenneth Blum
- Division of Addiction Research & Education, Center for Sports, Exercise & Mental Health, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Kai-Uwe Lewandrowski
- Center for Advanced Spine Care of Southern Arizona and Surgical Institute of Tucson, Tucson, AZ 85712, USA;
| | - Rossano K. A. Fiorelli
- Department of General and Specialized Surgery, Gaffrée e Guinle Universitary Hospital (EBSERH), Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro 22290-240, RJ, Brazil;
| |
Collapse
|
39
|
Shi Z, Han Z, Chen J, Zhou JC. Endoplasmic reticulum-resident selenoproteins and their roles in glucose and lipid metabolic disorders. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167246. [PMID: 38763408 DOI: 10.1016/j.bbadis.2024.167246] [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: 03/13/2024] [Revised: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024]
Abstract
Glucose and lipid metabolic disorders (GLMDs), such as diabetes, dyslipidemia, metabolic syndrome, nonalcoholic fatty liver disease, and obesity, are significant public health issues that negatively impact human health. The endoplasmic reticulum (ER) plays a crucial role at the cellular level for lipid and sterol biosynthesis, intracellular calcium storage, and protein post-translational modifications. Imbalance and dysfunction of the ER can affect glucose and lipid metabolism. As an essential trace element, selenium contributes to various human physiological functions mainly through 25 types of selenoproteins (SELENOs). At least 10 SELENOs, with experimental and/or computational evidence, are predominantly found on the ER membrane or within its lumen. Two iodothyronine deiodinases (DIOs), DIO1 and DIO2, regulate the thyroid hormone deiodination in the thyroid and some external thyroid tissues, influencing glucose and lipid metabolism. Most of the other eight members maintain redox homeostasis in the ER. Especially, SELENOF, SELENOM, and SELENOS are involved in unfolded protein responses; SELENOI catalyzes phosphatidylethanolamine synthesis; SELENOK, SELENON, and SELENOT participate in calcium homeostasis regulation; and the biological significance of thioredoxin reductase 3 in the ER remains unexplored despite its established function in the thioredoxin system. This review examines recent research advances regarding ER SELENOs in GLMDs and aims to provide insights on ER-related pathology through SELENOs regulation.
Collapse
Affiliation(s)
- Zhan Shi
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Ziyu Han
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Jingyi Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Ji-Chang Zhou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; Guangdong Provincial Engineering Laboratory for Nutrition Translation, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China.
| |
Collapse
|
40
|
Wang F, Wang RY, Zhong DB, Zhao P, Xia QY. Highly efficient expression of human extracellular superoxide dismutase (rhEcSOD) with ultraviolet-B-induced damage-resistance activity in transgenic silkworm cocoons. INSECT SCIENCE 2024; 31:1150-1164. [PMID: 38010045 DOI: 10.1111/1744-7917.13289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/20/2023] [Accepted: 09/24/2023] [Indexed: 11/29/2023]
Abstract
Extracellular superoxide dismutase (EcSOD) protects tissues from oxidative stress, and thus is considered as a therapeutic agent for many diseases such as atherosclerosis, hypertension, and cancer. However, cost-effective production of bioactive recombinant human EcSOD (rhEcSOD) remains a challenge. Herein, we developed an efficient strategy for producing active rhEcSOD by transgenic silkworms. rhEcSOD was successfully synthesized as homodimers and homotetramers in the middle silk gland and spun into the cocoons with a concentration of 9.48 ± 0.21 mg/g. Purification of rhEcSOD from the cocoons could be conveniently achieved with a purity of 99.50% and a yield of 3.5 ± 0.5 mg/g. Additionally, N-glycosylation at the only site of N89 in rhEcSOD with 10 types were identified. The purified rhEcSOD gained the potent enzymatic activity of 4 162 ± 293 U/mg after Cu/Zn ions incorporation. More importantly, rhEcSOD was capable of penetrating and accumulating in the nuclei of cells to maintain cell morphology and attenuate ultraviolet B-induced cell apoptosis by eliminating reactive oxygen species and inhibiting the C-Jun N-terminal kinase signaling pathway. These results demonstrated that the transgenic silkworm could successfully produce rhEcSOD with enzymatic and biological activities for biomedical applications.
