1
|
Stein M, Böttcher N, Tohidnezhad M, Fragoulis A, Bayer A, Klump H, Baron JM, Pufe T. Platelet-Released Growth Factors (PRGFs) Activate NRF2-ARE and Modulate Inflammatory Response in an NRF2-Dependent Manner in Primary Human Keratinocytes. J Cosmet Dermatol 2025; 24:e70228. [PMID: 40353553 PMCID: PMC12067853 DOI: 10.1111/jocd.70228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/29/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Platelet-released-growth factors (PRGF) and platelet-rich plasma (PRP) are blood-derived products used in regenerative treatments and in overall aesthetic rejuvenation. Keratinocytes possess distinctive characteristics responsible for protection against environmental stressors and oxidant clearance. One such mechanism is the transcription factor NRF2, which plays a critical role in regulating cytoprotective genes, inflammation, and oxidative stress response. Data on the activation of the NRF2-ARE and NF-κB axes by PRGF are very limited. AIM This study aims to investigate whether PRGF activates NRF2 and, if so, is responsible for the described anti-inflammatory effect of PRGF/PRP in an in vitro primary human keratinocyte model. METHODS NRF2 activation is analyzed by NQO1 and HO-1 western blotting, gene expression analysis, and by an ARE-promoter study using luciferase-based reporter gene assays in patient-derived keratinocytes. Besides direct determination of the PRGF-NRF2 interaction, we investigated the NF-κB response by treating cells with PRGF and the inflammatory stimuli TNF-α. Inflammatory parameters were analyzed using ELISAs for IL-1β, IL-4, Il-10, TNF-α and IL-6 in the supernatant, NF-κB luciferase reporter gene assays as well a-NF-κB western blotting. NRF2 involvement was tested by treating the cell-culture model with the NRF2-inhibitor ML-385. RESULTS We were able to show that ARE activity was significantly upregulated in PRGF-treated keratinocytes, leading subsequently to increased NQO1 and HO-1 protein expression. Inflammatory IL-secretion showed an association with NRF2 availability. CONCLUSIONS In summary, PRGFs activate NRF2 target proteins and downregulate NF-κB-associated inflammation in an NRF2-dependent manner. Therefore, we further suggest PRGF as an anti-inflammatory treatment after medical aesthetic procedures.
Collapse
Affiliation(s)
- Matthias Stein
- Department of Anatomy and Cell BiologyUniklinik RWTH Aachen UniversityAachenGermany
| | - Nicole Böttcher
- Department of Anatomy and Cell BiologyUniklinik RWTH Aachen UniversityAachenGermany
| | - Mersedeh Tohidnezhad
- Department of Anatomy and Cell BiologyUniklinik RWTH Aachen UniversityAachenGermany
| | | | - Andreas Bayer
- Center for Clinical AnatomyInstitute of Anatomy, Kiel UniversityKielGermany
| | - Hannes Klump
- Institute for Transfusion Medicine and Cell Therapeutics, Uniklinik RWTH Aachen UniversityAachenGermany
| | - Jens M. Baron
- Department of Dermatology and AllergologyUniklinik RWTH Aachen UniversityAachenGermany
| | - Thomas Pufe
- Department of Anatomy and Cell BiologyUniklinik RWTH Aachen UniversityAachenGermany
| |
Collapse
|
2
|
Li J, Luo J, Wang T, Tian X, Xu C, Wang W, Zhang D. DNA methylation associated with the serum alanine aminotransferase concentration: evidence from Chinese monozygotic twins. Clin Epigenetics 2025; 17:65. [PMID: 40296130 PMCID: PMC12039056 DOI: 10.1186/s13148-025-01869-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/30/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND To identify nongenetic factors influences on DNA methylation (DNAm) variations associated with blood Alanine Aminotransferase (ALT) concentration, this study conducted an epigenome-wide association study (EWAS) on Chinese monozygotic twins. METHODS A total of 61 pairs of Chinese monozygotic twins involved in this study. Whole blood samples were analyzed for DNAm profiling using the Reduced Representation Bisulfite Sequencing (RRBS) technique. We examined the relationship between DNAm levels at each CpG site and serum ALT using a linear mixed-effects model. Enrichment analysis and causal inference analysis was conducted, and differentially methylated regions (DMRs) were further identified. Candidate CpGs were validated in a community sample. Genome-wide significance were calculated by Bonferroni correction (p < 2.14 × 10-7). RESULTS We identified 85 CpGs reaching genome-wide significance (p < 2.14 × 10-7), located in 16 genes including FLT4, ADARB2, MRPS31P2, and RELB. Causal inference suggested that DNAm at 61 out of 85 significant CpGs within 14 genes influenced ALT level. 52 DMRs and 1765 pathways such as low voltage-gated calcium channel activity and focal adhesion were identified having influences on ALT levels. Further validation using community population found four CpGs mapped to FLT4 and three to RELB showing hypomethylation and hypermethylation in cases with abnormal ALT (ALT > 40 U/L), respectively. CONCLUSION This study identified several differentially methylated CpG sites associated with serum ALT in the Chinese population, particularly within FLT4 and RELB. These findings provide new insights into the epigenetic modifications underlying liver function.
Collapse
Affiliation(s)
- Jingxian Li
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao, 266071, Shandong Province, People's Republic of China
| | - Jia Luo
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao, 266071, Shandong Province, People's Republic of China
| | - Tong Wang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao, 266071, Shandong Province, People's Republic of China
| | - Xiaocao Tian
- Qingdao Municipal Centre for Disease Control and Prevention, No.175 Shandong Road, Qingdao, 266033, Shandong Province, People's Republic of China
| | - Chunsheng Xu
- Qingdao Municipal Centre for Disease Control and Prevention, No.175 Shandong Road, Qingdao, 266033, Shandong Province, People's Republic of China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao, 266071, Shandong Province, People's Republic of China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao, 266071, Shandong Province, People's Republic of China.
| |
Collapse
|
3
|
Špaková I, Smolko L, Sabolová G, Badovská Z, Kalinová K, Madreiter-Sokolowski C, Graier WF, Mareková M, Vašková J, Rabajdová M. Selective targeting of genes regulated by zinc finger proteins in endometriosis and endometrioid adenocarcinoma by zinc niflumato complex with neocuproine. Sci Rep 2025; 15:10126. [PMID: 40128272 PMCID: PMC11933352 DOI: 10.1038/s41598-025-94249-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 03/12/2025] [Indexed: 03/26/2025] Open
Abstract
Inadequate angiogenesis of endometriotic implants stimulated by the inflammatory microenvironment in the uterine region leads to the development of gynecological diseases, which significantly reduce the fertility and vitality of young women. Angiogenic processes are controlled by factors whose activities are regulated at the gene level by reactive oxygen species (ROS), hypoxia-induced factors (HIFs), and zinc-finger proteins (ZnFs) or posttranscriptionally via non-coding RNAs. The cooperation of these factors is responsible for the manifestation of pathological stimuli in the form of endometriosis of the body of the uterus, ovaries, or peritoneum, from which endometrioid carcinoma can develop. Molecules that can control gene expression by their intercalation to target DNA sequence, such as [Zn(neo)(nif)2], could prevent the hyperactivation of pro-angiogenic pathways (decrease HIF-1α, VEGF-A, TGF-β1, COX2, and ANG2/ANG1), reduce the formation of ROS, and reduce the risk of uterine neoplasticity. The NSAID-metal complex [Zn(neo)(nif)2] shows an ability to intercalate into ZNF3-7 target DNA sequence at a higher rate, which could explain its effect on genes regulated by this transcription factor. In addition, [Zn(neo)(nif)2] affects ROS production and Ca2+ level, possibly pointing to mitochondrial dysfunction as a potential cause for the described apoptosis.
Collapse
Affiliation(s)
- Ivana Špaková
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Lukáš Smolko
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Gabriela Sabolová
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Zuzana Badovská
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Katarína Kalinová
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biohemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, T8010, Graz, Austria
| | - Corina Madreiter-Sokolowski
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biohemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, T8010, Graz, Austria
| | - Wolfgang F Graier
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biohemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, T8010, Graz, Austria
| | - Mária Mareková
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Janka Vašková
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Miroslava Rabajdová
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia.
| |
Collapse
|
4
|
Bukowska P, Bralewska M, Pietrucha T, Sakowicz A. Nutraceuticals as Modulators of Molecular Placental Pathways: Their Potential to Prevent and Support the Treatment of Preeclampsia. Int J Mol Sci 2024; 25:12167. [PMID: 39596234 PMCID: PMC11594370 DOI: 10.3390/ijms252212167] [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: 10/31/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Preeclampsia (PE) is a serious condition characterized by new-onset hypertension and proteinuria or organ dysfunction after the 20th week of gestation, making it a leading cause of maternal and fetal mortality worldwide. Despite extensive research, significant gaps remain in understanding the mechanisms underlying PE, contributing to the ineffectiveness of current prevention and treatment strategies. Consequently, premature cesarean sections often become the primary intervention to safeguard maternal and fetal health. Emerging evidence indicates that placental insufficiency, driven by molecular disturbances, plays a central role in the development of PE. Additionally, the maternal microbiome may be implicated in the pathomechanism of preeclampsia by secreting metabolites that influence maternal inflammation and oxidative stress, thereby affecting placental health. Given the limitations of pharmaceuticals during pregnancy due to potential risks to fetal development and concerns about teratogenic effects, nutraceuticals may provide safer alternatives. Nutraceuticals are food products or dietary supplements that offer health benefits beyond basic nutrition, including plant extracts or probiotics. Their historical use in traditional medicine has provided valuable insights into their safety and efficacy, including for pregnant women. This review will examine how the adoption of nutraceuticals can enhance dysregulated placental pathways, potentially offering benefits in the prevention and treatment of preeclampsia.