Collapse
Affiliation(s)
- Feng Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
| | - Ri-Yuan Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
| | - De-Bin Zhong
- Century Legend Biotechnology Research Institute (Chongqing) Co., Ltd., Chongqing, China
| | - Ping Zhao
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
| | - Qing-You Xia
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
| |
Collapse
|
41
|
Dai H, Wu B, Ge Y, Hao Y, Zhou L, Hong R, Zhang J, Jiang W, Zhang Y, Li H, Zhang L. Deubiquitylase OTUD3 regulates integrated stress response to suppress progression and sorafenib resistance of liver cancer. Cell Rep 2024; 43:114487. [PMID: 38996071 DOI: 10.1016/j.celrep.2024.114487] [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: 01/29/2024] [Revised: 05/13/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
The integrated stress response (ISR) is activated in response to intrinsic and extrinsic stimuli, playing a role in tumor progression and drug resistance. The regulatory role and mechanism of ISR in liver cancer, however, remain largely unexplored. Here, we demonstrate that OTU domain-containing protein 3 (OTUD3) is a deubiquitylase of eukaryotic initiation factor 2α (eIF2α), antagonizing ISR and suppressing liver cancer. OTUD3 decreases interactions between eIF2α and the kinase EIF2ΑK3 by removing K27-linked polyubiquitylation on eIF2α. OTUD3 deficiency in mice leads to enhanced ISR and accelerated progression of N-nitrosodiethylamine-induced hepatocellular carcinoma. Additionally, decreased OTUD3 expression associated with elevated eIF2α phosphorylation correlates with the progression of human liver cancer. Moreover, ISR activation due to decreased OTUD3 expression renders liver cancer cells resistant to sorafenib, while the combined use of the ISR inhibitor ISRIB significantly improves their sensitivity to sorafenib. Collectively, these findings illuminate the regulatory mechanism of ISR in liver cancer and provide a potential strategy to counteract sorafenib resistance.
Collapse
Affiliation(s)
- Hongmiao Dai
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China; Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Bo Wu
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China
| | - Yingwei Ge
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China
| | - Yang Hao
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China
| | - Lijie Zhou
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China; School of Medicine, Tsinghua University, Beijing 100084, China
| | - Ruolin Hong
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China; Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| | - Jinhao Zhang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China; Department of Cell Biology, School of Basic Medicine, Medical College, Qingdao University, Qingdao 266071, China
| | - Wenli Jiang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China; School of Life Sciences, Hebei University, Baoding, Hebei 071002, China
| | - Yuting Zhang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China; School of Life Sciences, Hebei University, Baoding, Hebei 071002, China
| | - Hongchang Li
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China.
| | - Lingqiang Zhang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China.
| |
Collapse
|
42
|
Matsuzaki T, Weistuch C, de Graff A, Dill KA, Balázsi G. Transcriptional drift in aging cells: A global decontroller. Proc Natl Acad Sci U S A 2024; 121:e2401830121. [PMID: 39012826 PMCID: PMC11287169 DOI: 10.1073/pnas.2401830121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/11/2024] [Indexed: 07/18/2024] Open
Abstract
As cells age, they undergo a remarkable global change: In transcriptional drift, hundreds of genes become overexpressed while hundreds of others become underexpressed. Using archetype modeling and Gene Ontology analysis on data from aging Caenorhabditis elegans worms, we find that the up-regulated genes code for sensory proteins upstream of stress responses and down-regulated genes are growth- and metabolism-related. We observe similar trends within human fibroblasts, suggesting that this process is conserved in higher organisms. We propose a simple mechanistic model for how such global coordination of multiprotein expression levels may be achieved by the binding of a single factor that concentrates with age in C. elegans. A key implication is that a cell's own responses are part of its aging process, so unlike wear-and-tear processes, intervention might be able to modulate these effects.