Collapse
Affiliation(s)
| | | | | | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| |
Collapse
|
5
|
Li D, Lu Y, Xiao F, Cheng X, Hu C, Zhu X, Wang X, Duan H, Du L, Zhang Q. A recombinant plasmid encoding human hepatocyte growth factor promotes healing of combined radiation-trauma skin injury involved in regulating Nrf2 pathway in mice. JOURNAL OF RADIATION RESEARCH 2024; 65:279-290. [PMID: 38682896 PMCID: PMC11115442 DOI: 10.1093/jrr/rrae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 01/01/2024] [Indexed: 05/01/2024]
Abstract
Combined radiation-trauma skin injury represents a severe and intractable condition that urgently requires effective therapeutic interventions. In this context, hepatocyte growth factor (HGF), a multifunctional growth factor with regulating cell survival, angiogenesis, anti-inflammation and antioxidation, may be valuable for the treatment of combined radiation-trauma injury. This study investigated the protective effects of a recombinant plasmid encoding human HGF (pHGF) on irradiated human immortalized keratinocytes (HaCaT) cells in vitro, and its capability to promote the healing of combined radiation-trauma injuries in mice. The pHGF radioprotection on irradiated HaCaT cells in vitro was assessed by cell viability, the expression of Nrf2, Bcl-2 and Bax, as well as the secretion of inflammatory cytokines. In vivo therapeutic treatment, the irradiated mice with full-thickness skin wounds received pHGF local injection. The injuries were appraised based on relative wound area, pathology, immunohistochemical detection, terminal deoxynucleotidyl transferase dUTP nick end labelling assay and cytokine content. The transfection of pHGF increased the cell viability and Nrf2 expression in irradiated HaCaT cells. pHGF also significantly upregulated Bcl-2 expression, decreased the Bax/Bcl-2 ratio and inhibited the expression of interleukin-1β and tumor necrosis factor-α in irradiated cells. Local pHGF injection in vivo caused high HGF protein expression and noticeable accelerated healing of combined radiation-trauma injury. Moreover, pHGF administration upregulated Nrf2, vascular endothelial growth factor, Bcl-2 expression, downregulated Bax expression and mitigated inflammatory response. In conclusion, the protective effect of pHGF may be related to inhibiting apoptosis and inflammation involving by upregulating Nrf2. Local pHGF injection distinctly promoted the healing of combined radiation-trauma injury and demonstrates potential as a gene therapy intervention for combined radiation-trauma injury in clinic.
Collapse
Affiliation(s)
- Dujuan Li
- Department of Pharmacy & Pharmacology, University of South China, 28 Changsheng West Road, Zhengxiang District, Hengyang, Hunan 421001, China
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Yuxin Lu
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Fengjun Xiao
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Xiaochen Cheng
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Chunsheng Hu
- Department of Pharmacology, College of Pharmacy & International Academy of Targeted Therapeutics and Innovation, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing University of Arts and Sciences, 319 Honghe avenue, Yongchuan District, Chongqing 402160, China
| | - Xuefeng Zhu
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Xiaoying Wang
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Haiying Duan
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Li Du
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Qinglin Zhang
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing 100850, China
| |
Collapse
|
6
|
Benchaprathanphorn K, Muangman P, Chinaroonchai K, Namviriyachote N, Ampawong S, Angkhasirisap W, Kengkoom K, Viravaidya-Pasuwat K. Translational application of human keratinocyte-fibroblast cell sheets for accelerated wound healing in a clinically relevant type 2 diabetic rat model. Cytotherapy 2024; 26:360-371. [PMID: 38363247 DOI: 10.1016/j.jcyt.2024.01.003] [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: 08/09/2023] [Revised: 12/27/2023] [Accepted: 01/20/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND AIMS Despite advancements in wound care, wound healing remains a challenge, especially in individuals with type 2 diabetes. Cell sheet technology has emerged as an efficient and promising therapy for tissue regeneration and wound repair. Among these, bilayered human keratinocyte-fibroblast cell sheets constructed using temperature-responsive culture surfaces have been shown to mimic a normal tissue-like structure and secrete essential cytokines and growth factors that regulate the wound healing process. METHODS This study aimed to evaluate the safety and therapeutic potential of human skin cell sheets to treat full-thickness skin defects in a rat model of type 2 diabetes. RESULTS Our findings demonstrate that diabetic wounds transplanted with bilayered cell sheets resulted in accelerated re-epithelialization, increased angiogenesis, enhanced macrophage polarization and regeneration of tissue that closely resembled healthy skin. In contrast, the control group that did not receive cell sheet transplantation presented characteristic symptoms of impaired and delayed wound healing associated with type 2 diabetes. CONCLUSIONS The secretory cytokines and the upregulation of Nrf2 expression in response to cell sheet transplantation are believed to have played a key role in the improved wound healing observed in diabetic rats. Our study suggests that human keratinocyte-fibroblast cell sheets hold great potential as a therapeutic alternative for diabetic ulcers.
Collapse
Affiliation(s)
- Kanokaon Benchaprathanphorn
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Pornprom Muangman
- Trauma Surgery Division, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kusuma Chinaroonchai
- Trauma Surgery Division, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nantaporn Namviriyachote
- Trauma Surgery Division, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wannee Angkhasirisap
- Research and Academic Support Office, National Laboratory Animal Center, Mahidol University, Nakorn Pathom, Thailand
| | - Kanchana Kengkoom
- Research and Academic Support Office, National Laboratory Animal Center, Mahidol University, Nakorn Pathom, Thailand
| | - Kwanchanok Viravaidya-Pasuwat
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand; Chemical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand.
| |
Collapse
|
7
|
Song N, Fan X, Guo X, Tang J, Li H, Tao R, Li F, Li J, Yang D, Yao C, Liu P. A DNA/Upconversion Nanoparticle Complex Enables Controlled Co-Delivery of CRISPR-Cas9 and Photodynamic Agents for Synergistic Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309534. [PMID: 38199243 DOI: 10.1002/adma.202309534] [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: 09/14/2023] [Revised: 11/27/2023] [Indexed: 01/12/2024]
Abstract
Photodynamic therapy (PDT) depends on the light-irradiated exciting of photosensitizer (PS) to generate reactive oxygen species (ROS), which faces challenges and limitations in hypoxia and antioxidant response of cancer cells, and limited tissue-penetration of light. Herein, a multifunctional DNA/upconversion nanoparticles (UCNPs) complex is developed which enables controlled co-delivery of CRISPR-Cas9, hemin, and protoporphyrin (PP) for synergistic PDT. An ultralong single-stranded DNA (ssDNA) is prepared via rolling circle amplification (RCA), which contains recognition sequences of single guide RNA (sgRNA) for loading Cas9 ribonucleoprotein (RNP), G-quadruplex sequences for loading hemin and PP, and linker sequences for combining UCNP. Cas9 RNP cleaves the antioxidant regulator nuclear factor E2-related factor 2 (Nrf2), improving the sensitivity of cancer cells to ROS, and enhancing the synergistic PDT effect. The G-quadruplex/hemin DNAzyme mimicks horseradish peroxidase (HRP) to catalyze the endogenous H2O2 to O2, overcoming hypoxia condition in tumors. The introduced UCNP converts NIR irradiation with deep tissue penetration to light with shorter wavelength, exciting PP to transform the abundant O2 to 1O2. The integration of gene editing and PDT allows substantial accumulation of 1O2 in cancer cells for enhanced cell apoptosis, and this synergistic PDT has shown remarkable therapeutic efficacy in a breast cancer mouse model.