Collapse
Affiliation(s)
- Tyler Matsuzaki
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, New York, NY11794
| | - Corey Weistuch
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY10065
| | | | - Ken A. Dill
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, New York, NY11794
| | - Gábor Balázsi
- Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, New York, NY11794
- Department of Biomedical Engineering, Stony Brook University, New York, NY11794
- Stony Brook Cancer Center, Stony Brook University, New York, NY11794
| |
Collapse
|
43
|
Idowu M, Taiwo G, Sidney T, Adewoye A, Ogunade IM. Plasma proteomic analysis reveals key pathways associated with divergent residual body weight gain phenotype in beef steers. Front Vet Sci 2024; 11:1415594. [PMID: 39104547 PMCID: PMC11298483 DOI: 10.3389/fvets.2024.1415594] [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: 06/17/2024] [Indexed: 08/07/2024] Open
Abstract
We utilized plasma proteomics profiling to explore metabolic pathways and key proteins associated with divergent residual body weight gain (RADG) phenotype in crossbred (Angus × Hereford) beef steers. A group of 108 crossbred growing beef steers (average BW = 282.87 ± 30 kg; age = 253 ± 28 days) were fed a high-forage total mixed ration for 49 days in five dry lot pens (20-22 beef steers per pen), each equipped with two GrowSafe8000 intake nodes to determine their RADG phenotype. After RADG identification, blood samples were collected from the beef steers with the highest RADG (most efficient; n = 15; 0.76 kg/d) and lowest RADG (least efficient; n = 15; -0.65 kg/d). Plasma proteomics analysis was conducted on all plasma samples using a nano LC-MS/MS platform. Proteins with FC ≥ 1.2 and false-discovery rate-adjusted p-values (FDR) ≤ 0.05 were considered significantly differentially abundant. The analysis identified 435 proteins, with 59 differentially abundant proteins (DAPs) between positive and negative-RADG beef steers. Plasma abundance of 38 proteins, such as macrophage stimulating 1 and peptidase D was upregulated (FC ≥ 1.2, FDR ≤ 0.05) in positive-RADG beef steers, while 21 proteins, including fibronectin and ALB protein were greater (FC < 1.2, FDR ≤ 0.05) in negative-RADG beef steers. The results of the Gene Ontology (GO) analysis of all the DAPs showed enrichment of pathways such as metabolic processes, biological regulation, and catalytic activity in positive-RADG beef steers. Results of the EuKaryotic Orthologous Groups (KOG) analysis revealed increased abundance of DAPs involved in energy production and conversion, amino acid transport and metabolism, and lipid transport and metabolism in positive-RADG beef steers. The results of this study revealed key metabolic pathways and proteins associated with divergent RADG phenotype in beef cattle which give more insight into the biological basis of feed efficiency in crossbred beef cattle.
Collapse
Affiliation(s)
- Modoluwamu Idowu
- Division of Animal Science, West Virginia University, Morgantown, WV, United States
| | - Godstime Taiwo
- Division of Animal Science, West Virginia University, Morgantown, WV, United States
| | - Taylor Sidney
- Division of Animal Science, West Virginia University, Morgantown, WV, United States
| | - Anjola Adewoye
- Department of Chemistry, West Virginia University, Morgantown, WV, United States
| | - Ibukun M. Ogunade
- Division of Animal Science, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
44
|
Cai W, Rong D, Ding J, Zhang X, Wang Y, Fang Y, Xiao J, Yang S, Wang H. Activation of the PERK/eIF2α axis is a pivotal prerequisite of taxanes to cancer cell apoptosis and renders synergism to overcome paclitaxel resistance in breast cancer cells. Cancer Cell Int 2024; 24:249. [PMID: 39020371 PMCID: PMC11256575 DOI: 10.1186/s12935-024-03443-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Microtubule polymerization is usually considered as the upstream of apoptotic cell death induced by taxanes, but recently published studies provide more insights into the mechanisms responsible for the antineoplastic effect of taxanes. In this study, we figure out the role of the stress-related PERK/eIF2α axis in tumor cell death upon taxane treatment along with paclitaxel resistance. METHODS Utilizing immunoblot assay, the activation status of PERK-eIF2α signaling was detected in a panel of cancer cell lines after the treatment of taxanes. The causal role of PERK-eIF2α signaling in the cancer cell apoptosis induced by taxanes was examined via pharmacological and genetic inhibitions of PERK. The relationship between microtubule polymerization and PERK-eIF2α activation was explored by immunofluorescent and immunoblotting assays. Eventaually, the combined therapeutic effect