Collapse
Affiliation(s)
- Nachuan Song
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai, 200438, P. R. China
| | - Xiaoting Fan
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Xiaocui Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Jianpu Tang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai, 200438, P. R. China
| | - Hongjin Li
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Ruoyu Tao
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Fengqin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Junru Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| | - Dayong Yang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai, 200438, P. R. China
| | - Chi Yao
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Peifeng Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, P. R. China
| |
Collapse
|
8
|
Keshavarzi F, Salehi MS, Pandamooz S, Zare R, Zamani M, Mostafavi-Pour Z, Pooneh Mokarram P. Valproic acid and/or rapamycin preconditioning protects hair follicle stem cells from oxygen glucose serum deprivation-induced oxidative injury via activating Nrf2 pathway. MOLECULAR BIOLOGY RESEARCH COMMUNICATIONS 2024; 13:103-116. [PMID: 38915453 PMCID: PMC11194030 DOI: 10.22099/mbrc.2024.49302.1922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Among leading causes of the ischemic stroke pathogenesis, oxidative stress strongly declines rate of stem cell engraftment at the injury site, and disables stem cell-based therapy as a key treatment for ischemia stroke. To overcome this therapeutic limitation, preconditioning has been represented a possible approach to augment the adaptation and viability of stem cells to oxidative stress. Here, we illustrated protective impacts of valproic acid (VPA) and/or rapamycin (RAPA) preconditioning unto oxygen glucose and serum deprivation (OGSD)-stimulated cell damage in hair follicle-derived stem cells (HFSCs) and surveyed the plausible inducement mechanisms. OGSD, as an in vitro cell injury model, was established and HFSCs viability was observed using MTT assay after VPA, RAPA, and VPA-RAPA preconditioning under OGSD. ROS and MDA production was assessed to reflect oxidative stress. Real-time PCR and western blotting were employed to investigate Nrf2 expression. The activity of Nrf2-related antioxidant enzymes including NQO1, GPx and GSH level were examined. VEGF and BDNF mRNA expression levels were analyzed. Our results showed that VPA and/or RAPA preconditioning ameliorated OGSD-induced decline in HFSCs viability. In addition, they considerably prohibited ROS and MDA generation in the OGSD-treated HFSCs. Furthermore, VPA and/or RAPA preconditioning stimulated Nrf2 nuclear repositioning and NQO1 and GPx activity and GSH amount, as well as expression of paracrine factors VEGF and BDNF in OGSD-treated HFSCs. Thus, the protective effects afforded by VPA and/or RAPA preconditioning, which involved Nrf2-modulated oxidant stress and regulation of VEGF and BDNF expression, display a simple strategy to augment cell-transplantation efficiency for ischemic stroke.
Collapse
Affiliation(s)
- Fatemeh Keshavarzi
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Zare
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhdeh Zamani
- Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zohreh Mostafavi-Pour
- Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooneh Pooneh Mokarram
- Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Che J, Yang X, Jin Z, Xu C. Nrf2: A promising therapeutic target in bone-related diseases. Biomed Pharmacother 2023; 168:115748. [PMID: 37865995 DOI: 10.1016/j.biopha.2023.115748] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023] Open
Abstract
Nuclear factor erythroid-2-related factor 2 (Nrf2) plays an important role in maintaining cellular homeostasis, as it suppresses cell damage caused by external stimuli by regulating the transcription of intracellular defense-related genes. Accumulating evidence has highlighted the crucial role of reduction-oxidation (REDOX) imbalance in the development of bone-related diseases. Nrf2, a transcription factor linked to nuclear factor-erythrocyte 2, plays a pivotal role in the regulation of oxidative stress and induction of antioxidant defenses. Therefore, further investigation of the mechanism and function of Nrf2 in bone-related diseases is essential. Considerable evidence suggests that increased nuclear transcription of Nrf2 in response to external stimuli promotes the expression of intracellular antioxidant-related genes, which in turn leads to the inhibition of bone remodeling imbalance, improved fracture recovery, reduced occurrence of osteoarthritis, and greater tumor resistance. Certain natural extracts can selectively target Nrf2, potentially offering therapeutic benefits for osteogenic arthropathy. In this article, the biological characteristics of Nrf2 are reviewed, the intricate interplay between Nrf2-regulated REDOX imbalance and bone-related diseases is explored, and the potential preventive and protective effects of natural products targeting Nrf2 in these diseases are elucidated. A comprehensive understanding of the role of Nrf2 in the development of bone-related diseases provides valuable insights into clinical interventions and can facilitate the discovery of novel Nrf2-targeting drugs.
Collapse
Affiliation(s)
- Jingmin Che
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China.
| | - Xiaoli Yang
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Zhankui Jin
- Department of Orthopedics, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China.
| | - Cuixiang Xu
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
10
|
Segherlou ZH, Siyanaki MRH, Lucke-Wold B. Potential Effects of Adropin in Subarachnoid Hemorrhage. NEUROSCIENCE INTERNATIONAL 2023; 13:12-19. [PMID: 37654780 PMCID: PMC10469087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Subarachnoid Hemorrhage (SAH) typically, occurs in patients over 55 years of age and can cause a significant loss of productivity. SAH also has a high mortality rate and those who survive often suffer from early and secondary brain injuries that can result from the condition. By gaining a better understanding of the pathophysiology of SAH, it may be possible to identify therapeutic agents to improve outcomes. Adropin is a novel peptide that is primarily secreted in the liver and brain. Research has shown that adropin can activate endothelial NO synthase through post-transcriptional mechanisms. Studies in animal models have demonstrated that therapies using synthetic adropin peptide or adropin overexpression can have positive effects on reducing infarct dimensions and enhancing neurological functioning. In this review, we aim to discuss the potential effect of Adropin on SAH and its potential as a therapeutic agent.
Collapse
|
11
|
Rahman SO, Khan T, Iqubal A, Agarwal S, Akhtar M, Parvez S, Shah ZA, Najmi AK. Association between insulin and Nrf2 signalling pathway in Alzheimer's disease: A molecular landscape. Life Sci 2023:121899. [PMID: 37394097 DOI: 10.1016/j.lfs.2023.121899] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/17/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023]
Abstract
Insulin, a well-known hormone, has been implicated as a regulator of blood glucose levels for almost a century now. Over the past few decades, the non-glycemic actions of insulin i.e. neuronal growth and proliferation have been extensively studied. In 2005, Dr. Suzanne de La Monte and her team reported that insulin might be involved in the pathogenesis of Alzheimer's Disease (AD) and thus coined a term "Type-3 diabetes" This hypothesis was supported by several subsequent studies. The nuclear factor erythroid 2- related factor 2 (Nrf2) triggers a cascade of events under the regulation of distinct mechanisms including protein stability, phosphorylation and nuclear cytoplasmic shuttling, finally leading to the protection against oxidative damage. The Nrf2 pathway has been investigated extensively in relevance to neurodegenerative disorders, particularly AD. Many studies have indicated a strong correlation between insulin and Nrf2 signalling pathways both in the periphery and the brainbut merely few of them have focused on elucidating their inter-connective role in AD. The present review emphasizes key molecular pathways that correlate the role of insulin with Nrf2 during AD. The review has also identified key unexplored areas that could be investigated in future to further establish the insulin and Nrf2 influence in AD.
Collapse
Affiliation(s)
- Syed Obaidur Rahman
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Tahira Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Shivani Agarwal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Suhel Parvez
- Neurobehavioral Pharmacology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Zahoor Ahmad Shah
- Department of Medicinal and Biological Chemistry, University of Toledo, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
12
|
Tossetta G, Fantone S, Piani F, Crescimanno C, Ciavattini A, Giannubilo SR, Marzioni D. Modulation of NRF2/KEAP1 Signaling in Preeclampsia. Cells 2023; 12:1545. [PMID: 37296665 PMCID: PMC10252212 DOI: 10.3390/cells12111545] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/24/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Placentation is a key and tightly regulated process that ensures the normal development of the placenta and fetal growth. Preeclampsia (PE) is a hypertensive pregnancy-related disorder involving about 5-8% of all pregnancies and clinically characterized by de novo maternal hypertension and proteinuria. In addition, PE pregnancies are also characterized by increased oxidative stress and inflammation. The NRF2/KEAP1 signaling pathway plays an important role in protecting cells against oxidative damage due to increased reactive oxygen species (ROS) levels. ROS activate NRF2, allowing its binding to the antioxidant response element (ARE) region present in the promoter of several antioxidant genes such as heme oxygenase, catalase, glutathione peroxidase and superoxide dismutase that neutralize ROS, protecting cells against oxidative stress damages. In this review, we analyze the current literature regarding the role of the NRF2/KEAP1 pathway in preeclamptic pregnancies, discussing the main cellular modulators of this pathway. Moreover, we also discuss the main natural and synthetic compounds that can regulate this pathway in in vivo and in vitro models.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.F.); (D.M.)
| | - Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.F.); (D.M.)
| | - Federica Piani
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40128 Bologna, Italy;
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Caterina Crescimanno
- School of Human and Social Science, University “Kore” of Enna, 94100 Enna, Italy;
| | - Andrea Ciavattini
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, 60123 Ancona, Italy; (A.C.); (S.R.G.)
| | - Stefano Raffaele Giannubilo
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, 60123 Ancona, Italy; (A.C.); (S.R.G.)
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.F.); (D.M.)
| |
Collapse
|
13
|
Fragoulis A, Tohidnezhad M, Kubo Y, Wruck CJ, Craveiro RB, Bock A, Wolf M, Pufe T, Jahr H, Suhr F. The Contribution of the Nrf2/ARE System to Mechanotransduction in Musculoskeletal and Periodontal Tissues. Int J Mol Sci 2023; 24:ijms24097722. [PMID: 37175428 PMCID: PMC10177782 DOI: 10.3390/ijms24097722] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Mechanosensing plays an essential role in maintaining tissue functions. Across the human body, several tissues (i.e., striated muscles, bones, tendons, ligaments, as well as cartilage) require mechanical loading to exert their physiological functions. Contrary, mechanical unloading triggers pathological remodeling of these tissues and, consequently, human body dysfunctions. At the cellular level, both mechanical loading and unloading regulate a wide spectrum of cellular pathways. Among those, pathways regulated by oxidants such as reactive oxygen species (ROS) represent an essential node critically controlling tissue organization and function. Hence, a sensitive balance between the generation and elimination of oxidants keeps them within a physiological range. Here, the Nuclear Factor-E2-related factor 2/Antioxidant response element (Nrf2/ARE) system plays an essential role as it constitutes the major cellular regulation against exogenous and endogenous oxidative stresses. Dysregulations of this system advance, i.a., liver, neurodegenerative, and cancer diseases. Herein, we extend our comprehension of the Nrf2 system to the aforementioned mechanically sensitive tissues to explore its role in their physiology and pathology. We demonstrate the relevance of it for the tissues' functionality and highlight the imperative to further explore the Nrf2 system to understand the physiology and pathology of mechanically sensitive tissues in the context of redox biology.
Collapse
Affiliation(s)
- Athanassios Fragoulis
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Mersedeh Tohidnezhad
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Yusuke Kubo
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Christoph Jan Wruck
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Rogerio Bastos Craveiro
- Department of Orthodontics, Dental Clinic, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Anna Bock
- Department of Oral and Maxillofacial Surgery, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Michael Wolf
- Department of Orthodontics, Dental Clinic, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Holger Jahr
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
- Institute of Structural Mechanics and Lightweight Design, RWTH Aachen University, 52062 Aachen, Germany
| | - Frank Suhr
- Division of Molecular Exercise Physiology, Faculty of Life Sciences: Food, Nutrition and Health, University of Bayreuth, 95326 Kulmbach, Germany
| |
Collapse
|
14
|
Novel Anti-Cancer Products Targeting AMPK: Natural Herbal Medicine against Breast Cancer. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020740. [PMID: 36677797 PMCID: PMC9863744 DOI: 10.3390/molecules28020740] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 01/15/2023]
Abstract
Breast cancer is a common cancer in women worldwide. The existing clinical treatment strategies have been able to limit the progression of breast cancer and cancer metastasis, but abnormal metabolism, immunosuppression, and multidrug resistance involving multiple regulators remain the major challenges for the treatment of breast cancer. Adenosine 5'-monophosphate (AMP)-Activated Protein Kinase (AMPK) can regulate metabolic reprogramming and reverse the "Warburg effect" via multiple metabolic signaling pathways in breast cancer. Previous studies suggest that the activation of AMPK suppresses the growth and metastasis of breast cancer cells, as well as stimulating the responses of immune cells. However, some other reports claim that the development and poor prognosis of breast cancer are related to the overexpression and aberrant activation of AMPK. Thus, the role of AMPK in the progression of breast cancer is still controversial. In this review, we summarize the current understanding of AMPK, particularly the comprehensive bidirectional functions of AMPK in cancer progression; discuss the pharmacological activators of AMPK and some specific molecules, including the natural products (including berberine, curcumin, (-)-epigallocatechin-3-gallate, ginsenosides, and paclitaxel) that influence the efficacy of these activators in cancer therapy; and elaborate the role of AMPK as a potential therapeutic target for the treatment of breast cancer.
Collapse
|
15
|
Hermilasari RD, Rizal DM, Wirohadidjojo YW. Potential Mechanism of Platelet-rich Plasma Treatment on Testicular Problems Related to Diabetes Mellitus. Prague Med Rep 2023; 124:344-358. [PMID: 38069642 DOI: 10.14712/23362936.2023.27] [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: 12/18/2023] Open
Abstract
Diabetes mellitus is a condition of continuously increased blood glucose levels that causes hyperglycemia. This condition can result in disorders of various organs including testicular problems. The use of platelet-rich plasma (PRP) which is contained in several growth factors shows its potential in overcoming testicular problems. This literature review study was conducted to identify the potential of PRP in overcoming various testicular problems due to diabetic conditions.
Collapse
Affiliation(s)
- Rista Dwi Hermilasari
- Department of Public Health, Faculty of Public Health, University of Jember, Jember, Indonesia
| | - Dicky Moch Rizal
- Department of Physiology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Yohanes Widodo Wirohadidjojo
- Department of Dermatology and Venereology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
16
|
Awuah WA, Toufik AR, Yarlagadda R, Mikhailova T, Mehta A, Huang H, Kundu M, Lopes L, Benson S, Mykola L, Vladyslav S, Alexiou A, Alghamdi BS, Hashem AM, Md Ashraf G. Exploring the role of Nrf2 signaling in glioblastoma multiforme. Discov Oncol 2022; 13:94. [PMID: 36169772 PMCID: PMC9519816 DOI: 10.1007/s12672-022-00556-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/01/2022] [Indexed: 11/05/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive glial cell tumors in adults. Although current treatment options for GBM offer some therapeutic benefit, median survival remains poor and does not generally exceed 14 months. Several genes, such as isocitrate dehydrogenase (IDH) enzyme and O6-methylguanine-DNA methyltransferase (MGMT), have been implicated in pathogenesis of the disease. Treatment is often adapted based on the presence of IDH mutations and MGMT promoter methylation status. Recent GBM cell line studies have associated Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) expression with high-grade tumors. Increased Nrf2 expression is often found in tumors with IDH-1 mutations. Nrf2 is an important transcription factor with anti-apoptotic, antioxidative, anti-inflammatory, and proliferative properties due to its complex interactions with multiple regulatory pathways. In addition, evidence suggests that Nrf2 promotes GBM cell survival in hypoxic environment,by up-regulating hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF). Downregulation of Nrf2 has been shown to improve GBM sensitivity to chemotherapy drugs such as Temozolomide. Thus, Nrf2 could be a key regulator of GBM pathways and potential therapeutic target. Further research efforts exploring an interplay between Nrf2 and major molecular signaling mechanisms could offer novel GBM drug candidates with a potential to significantly improve patients prognosis.
Collapse
Affiliation(s)
| | | | - Rohan Yarlagadda
- Rowan University School of Osteopathic Medicine, Stratford, NJ USA
| | | | - Aashna Mehta
- University of Debrecen-Faculty of Medicine, Debrecen, 4032 Hungary
| | - Helen Huang
- Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Mrinmoy Kundu
- Institute of Medical Sciences and SUM Hospital, Bhubaneswar, India
| | - Leilani Lopes
- College of Osteopathic Medicine of the Pacific-Northwest, Western University of Health Sciences, Lebanon, OR USA
| | | | | | | | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770 Australia
- AFNP Med, 1030 Vienna, Austria
| | - Badrah S. Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anwar M. Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
| |
Collapse
|
17
|
Fragoulis A, Schenkel J, Schröder N, Brandt EF, Weiand M, Neu T, Ramadori P, Caspers T, Kant S, Pufe T, Mohs A, Trautwein C, Longerich T, Streetz KL, Wruck CJ. Nrf2 induces malignant transformation of hepatic progenitor cells by inducing β-catenin expression. Redox Biol 2022; 57:102453. [PMID: 36209041 PMCID: PMC9618468 DOI: 10.1016/j.redox.2022.102453] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 07/22/2022] [Accepted: 08/19/2022] [Indexed: 11/28/2022] Open
Abstract
The Nrf2 signaling pathway prevents cancer initiation, but genetic mutations that activate this pathway are found in various types of cancer. The molecular mechanisms underlying this Janus-headed character are still not understood. Here, we show that sustained Nrf2 activation induces proliferation and dedifferentiation of a Wnt-responsive perivenular hepatic progenitor cell population, transforming them into metastatic cancer cells. The neoplastic lesions display many histological features known from human hepatoblastoma. We describe an Nrf2-induced upregulation of β-catenin expression and its activation as the underlying mechanism for the observed malignant transformation. Thus, we have identified the Nrf2–β-catenin axis promoting proliferation of hepatic stem cells and triggering tumorigenesis. These findings support the concept that different functional levels of Nrf2 control both the protection against various toxins as well as liver regeneration by activating hepatic stem cells. Activation of the hepatic stem cell compartment confers the observation that unbridled Nrf2 activation may trigger tumorigenesis.
Collapse
Affiliation(s)
| | - Julia Schenkel
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Germany
| | - Nicole Schröder
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Germany
| | | | - Mathias Weiand
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Germany
| | - Tabita Neu
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Germany
| | - Pierluigi Ramadori
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Caspers
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Germany
| | - Sebastian Kant
- Institute of Molecular and Cellular Anatomy, Uniklinik RWTH Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Germany
| | - Antje Mohs
- Department of Medicine III, Uniklinik RWTH Aachen, Germany
| | | | | | | | | |
Collapse
|
18
|
Kumar H, Kumar RM, Bhattacharjee D, Somanna P, Jain V. Role of Nrf2 Signaling Cascade in Breast Cancer: Strategies and Treatment. Front Pharmacol 2022; 13:720076. [PMID: 35571115 PMCID: PMC9098811 DOI: 10.3389/fphar.2022.720076] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 03/31/2022] [Indexed: 12/30/2022] Open
Abstract
Breast cancer is the second leading cancer among all types of cancers. It accounts for 12% of the total cases of cancers. The complex and heterogeneous nature of breast cancer makes it difficult to treat in advanced stages. The expression of various enzymes and proteins is regulated by several molecular pathways. Oxidative stress plays a vital role in cellular events that are generally regulated by nuclear factor erythroid 2-related factor 2 (Nrf2). The exact mechanism of Nrf2 behind cytoprotective and antioxidative properties is still under investigation. In healthy cells, Nrf2 expression is lower, which maintains antioxidative stress; however, cancerous cells overexpress Nrf2, which is associated with various phenomena, such as the development of drug resistance, angiogenesis, development of cancer stem cells, and metastasis. Aberrant Nrf2 expression diminishes the toxicity and potency of therapeutic anticancer drugs and provides cytoprotection to cancerous cells. In this article, we have discussed the attributes associated with Nrf2 in the development of drug resistance, angiogenesis, cancer stem cell generation, and metastasis in the specific context of breast cancer. We also discussed the therapeutic strategies employed against breast cancer exploiting Nrf2 signaling cascades.