of paclitaxel (PTX) and CCT020312, a PERK agonist, was investigated in PTX-resistant breast cancer cells in vitro and in vivo. RESULTS PERK-eIF2α axis was dramatically activated by taxanes in several cancer cell types. Pharmacological or genetic inhibition of PERK efficiently impaired taxane-induced apoptotic cell death, independent of the cellular microtubule polymerization status. Moreover, PTX was able to activate the PERK/eIF2α axis in a very low concentration without triggering microtubule polymerization. In PTX-resistant breast cancer cells, the PERK/eIF2α axis was attenuated in comparison with the PTX-sensitive counterparts. Reactivation of the PERK/eIF2α axis in the PTX-resistant breast cancer cells with PERK agonist sensitized them to PTX in vitro. Combination treatment of the xenografted PTX-resistant breast tumors with PERK agonist and PTX validated the synergic effect of PTX and PERK activation in vivo. CONCLUSION Activation of the PERK/eIF2α axis is a pivotal prerequisite of taxanes to initiate cancer cell apoptosis, which is independent of the well-known microtubule polymerization-dependent manner. Simultaneous activation of PERK-eIF2α signaling would be a promising therapeutic strategy to overcome PTX resistance in breast cancer or other cancers.
Collapse
Affiliation(s)
- Wanhua Cai
- Center for Translational Medicine, the First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Dade Rong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Jiayu Ding
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Xiaomei Zhang
- Center for Translational Medicine, the First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China
| | - Yuwei Wang
- Center for Translational Medicine, the First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China
- School of Medicine, Xizang Minzu University, No.6 Wenhui Donglu, Xianyang, 712082, China
| | - Ying Fang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Jing Xiao
- Department of Clinical Laboratory, Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, China.
| | - Shulan Yang
- Center for Translational Medicine, the First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China.
| | - Haihe Wang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China.
- School of Medicine, Xizang Minzu University, No.6 Wenhui Donglu, Xianyang, 712082, China.
- Clinical Medical Research Centre for Plateau Gastroenterological Disease of Xizang Autonomous Region, Xizang Minzu University, Xianyang 712082, China.
| |
Collapse
|
45
|
Chen Y, Jiao W, Wang Y, Liang Z, Wang L, Li D, Liang Y, Niu H. Microtubule interacting and trafficking domain containing 1 deficiency leads to poor survival via tissue factor-mediated coagulation in bladder cancer. J Thromb Haemost 2024; 22:1956-1972. [PMID: 38554936 DOI: 10.1016/j.jtha.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/08/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Patients with cancer are at an increased risk of developing a hypercoagulative phenotype and venous thromboembolism. However, no clinical trial has yet confirmed that anticoagulant therapy improves cancer prognosis, and the mechanism underlying hypercoagulation in patients with bladder cancer is not well understood. OBJECTIVES We hypothesized that the prognostic genes affect tumor progression via tumor-mediated coagulation. METHODS We detected the most significant prognostic genes of bladder cancer with The Cancer Genome Atlas dataset and validated them in 2 Gene Expression Omnibus datasets and 1 ArrayExpress dataset. Immunohistochemical tests were performed on a cohort of 80 individuals to further examine the prognostic genes. For the most reliable prognostic gene, its influence on coagulation was evaluated with gene knockdown followed by next-generation sequencing and cellular and animal experiments. RESULTS Depletion of microtubule interacting and trafficking domain containing 1 (MITD1), a major prognostic gene of bladder cancer, significantly increased the tissue factor (TF) expression. MITD1 deficiency led to cytokinesis arrest, which, in turn, promoted the TF expression via unfolded protein response and c-Jun. The knockdown of IRE1, an essential kinase of unfolded protein response or the inactivation of c-Jun using c-Jun N-terminal kinase inhibitors weakened MITD1 deficiency- or dithiothreitol-induced TF upregulation. Cells lacking MITD1 promoted coagulation and metastasis in the experimental metastasis assay. CONCLUSION Our findings suggest the novel role of tumor prognostic genes upon the development of hypercoagulative phenotype and venous thromboembolism, thereby underlining the importance of anticoagulant therapy and shedding light on the therapeutic value of targeting MITD1 in bladder cancer.
Collapse
Affiliation(s)
- Yuanbin Chen
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Jiao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yonghua Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhijuan Liang
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liping Wang
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dan Li
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ye Liang
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Haitao Niu
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China; Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
46
|
Mu W, Zhi Y, Zhou J, Wang C, Chai K, Fan Z, Lv G. Endoplasmic reticulum stress and quality control in relation to cisplatin resistance in tumor cells. Front Pharmacol 2024; 15:1419468. [PMID: 38948460 PMCID: PMC11211601 DOI: 10.3389/fphar.2024.1419468] [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: 04/18/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024] Open
Abstract
The endoplasmic reticulum (ER) is a crucial organelle that orchestrates key cellular functions like protein folding and lipid biosynthesis. However, it is highly sensitive to disturbances that lead to ER stress. In response, the unfolded protein response (UPR) activates to restore ER homeostasis, primarily through three sensors: IRE1, ATF6, and PERK. ERAD and autophagy are crucial in mitigating ER stress, yet their dysregulation can lead to the accumulation of misfolded proteins. Cisplatin, a commonly used chemotherapy drug, induces ER stress in tumor cells, activating complex signaling pathways. Resistance to cisplatin stems from reduced drug accumulation, activation of DNA repair, and anti-apoptotic mechanisms. Notably, cisplatin-induced ER stress can dualistically affect tumor cells, promoting either survival or apoptosis, depending on the context. ERAD is crucial for degrading misfolded proteins, whereas autophagy can protect cells from apoptosis or enhance ER stress-induced apoptosis. The complex interaction between ER stress, cisplatin resistance, ERAD, and autophagy opens new avenues for cancer treatment. Understanding these processes could lead to innovative strategies that overcome chemoresistance, potentially improving outcomes of cisplatin-based cancer treatments. This comprehensive review provides a multifaceted perspective on the complex mechanisms of ER stress, cisplatin resistance, and their implications in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhongqi Fan
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
47
|
Huang S, Zhang Y, Shu H, Liu W, Zhou X, Zhou X. Advances of the MAPK pathway in the treatment of spinal cord injury. CNS Neurosci Ther 2024; 30:e14807. [PMID: 38887853 PMCID: PMC11183187 DOI: 10.1111/cns.14807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/23/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
Spinal cord injury (SCI) represents a complex pathology within the central nervous system (CNS), leading to severe sensory and motor impairments. It activates various signaling pathways, notably the mitogen-activated protein kinase (MAPK) pathway. Present treatment approaches primarily focus on symptomatic relief, lacking efficacy in addressing the underlying pathophysiological mechanisms. Emerging research underscores the significance of the MAPK pathway in neuronal differentiation, growth, survival, axonal regeneration, and inflammatory responses post-SCI. Modulating this pathway post-injury has shown promise in attenuating inflammation, minimizing apoptosis, alleviating neuropathic pain, and fostering neural regeneration. Given its pivotal role, the MAPK pathway emerges as a potential therapeutic target in SCI management. This review synthesizes current knowledge on SCI pathology, delineates the MAPK pathway's characteristics, and explores its dual roles in SCI pathology and therapeutic interventions. Furthermore, it addresses the existing challenges in MAPK research in the context of SCI, proposing solutions to overcome these hurdles. Our aim is to offer a comprehensive reference for future research on the MAPK pathway and SCI, laying the groundwork for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Shixue Huang
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Yinuo Zhang