Collapse
Affiliation(s)
| | | | | | | | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, India
| |
Collapse
|
19
|
Pérez-Alfayate R, Grasso G. State of the Art and Future Direction in Diagnosis, Molecular Biology, Genetics, and Treatment of Brain Arteriovenous Malformations. World Neurosurg 2022; 159:362-372. [PMID: 35255635 DOI: 10.1016/j.wneu.2021.08.111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022]
Abstract
Brain arteriovenous malformations (bAVMs) are uncommon and represent a heterogeneous group of lesions. Although these 2 facts have delayed research on this topic, knowledge about the pathophysiology, diagnosis, and treatment of bAVMs has evolved in recent years. We conducted a review of the literature to update the knowledge about diagnosis, molecular biology, genetic, pathology, and treatment by searching for the following terms: "Epidemiology AND Natural History," "risk of hemorrhage," "intracranial hemorrhage," "diagnosis," "angiogenesis," "molecular genetics," "VEGF," "KRAS," "radiosurgery," "endovascular," "microsurgery," or "surgical resection." Our understanding of bAVMs has significantly evolved in recent years. The latest investigations have helped in defining some molecular pathways involved in the pathology of bAVM. Although there is still more to learn and discover, describing these pathways will allow the creation of targeted treatments that could improve the prognosis of patients with bAVMs.
Collapse
Affiliation(s)
- Rebeca Pérez-Alfayate
- Department of Neurosurgery, Neuroscience Institute, Hospital Clínico San Carlos, Madrid, Spain.
| | - Giovanni Grasso
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| |
Collapse
|
20
|
Sheppard AJ, Barfield AM, Barton S, Dong Y. Understanding Reactive Oxygen Species in Bone Regeneration: A Glance at Potential Therapeutics and Bioengineering Applications. Front Bioeng Biotechnol 2022; 10:836764. [PMID: 35198545 PMCID: PMC8859442 DOI: 10.3389/fbioe.2022.836764] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/19/2022] [Indexed: 01/24/2023] Open
Abstract
Although the complex mechanism by which skeletal tissue heals has been well described, the role of reactive oxygen species (ROS) in skeletal tissue regeneration is less understood. It has been widely recognized that a high level of ROS is cytotoxic and inhibits normal cellular processes. However, with more recent discoveries, it is evident that ROS also play an important, positive role in skeletal tissue repair, specifically fracture healing. Thus, dampening ROS levels can potentially inhibit normal healing. On the same note, pathologically high levels of ROS cause a sharp decline in osteogenesis and promote nonunion in fracture repair. This delicate balance complicates the efforts of therapeutic and engineering approaches that aim to modulate ROS for improved tissue healing. The physiologic role of ROS is dependent on a multitude of factors, and it is important for future efforts to consider these complexities. This review first discusses how ROS influences vital signaling pathways involved in the fracture healing response, including how they affect angiogenesis and osteogenic differentiation. The latter half glances at the current approaches to control ROS for improved skeletal tissue healing, including medicinal approaches, cellular engineering, and enhanced tissue scaffolds. This review aims to provide a nuanced view of the effects of ROS on bone fracture healing which will inspire novel techniques to optimize the redox environment for skeletal tissue regeneration.
Collapse
Affiliation(s)
- Aaron J. Sheppard
- Department of Orthopaedic Surgery, Louisiana State University Health Shreveport, Shreveport, LA, United States
- School of Medicine, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Ann Marie Barfield
- Department of Orthopaedic Surgery, Louisiana State University Health Shreveport, Shreveport, LA, United States
- School of Medicine, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Shane Barton
- Department of Orthopaedic Surgery, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Yufeng Dong
- Department of Orthopaedic Surgery, Louisiana State University Health Shreveport, Shreveport, LA, United States
| |
Collapse
|
21
|
Tantengco OAG, de Castro Silva M, Shahin H, Bento GFC, Cursino GC, Cayenne S, da Silva MG, Menon R. The role of nuclear factor erythroid 2-related factor 2 (NRF2) in normal and pathological pregnancy: A systematic review. Am J Reprod Immunol 2021; 86:e13496. [PMID: 34467607 DOI: 10.1111/aji.13496] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE A homeostatic balance between reactive oxygen species production and the antioxidant redox system is an important component of normal pregnancy. Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) preserves cellular homeostasis by enhancing the cell's innate antioxidant status to reduce oxidative stress and inflammatory damage to the cell during pregnancy. Active Nrf2, in the nucleus of the cell, transactivates various antioxidant genes. The objective of this systematic review was to synthesize evidence on the role of Nrf2 in various adverse pregnancy outcomes (APOs). METHODS We conducted a systematic review of the role of Nrf2 in pregnancy. Articles written in English, Portuguese, and Spanish were obtained from three different databases from inception until January 2021. The titles, abstracts and full text were reviewed independently by six reviewers. The quality of the included studies was assessed using a quality assessment tool developed to assess basic science and clinical studies. Nrf2 expression (gene and protein), functional contributions, and association with APOs were assessed. RESULTS A total of 747 citations were identified; 80 were retained for full review. Most studies on Nrf2 have been carried out using placental tissues and placenta-derived cells. Limited studies have been conducted using fetal membranes, uterus, and cervix. Nuclear translocation of Nrf2 results in transactivation of antioxidant enzymes, including glutathione peroxidase, hemeoxygenase-1, and superoxide dismutase in gestational cells during pregnancy. This antioxidant response maintains cellular homeostasis during pregnancy. This promotes trophoblast cell survival and prevents cell death and abnormal angiogenesis in the placenta. Excessive and insufficient Nrf2 response may promote oxidative and reductive stress, respectively. This Nrf2 dysregulation has been associated with APOs including gestational diabetes mellitus, intrauterine growth restriction, reproductive toxicity, preeclampsia, and preterm birth. CONCLUSION Several studies have localized and reported an association between Nrf2's differential expression in reproductive tissues and the pathogenesis of APOs. However, a comprehensive functional understanding of Nrf2 in reproductive tissues is still lacking. Nrf2's activation and functions are complex, and therefore, current in vitro and in vivo studies are limited in their experimental approaches. We have identified key areas for future Nrf2 research that is needed to fill knowledge gaps.
Collapse
Affiliation(s)
- Ourlad Alzeus G Tantengco
- Division of Basic & Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Mariana de Castro Silva
- Department of Pathology, Botucatu Medical School, Universidade Estadual Paulista, UNESP, Botucatu, São Paulo, Brazil
| | - Hend Shahin
- Division of Basic & Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Giovana Fernanda Cosi Bento
- Department of Pathology, Botucatu Medical School, Universidade Estadual Paulista, UNESP, Botucatu, São Paulo, Brazil
| | - Geovanna Cristofani Cursino
- Department of Pathology, Botucatu Medical School, Universidade Estadual Paulista, UNESP, Botucatu, São Paulo, Brazil
| | - Samir Cayenne
- The University of Texas at Austin, Austin, Texas, USA
| | - Marcia Guimarães da Silva
- Department of Pathology, Botucatu Medical School, Universidade Estadual Paulista, UNESP, Botucatu, São Paulo, Brazil
| | - Ramkumar Menon
- Division of Basic & Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| |
Collapse
|
22
|
Lee BR, Paing MH, Sharma-Walia N. Cyclopentenone Prostaglandins: Biologically Active Lipid Mediators Targeting Inflammation. Front Physiol 2021; 12:640374. [PMID: 34335286 PMCID: PMC8320392 DOI: 10.3389/fphys.2021.640374] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
Cyclopentenone prostaglandins (cyPGs) are biologically active lipid mediators, including PGA2, PGA1, PGJ2, and its metabolites. cyPGs are essential regulators of inflammation, cell proliferation, apoptosis, angiogenesis, cell migration, and stem cell activity. cyPGs biologically act on multiple cellular targets, including transcription factors and signal transduction pathways. cyPGs regulate the inflammatory response by interfering with NF-κB, AP-1, MAPK, and JAK/STAT signaling pathways via both a group of nuclear receptor peroxisome proliferator-activated receptor-gamma (PPAR-γ) dependent and PPAR-γ independent mechanisms. cyPGs promote the resolution of chronic inflammation associated with cancers and pathogen (bacterial, viral, and parasitic) infection. cyPGs exhibit potent effects on viral infections by repressing viral protein synthesis, altering viral protein glycosylation, inhibiting virus transmission, and reducing virus-induced inflammation. We summarize their anti-proliferative, pro-apoptotic, cytoprotective, antioxidant, anti-angiogenic, anti-inflammatory, pro-resolution, and anti-metastatic potential. These properties render them unique therapeutic value, especially in resolving inflammation and could be used in adjunct with other existing therapies. We also discuss other α, β -unsaturated carbonyl lipids and cyPGs like isoprostanes (IsoPs) compounds.