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Haoming Shu
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Wei Liu
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Xin Zhou
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
- Translational Research Centre of Orthopedics, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
48
|
Liu Q, Jiao L, Ye MS, Ma Z, Yu J, Su LY, Zou WY, Yang LX, Chen C, Yao YG. GSNOR negatively regulates the NLRP3 inflammasome via S-nitrosation of MAPK14. Cell Mol Immunol 2024; 21:561-574. [PMID: 38570588 PMCID: PMC11143353 DOI: 10.1038/s41423-024-01155-9] [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: 09/03/2023] [Accepted: 03/17/2024] [Indexed: 04/05/2024] Open
Abstract
Hyperactivation of the NLRP3 inflammasome has been implicated in the pathogenesis of numerous diseases. However, the precise molecular mechanisms that modulate the transcriptional regulation of NLRP3 remain largely unknown. In this study, we demonstrated that S-nitrosoglutathione reductase (GSNOR) deficiency in macrophages leads to significant increases in the Nlrp3 and Il-1β expression levels and interleukin-1β (IL-1β) secretion in response to NLRP3 inflammasome stimulation. Furthermore, in vivo experiments utilizing Gsnor-/- mice revealed increased disease severity in both lipopolysaccharide (LPS)-induced septic shock and dextran sodium sulfate (DSS)-induced colitis models. Additionally, we showed that both LPS-induced septic shock and DSS-induced colitis were ameliorated in Gsnor-/- Nlrp3-/- double-knockout (DKO) mice. Mechanistically, GSNOR deficiency increases the S-nitrosation of mitogen-activated protein kinase 14 (MAPK14) at the Cys211 residue and augments MAPK14 kinase activity, thereby promoting Nlrp3 and Il-1β transcription and stimulating NLRP3 inflammasome activity. Our findings suggested that GSNOR is a regulator of the NLRP3 inflammasome and that reducing the level of S-nitrosylated MAPK14 may constitute an effective strategy for alleviating diseases associated with NLRP3-mediated inflammation.
Collapse
Affiliation(s)
- Qianjin Liu
- Key Laboratory of Genetic Evolution & Animal Models, and Key Laboratory of Animal Models & Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650201, Kunming, Yunnan, China.
| | - Lijin Jiao
- Key Laboratory of Genetic Evolution & Animal Models, and Key Laboratory of Animal Models & Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650201, Kunming, Yunnan, China
| | - Mao-Sen Ye
- Key Laboratory of Genetic Evolution & Animal Models, and Key Laboratory of Animal Models & Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650201, Kunming, Yunnan, China
| | - Zhiyu Ma
- Key Laboratory of Genetic Evolution & Animal Models, and Key Laboratory of Animal Models & Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650201, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, 650204, Kunming, Yunnan, China
| | - Jinsong Yu
- Key Laboratory of Genetic Evolution & Animal Models, and Key Laboratory of Animal Models & Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650201, Kunming, Yunnan, China
| | - Ling-Yan Su
- Key Laboratory of Genetic Evolution & Animal Models, and Key Laboratory of Animal Models & Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650201, Kunming, Yunnan, China
| | - Wei-Yin Zou
- Key Laboratory of Genetic Evolution & Animal Models, and Key Laboratory of Animal Models & Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650201, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, 650204, Kunming, Yunnan, China
| | - Lu-Xiu Yang
- Key Laboratory of Genetic Evolution & Animal Models, and Key Laboratory of Animal Models & Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650201, Kunming, Yunnan, China
| | - Chang Chen
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yong-Gang Yao
- Key Laboratory of Genetic Evolution & Animal Models, and Key Laboratory of Animal Models & Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650201, Kunming, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, 650204, Kunming, Yunnan, China.
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, 650201, Kunming, Yunnan, China.
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, 650201, Kunming, China.