Collapse
|
23
|
Mende W, Götzl R, Kubo Y, Pufe T, Ruhl T, Beier JP. The Role of Adipose Stem Cells in Bone Regeneration and Bone Tissue Engineering. Cells 2021; 10:cells10050975. [PMID: 33919377 PMCID: PMC8143357 DOI: 10.3390/cells10050975] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Bone regeneration is a complex process that is influenced by tissue interactions, inflammatory responses, and progenitor cells. Diseases, lifestyle, or multiple trauma can disturb fracture healing, which might result in prolonged healing duration or even failure. The current gold standard therapy in these cases are bone grafts. However, they are associated with several disadvantages, e.g., donor site morbidity and availability of appropriate material. Bone tissue engineering has been proposed as a promising alternative. The success of bone-tissue engineering depends on the administered cells, osteogenic differentiation, and secretome. Different stem cell types offer advantages and drawbacks in this field, while adipose-derived stem or stromal cells (ASCs) are in particular promising. They show high osteogenic potential, osteoinductive ability, and immunomodulation properties. Furthermore, they can be harvested through a noninvasive process in high numbers. ASCs can be induced into osteogenic lineage through bioactive molecules, i.e., growth factors and cytokines. Moreover, their secretome, in particular extracellular vesicles, has been linked to fracture healing. The aim of this review is a comprehensive overview of ASCs for bone regeneration and bone tissue engineering.
Collapse
Affiliation(s)
- Wolfgang Mende
- Hand Surgery-Burn Center, Department of Plastic Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Rebekka Götzl
- Hand Surgery-Burn Center, Department of Plastic Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Yusuke Kubo
- Department of Anatomy and Cell Biology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Tim Ruhl
- Hand Surgery-Burn Center, Department of Plastic Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Justus P Beier
- Hand Surgery-Burn Center, Department of Plastic Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany
| |
Collapse
|
24
|
Süntar I, Çetinkaya S, Panieri E, Saha S, Buttari B, Profumo E, Saso L. Regulatory Role of Nrf2 Signaling Pathway in Wound Healing Process. Molecules 2021; 26:molecules26092424. [PMID: 33919399 PMCID: PMC8122529 DOI: 10.3390/molecules26092424] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 12/30/2022] Open
Abstract
Wound healing involves a series of cellular events in damaged cells and tissues initiated with hemostasis and finally culminating with the formation of a fibrin clot. However, delay in the normal wound healing process during pathological conditions due to reactive oxygen species, inflammation and immune suppression at the wound site represents a medical challenge. So far, many therapeutic strategies have been developed to improve cellular homeostasis and chronic wounds in order to accelerate wound repair. In this context, the role of Nuclear factor erythroid 2-related factor 2 (Nrf2) during the wound healing process has been a stimulating research topic for therapeutic perspectives. Nrf2 is the main regulator of intracellular redox homeostasis. It increases cytoprotective gene expression and the antioxidant capacity of mammalian cells. It has been reported that some bioactive compounds attenuate cellular stress and thus accelerate cell proliferation, neovascularization and repair of damaged tissues by promoting Nrf2 activation. This review highlights the importance of the Nrf2 signaling pathway in wound healing strategies and the role of bioactive compounds that support wound repair through the modulation of this crucial transcription factor.
Collapse
Affiliation(s)
- Ipek Süntar
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Etiler, Ankara 06330, Turkey
- Correspondence: ; Tel.: +90-31-2202-3176
| | - Sümeyra Çetinkaya
- Biotechnology Research Center of Ministry of Agriculture and Forestry, Yenimahalle, Ankara 06330, Turkey;
| | - Emiliano Panieri
- Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University, 00185 Rome, Italy; (E.P.); (L.S.)
| | - Sarmistha Saha
- Department of Cardiovascular and Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (S.S.); (B.B.); (E.P.)
| | - Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (S.S.); (B.B.); (E.P.)
| | - Elisabetta Profumo
- Department of Cardiovascular and Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (S.S.); (B.B.); (E.P.)
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University, 00185 Rome, Italy; (E.P.); (L.S.)
| |
Collapse
|
25
|
Almeida MPO, Mota CM, Mineo TWP, Ferro EAV, Barbosa BF, Silva NM. Heme Oxygenase-1 Induction in Human BeWo Trophoblast Cells Decreases Toxoplasma gondii Proliferation in Association With the Upregulation of p38 MAPK Phosphorylation and IL-6 Production. Front Microbiol 2021; 12:659028. [PMID: 33912151 PMCID: PMC8071940 DOI: 10.3389/fmicb.2021.659028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/09/2021] [Indexed: 11/13/2022] Open
Abstract
Heme oxygenase-1 (HO-1) enzyme exerts beneficial effects at the maternal-fetal interface, especially in trophoblasts, being involved in survival and maturation of these cell phenotypes. Trophoblast cells play essential roles throughout pregnancy, being the gateway for pathogens vertically transmitted, such as Toxoplasma gondii. It was previously shown that HO-1 activity was involved in the control of T. gondii infection in vivo; however, its contribution in trophoblast cells during T. gondii infection, remain undefined. Thus, this study aimed to investigate the influence of HO-1 in T. gondii-infected BeWo and HTR-8/SVneo human trophoblast cells. For this purpose, trophoblast cells were infected and the HO-1 expression was evaluated. T. gondii-infected BeWo cells were treated with hemin or CoPPIX, as inducers of HO-1, or with bilirubin, an end-product of HO-1, and the parasitism was quantified. The involvement of p38 MAPK, a regulator of HO-1, and the cytokine production, were also evaluated. It was found that T. gondii decreased the HO-1 expression in BeWo but not in HTR-8/SVneo cells. When treated with the HO-1 inducers or bilirubin, BeWo cells reduced the parasite proliferation. T. gondii also decreased the p38 MAPK phosphorylation in BeWo cells; on the other hand, HO-1 induction sustained its activation. Finally, the IL-6 production was upregulated by HO-1 induction in T. gondii-infected cells, which was associated with the control of infection.
Collapse
Affiliation(s)
- Marcos Paulo Oliveira Almeida
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Caroline Martins Mota
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo," Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Tiago Wilson Patriarca Mineo
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo," Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Neide Maria Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
26
|
Pacheco-Romero J, Acosta O, Huerta D, Cabrera S, Vargas M, Mascaro P, Huamán M, Sandoval J, López R, Mateus J, Gil E, Guevara E, Butrica N, Catari D, Bellido D, Custodio G, Naranjo A. Genetic markers for preeclampsia in Peruvian women. Colomb Med (Cali) 2021; 52:e2014437. [PMID: 33911318 PMCID: PMC8054708 DOI: 10.25100/cm.v52i1.4437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/13/2020] [Accepted: 01/28/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Preeclampsia is a multiorgan disorder associated with maternal and perinatal morbi-mortality. In Peru, incidence is 10% and accounts for 22% of maternal deaths. Genome and genetic epidemiological studies have found an association between preeclampsia and genetic polymorphisms. OBJECTIVE To determine the association of the vascular endothelial growth factor (VEGF) +936 C/T and +405 G/C, interleukine-6 (IL-6) -174 G/C, IL-1β-511 C/T, Apo A-1-75 G/A, Apo B-100 2488 C/T (Xbal) polymorphisms with preeclampsia in pregnant Peruvian women. METHODS Were included preeclamptic and healthy (control) pregnant women. Maternal blood samples were subjected to DNA extraction, and molecular genetic analysis was conducted using the PCR-RFLP technique and following a specific protocol for each gene. Allele and genotypic frequencies in the cases and controls were compared. RESULTS No association was found between the VEGF+936C/T and VEGF+405 polymorphisms and preeclampsia. The frequencies of the GG genotypes and the G allele of the -174 G/C polymorphism in the IL6 gene in preeclamptic and controls showed significant differences, with higher frequencies in cases. For the -511 C/T polymorphism of the IL-1β gene, no significant differences were found in the frequencies of TT genotypes compared with CT+CC. The genotypes and alleles of the Apo-A1-75 G/A and Apo-B100 Xbal variants showed no significant differences between cases and controls. CONCLUSION No association was found between the studied genetic markers and preeclampsia. However, in the -174G/C polymorphism of the IL-6 gene, significant differences were found mainly in the GG genotype and G allele.
Collapse
Affiliation(s)
- José Pacheco-Romero
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Oscar Acosta
- Universidad Nacional Mayor de San Marcos, Faculty of Pharmacy and Biochemistry, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of Pharmacy and BiochemistryLimaPeru
| | - Doris Huerta
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Santiago Cabrera
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Marlene Vargas
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Pedro Mascaro
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Moisés Huamán
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - José Sandoval
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Rudy López
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Julio Mateus
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
- Atrium Health, Charlotte, North Carolina, USA.Atrium HealthCharlotteNorth CarolinaUSA
| | - Enrique Gil
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Enrique Guevara
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Nitza Butrica
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Diana Catari
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - David Bellido
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Gina Custodio
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| | - Andrea Naranjo
- Universidad Nacional Mayor de San Marcos, Faculty of Medicine. Medicina y Genética Molecular Materno Perinatal-MEGEMAPE Research Group, Lima, Peru.Universidad Nacional Mayor de San MarcosUniversidad Nacional Mayor de San MarcosFaculty of MedicineMedicina y Genética Molecular Materno Perinatal-MEGEMAPE Research GroupLimaPeru
| |
Collapse
|
27
|
Wu Y, Mi Y, Zhang F, Cheng Y, Wu X. Suppression of bromodomain-containing protein 4 protects trophoblast cells from oxidative stress injury by enhancing Nrf2 activation. Hum Exp Toxicol 2020; 40:742-753. [PMID: 33094643 DOI: 10.1177/0960327120968857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Oxidative stress is considered a key hallmark of preeclampsia, which causes the dysregulation of trophoblast cells, and it contributes to the pathogenesis of preeclampsia. Emerging evidence has suggested bromodomain-containing protein 4 (BRD4) as a key regulator of oxidative stress in multiple cell types. However, whether BRD4 participates in regulating oxidative stress in trophoblast cells remains undetermined. The current study was designed to explore the potential function of BRD4 in the regulation of oxidative stress in trophoblast cells. Our data revealed that BRD4 expression was elevated in trophoblast cells stimulated with hydrogen peroxide. Exposure to hydrogen peroxide caused marked decreases in the levels of proliferation and invasion but promoted apoptosis and the production of ROS in trophoblast cells. Knockdown of BRD4, or treatment with a BRD4 inhibitor, markedly increased the levels of cell proliferation and invasion and decreased apoptosis and ROS production following the hydrogen peroxide challenge. Further data indicated that suppression of BRD4 markedly decreased the expression levels of Keap1, but increased the nuclear expression of Nrf2 and enhanced Nrf2-mediated transcriptional activity. BRD4 inhibition-mediated protective effects were markedly reversed by Keap1 overexpression or Nrf2 inhibition. Overall, these results demonstrated that BRD4 inhibition attenuated hydrogen peroxide-induced oxidative stress injury in trophoblast cells by enhancing Nrf2 activation via the downregulation of Keap1. Our study highlights the potential importance of the BRD4/Keap1/Nrf2 axis in the modulation of the oxidative stress response in trophoblast cells. Targeted inhibition of BRD4 may offer new opportunities for the development of innovative therapeutic approaches to treat preeclampsia.
Collapse
Affiliation(s)
- Yiqing Wu
- Department of Obstetrics, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Yang Mi
- Department of Obstetrics, Northwest Women's and Children's Hospital, Xi'an, Shaanxi, China
| | - Fan Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of 117799Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yimin Cheng
- The Hospital of Xi'an Shiyou University, Xi'an, Shaanxi, China
| | - Xiaoling Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of 117799Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
28
|
Rossino MG, Lulli M, Amato R, Cammalleri M, Dal Monte M, Casini G. Oxidative Stress Induces a VEGF Autocrine Loop in the Retina: Relevance for Diabetic Retinopathy. Cells 2020; 9:E1452. [PMID: 32545222 PMCID: PMC7349409 DOI: 10.3390/cells9061452] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/01/2020] [Accepted: 06/10/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Oxidative stress (OS) plays a central role in diabetic retinopathy (DR), triggering expression and release of vascular endothelial growth factor (VEGF), the increase of which leads to deleterious vascular changes. We tested the hypothesis that OS-stimulated VEGF induces its own expression with an autocrine mechanism. METHODS MIO-M1 cells and ex vivo mouse retinal explants were treated with OS, with exogenous VEGF or with conditioned media (CM) from OS-stressed cultures. RESULTS Both in MIO-M1 cells and in retinal explants, OS or exogenous VEGF induced a significant increase of VEGF mRNA, which was abolished by VEGF receptor 2 (VEGFR-2) inhibition. OS also caused VEGF release. In MIO-M1 cells, CM induced VEGF expression, which was abolished by a VEGFR-2 inhibitor. Moreover, the OS-induced increase of VEGF mRNA was abolished by a nuclear factor erythroid 2-related factor 2 (Nrf2) blocker, while the effect of exo-VEGF resulted Nrf2-independent. Finally, both the exo-VEGF- and the OS-induced increase of VEGF expression were blocked by a hypoxia-inducible factor-1 inhibitor. CONCLUSIONS These results are consistent with the existence of a retinal VEGF autocrine loop triggered by OS. This mechanism may significantly contribute to the maintenance of elevated VEGF levels and therefore it may be of central importance for the onset and development of DR.
Collapse
Affiliation(s)
- Maria Grazia Rossino
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (M.G.R.); (R.A.); (M.C.)
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Rosario Amato
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (M.G.R.); (R.A.); (M.C.)
| | - Maurizio Cammalleri
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (M.G.R.); (R.A.); (M.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| | - Massimo Dal Monte
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (M.G.R.); (R.A.); (M.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| | - Giovanni Casini
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (M.G.R.); (R.A.); (M.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| |
Collapse
|
29
|
Guo Z, Mo Z. Keap1‐Nrf2 signaling pathway in angiogenesis and vascular diseases. J Tissue Eng Regen Med 2020; 14:869-883. [PMID: 32336035 DOI: 10.1002/term.3053] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 04/14/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Zi Guo
- Department of EndocrinologyThe Third Xiangya Hospital, Central South University Changsha China
| | - Zhaohui Mo
- Department of EndocrinologyThe Third Xiangya Hospital, Central South University Changsha China
| |
Collapse
|
30
|
Li X, Zhou H, Guo D, Hu Y, Fang X, Chen Y, Zhang F. Oxidative stress and inflammation: Early predictive indicators of multiple recurrent coronary in‐stent chronic total occlusions in elderly patients after coronary stenting. IUBMB Life 2020; 72:1023-1033. [PMID: 32022379 DOI: 10.1002/iub.2239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 01/23/2020] [Indexed: 11/07/2022]
Affiliation(s)
- Xia Li
- Department of GeriatricsThe Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Hualan Zhou
- Department of GeriatricsThe Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Dianxuan Guo
- Department of GeriatricsThe Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Youdong Hu
- Department of GeriatricsThe Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Xiang Fang
- Department of GeriatricsThe Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Ying Chen
- Department of GeriatricsThe Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Fenglin Zhang
- Department of GeriatricsThe Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| |
Collapse
|
31
|
A Review of Adropin as the Medium of Dialogue between Energy Regulation and Immune Regulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3947806. [PMID: 32190172 PMCID: PMC7073478 DOI: 10.1155/2020/3947806] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/26/2020] [Accepted: 02/10/2020] [Indexed: 11/18/2022]
Abstract
Adropin is a secretory protein encoded by the energy balance gene and is closely associated with regulation of energy metabolism and insulin resistance. The clinical findings demonstrated its decreased expression in various inflammatory diseases, its negative correlation with the expression levels of inflammatory cytokines, and its potential anti-inflammatory effects. We speculate that adropin plays a pivotal regulatory role in immune cells and inflammatory factors. In this study, we reviewed the advances in researches concentrated on immunological effects of adropin.
Collapse
|
32
|
Silvani Y, Lovita AND, Maharani A, Wiyasa IWA, Sujuti H, Ratnawati R, Raras TYM. Extra Virgin Olive Oil Modulates Vasodilator Enzyme Level by Repairing Angiogenesis Function in Rat Model of Preeclampsia. J Family Reprod Health 2020; 14:38-44. [PMID: 32863837 PMCID: PMC7428417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Objective: This study aimed to determine the effect of Extra Virgin Olive Oil (EVOO) on vasodilator enzyme by repairing angiogenic function in rat model of preeclampsia. Materials and methods: This research consisted of five groups; negative control (normal pregnant rats) group, positive control (preeclampsia rat model) group, preeclampsia rat model groups given EVOO in 3 different doses (0.5 ml/day, 1 ml/day, and 2 ml/day, respectively). Blood pressure measurements were carried out on day 12, 15, and 19 of pregnancy. After the rats were sacrificed, the placentas were collected to determine endothelial Nitric Oxide Synthase (eNOS) level of maternal plasma to determine soluble Fms-like Tyrosine Kinase 1 (sFlt-1) and Vascular Endothelial Growth Factor (VEGF) level. Results: There were significant higher sFlt-1 level (p < 0.001), lower VEGF level (p = 0.009), and lower eNOS level (p = 0.034) between negative and positive control groups. After EVOO administration, sFlt-1 level was lower in dose 1 and 2 groups but higher in dose 3 group in accordance with VEGF and eNOS levels that were increasing both in dose 1 and dose 2 groups but decreasing in dose 3. There were significant differences between positive control and dose 1 (p = 0.015) and dose 2 (p = 0.001) in sFlt-1 level. None of all dose groups were statistically different with positive control group in VEGF level (dose 1 p = 0.601; dose 2 p = 0.297; dose 3 p = 0.805). eNOS levels of all dose groups were statistically different from that of the positive control group (dose 1 p = 0.014; dose 2 p = 0.001; dose 3 p = 0.024). Conclusion: Administration of EVOO modulates eNOS as vasodilator enzyme by repairing the angiogenic function indicated by decreased sFlt-1 level and increased VEGF in rat model of preeclampsia.