| |
Collapse
|
49
|
Alasmari A. Achillea fragrantissima (Forssk.) Sch.Bip instigates the ROS/FADD/c-PARP expression that triggers apoptosis in breast cancer cell (MCF-7). PLoS One 2024; 19:e0304072. [PMID: 38820323 PMCID: PMC11142488 DOI: 10.1371/journal.pone.0304072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/03/2024] [Indexed: 06/02/2024] Open
Abstract
Achillea fragrantissima is a shrub plant that belongs to the Asteraceae family in Arabia and Egypt. It is used as folk medicine and is a good source of phenolic acids, flavonoids, and some active compounds. To investigate the anti-cancer effect of A.fragrantissima on breast cancer MCF-7 cells and find the critical mechanism involved in apoptosis. The toxicity and pharmacokinetic studies of ethanolic extract of A.fragrantissima was examined for anti-breast cancer properties. In turn, cytotoxicity and cell viability were achieved by the MTT method. Furthermore, the trypan blue exclusion and microscopy examination proved the presence of apoptotic cells. Again, fluorescent staining such as AO/EtBr, DCFH-DA, Rho-123, and Hoechst-33342 reveals the cellular cytoplasmic disciplines upon A. fragrantissima effect. Moreover, cellular functioning tests like wound healing, colony formation, and Transwell invasion assay were demonstrated. In addition, the qRT-PCR technique authenticates the A. fragrantissima -induced apoptotic network genes (Caspase-3, Caspase-8, Caspase-9, Cytochrome c, BCL-2, BID, BAX, PARP, PTEN, PI3K, and Akt) expression were evaluated. Mainly, the Immunoblot technique proved the expressed level of apoptotic proteins such as cleaved PARP, CYCS, and FADD. This study confirmed that the A. fragrantissima exerts cytotoxicity at 20 μg/mL for 24 hrs in MCF-7 cells. Also, decreases cellular viability, producing apoptotic cells and damaged cellular surfaces with dead matter. Consequently, it creates ROS species accumulation, loss of mitochondrial membrane potential, and fragmentation of DNA in MCF-7 cells. Furthermore, it arrests cell migration, induces colony-forming ability loss, and suppresses cell invasion. In addition, A. fragrantissima significantly upregulates genes such as caspase-3, 9, cytochrome c, BID, BAX, and PTEN while downregulating the Pi3K/ Akt signaling. Nonetheless, A.fragrantissima induced cleaved PARP, CYCS, and FADD proteins in MCF-7 cells to avail apoptosis.
Collapse
Affiliation(s)
- Abdulrahman Alasmari
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
- Biodiversity Genomics Unit, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
50
|
Cao J, Yang Y, Duan B, Zhang H, Xu Q, Han J, Lu B. LncRNA PCED1B-AS1 mediates miR-3681-3p/MAP2K7 axis to promote metastasis, invasion and EMT in gastric cancer. Biol Direct 2024; 19:34. [PMID: 38698487 PMCID: PMC11064384 DOI: 10.1186/s13062-024-00468-z] [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: 11/28/2023] [Accepted: 03/19/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND LncRNA PCED1B-AS1 is abnormally expressed in multiple cancers and has been confirmed as an oncogene. Our study aimed to investigate the regulatory mechanism of lncRNA PCED1B-AS1 in gastric cancer. METHODS TCGA database was used to analyze the abnormal expression of lncRNA PCED1B-AS1 in gastric cancer. By database prediction and mass spectrometric analysis, miR-3681-3p and MAP2K7 are potential downstream target molecules of lncRNA PCED1B-AS1 and verified by dual-luciferase report assay. RT-qPCR analysis and western blot were performed to detect the expressions of PCED1B-AS1 and MAP2K7 in gastric cancer cell lines and tissues. CCK-8 kit was applied to measure the cell viability. Wound healing and Transwell experiment were used to detect the migration and invasion. Western blot and immunohistochemical staining were performed to detect the expressions of EMT-related proteins in tissues. The changes of tumor proliferation were detected by xenograft experiment in nude mice. RESULTS PCED1B-AS1 expression was higher but miR-3681-3 expression was lower in gastric cancer cell lines or tissues, compared to normal group. Function analysis verified PCED1B-AS1 promoted cell proliferation and inhibited cell apoptosis in gastric cancer cells in vitro and in vivo. LncRNA PCED1B-AS1 could bind directly to miR-3681-3p, and MAP2K7 was found to be a downstream target of miR-3681-3p. MiR-3681-3p mimics or si-MAP2K7 could partly reverse the effect of PCED1B-AS1 on gastric cancer cells. CONCLUSION PCED1B-AS1 accelerated cell proliferation and inhibited cell apoptosis through sponging miR-3681-3p to upregulate MAP2K7 expression in gastric cancer, which indicated PCED1B-AS1/miR-3681-3p/MAP2K7 axis may serve as a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Jia Cao
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yicheng Yang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bensong Duan
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haibin Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qinwei Xu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junyi Han
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Bing Lu
- Department of General Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|