Collapse
Affiliation(s)
- Yulia Silvani
- Midwifery Department, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | | | - Afniari Maharani
- Midwifery Department, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - I Wayan Arsana Wiyasa
- Department of Obstetrics and Gynecology, Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Hidayat Sujuti
- Department of Biochemistry-Molecular Biology, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Retty Ratnawati
- Department of Physiology, Faculty of Medicine University of Brawijaya, Malang, Indonesia
| | - Tri Yudani Mardining Raras
- Department of Biochemistry-Molecular Biology, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| |
Collapse
|
33
|
Li H, Min J, Chen Y, Li H, Zhang Y. Polydatin attenuates orbital oxidative stress in Graves’ orbitopathy through the NRF2 pathway. Chem Biol Interact 2020; 315:108894. [DOI: 10.1016/j.cbi.2019.108894] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/23/2019] [Accepted: 11/05/2019] [Indexed: 12/30/2022]
|
34
|
Wu X, Li R, Song Q, Zhang C, Jia R, Han Z, Zhou L, Sui H, Liu X, Zhu H, Yang L, Wang Y, Ji Q, Li Q. JMJD2C promotes colorectal cancer metastasis via regulating histone methylation of MALAT1 promoter and enhancing β-catenin signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:435. [PMID: 31665047 PMCID: PMC6819649 DOI: 10.1186/s13046-019-1439-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/06/2019] [Indexed: 12/26/2022]
Abstract
Background Our previous work demonstrated that lncRNA-MALAT1 was overexpressed in recurrent colorectal cancer (CRC) and metastatic sites in post-surgical patients. However, the upstream regulatory mechanism of MALAT1 is not well-defined. Histone demethylase JMJD2C holds great potential of epigenetic regulating mechanism in tumor diseases, especially the moderating effect on the promoter activity of targeted genes associated closely with tumor development. Therefore, we herein investigated whether JMJD2C could epigeneticly regulate the promoter activity of MALAT1 and the downstream β-catenin signaling pathway, thereby affecting the metastatic abilities of CRC cells. Methods JMJD2C expressions in human CRC samples were detected by real-time PCR and immunohistochemistry staining. Gene silencing and overexpressing efficiencies of JMJD2C were confirmed by real-time PCR and western blot. The migration of CRC cells in vitro were tested by transwell and wound healing assays. The protein expression and cellular localization of JMJD2C and β-catenin were characterized by immunofluorescence staining and western blot. The histone methylation level of MALAT1 promoter region (H3K9me3 and H3K36me3) was tested by ChIP-PCR assays. The promoter activity of MALAT1 was detected by luciferase reporter assay. The expressions of MALAT1 and the downstream β-catenin signaling pathway related genes in CRC cells were detected by real-time PCR and western blot, respectively. The nude mice tail vein metastasis model was established to observe the effect of JMJD2C on the lung metastasis of CRC cells in vivo. Results Our present results indicated that histone demethylase JMJD2C was overexpressed in matched CRC tumor tissues of primary and metastatic foci, and CRC patients with lower JMJD2C expression in primary tumors had better prognosis with longer OS (Overall Survival). The following biological function observation suggested that JMJD2C promoted CRC metastasis in vitro and in vivo. Further molecular mechanism investigation demonstrated that JMJD2C protein translocated into the nuclear, lowered the histone methylation level of MALAT1 promoter in the sites of H3K9me3 and H3K36me3, up-regulated the expression of MALAT1, and enhanced the β-catenin signaling pathway in CRC cells. Conclusion Our data demonstrated that JMJD2C could enhance the metastatic abilities of CRC cells in vitro and in vivo by regulating the histone methylation level of MALAT1 promoter, thereby up-regulating the expression of MALAT1 and enhancing the activity of β-catenin signaling pathway, providing that JMJD2C might be a novel therapeutic target for CRC metastasis.
Collapse
Affiliation(s)
- Xinnan Wu
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ruixiao Li
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing Song
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Medical Oncology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Chengcheng Zhang
- Department of Medical Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ru Jia
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhifen Han
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lihong Zhou
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Sui
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuan Liu
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huirong Zhu
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu Yang
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Wang
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing Ji
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Qi Li
- Department of Medical Oncology and Cancer Institute of Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China. .,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
35
|
Kubo Y, Wruck CJ, Fragoulis A, Drescher W, Pape HC, Lichte P, Fischer H, Tohidnezhad M, Hildebrand F, Pufe T, Jahr H. Role of Nrf2 in Fracture Healing: Clinical Aspects of Oxidative Stress. Calcif Tissue Int 2019; 105:341-352. [PMID: 31236620 DOI: 10.1007/s00223-019-00576-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022]
Abstract
Fracture healing is a natural process that recapitulates embryonic skeletal development. In the early phase after fracture, reactive oxygen species (ROS) are produced under inflammatory and ischemic conditions due to vessel injury and soft tissue damage, leading to cell death. Usually, such damage during the course of fracture healing can be largely prevented by protective mechanisms and functions of antioxidant enzymes. However, intrinsic oxidative stress can cause excessive toxic radicals, resulting in irreversible damage to cells associated with bone repair during the fracture healing process. Clinically, patients with type-2 diabetes mellitus, osteoporosis, habitual drinkers, or heavy smokers are at risk of impaired fracture healing due to elevated oxidative stress. Although increased levels of oxidative stress markers upon fracture and effects of antioxidants on fracture healing have been reported, a detailed understanding of what causes impaired fracture healing under intrinsic conditions of oxidative stress is lacking. Nuclear factor erythroid 2-related factor 2 (Nrf2) has been identified as a key transcriptional regulator of the expression of antioxidants and detoxifying enzymes. It further not only plays a crucial role in preventing degenerative diseases in multiple organs, but also during fracture healing. This narrative review evaluates the influence of intrinsic oxidative stress on fracture healing and sheds new light on the intriguing role of Nrf2 during bone regeneration in pathological fractures.
Collapse
Affiliation(s)
- Yusuke Kubo
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| | - Christoph Jan Wruck
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Athanassios Fragoulis
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Wolf Drescher
- Department of Orthopaedics, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany
- Department of Orthopaedic Surgery of the Lower Limb and Arthroplasty, Hospital Rummelsberg, Rummelsberg 71, 90592, Schwarzenbruck, Germany
| | - Hans Christoph Pape
- Department of Trauma Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Philipp Lichte
- Department of Orthopaedic Trauma Surgery, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Mersedeh Tohidnezhad
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Frank Hildebrand
- Department of Orthopaedic Trauma Surgery, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Holger Jahr
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
- Department of Orthopaedic Surgery, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
36
|
Yu L, Wang T, Que R, Yang J, Wang Z, Jiang X, Wang L. The potentially protective role of ATP-binding cassette transporters in preeclampsia via Nrf2. Pregnancy Hypertens 2019; 18:21-28. [PMID: 31445436 DOI: 10.1016/j.preghy.2019.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 07/25/2019] [Accepted: 08/09/2019] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Preeclampsia (PE) is a severe placental syndrome that likely results from placental oxidative stress and inflammation, and can lead to maternal hypertension and premature delivery. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) activates several genes involved in antioxidant defense in the placentae, along with the ATP-binding cassette (ABC) transporters which regulate substrate flow between maternal and fetal circulation. Although several ABC transporters are down-regulated in PE, their exact mechanistic role is poorly understood. METHODS& RESULTS: In this study, we compared the levels of major ABC transporters and NRF2 in placentae of healthy full-term pregnant women and those with early and term onset PE. We found a significant decrease in the levels of Nrf2 and several ABC transporters in the placentae of early onset compared to term onset PE. In addition, women with term onset PE showed improved post-partum parameters (lower blood pressure, and greater placental and neonatal weights) compared to those with early onset PE. Mechanistically, Nrf2 knockdown/knockout downregulated the genes for ABC transporters and antioxidant enzymes, and upregulated pro-inflammatory factors, whereas Nrf2 upregulation had the opposite effects. CONCLUSIONS Nrf2 protects the placenta against PE by activating the ABC transporter-mediated efflux, indicating a novel target in PE therapy.
Collapse
Affiliation(s)
- Lu Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Ting Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China; Department of Pharmacy, Sichuan Cancer Hospital & Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Rongcheng Que
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jillian Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, Marshall B. Ketchum University, Fullerton, CA 92831, USA
| | - Zhijun Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Marshall B. Ketchum University, Fullerton, CA 92831, USA
| | - Xuehua Jiang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Ling Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
37
|
Patel A, Vendrell-Gonzalez S, Haas G, Marcinczyk M, Ziemkiewicz N, Talovic M, Fisher JS, Garg K. Regulation of Myogenic Activity by Substrate and Electrical Stimulation In Vitro. Biores Open Access 2019; 8:129-138. [PMID: 31367477 PMCID: PMC6664826 DOI: 10.1089/biores.2019.0016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle has a remarkable regenerative capacity in response to mild injury. However, when muscle is severely injured, muscle regeneration is impaired due to the loss of muscle-resident stem cells, known as satellite cells. Fibrotic tissue, primarily comprising collagen I (COL), is deposited with this critical loss of muscle. In recent studies, supplementation of laminin (LM)-111 has been shown to improve skeletal muscle regeneration in several models of disease and injury. Additionally, electrical stimulation (E-stim) has been investigated as a possible rehabilitation therapy to improve muscle's functional recovery. This study investigated the role of E-stim and substrate in regulating myogenic response. C2C12 myoblasts were allowed to differentiate into myotubes on COL- and LM-coated polydimethylsiloxane molds. The myotubes were subjected to E-stim and compared with nonstimulated controls. While E-stim resulted in increased myogenic activity, irrespective of substrate, LM supported increased proliferation and uniform distribution of C2C12 myoblasts. In addition, C2C12 myoblasts cultured on LM showed higher Sirtuin 1, mammalian target of rapamycin, desmin, nitric oxide, and vascular endothelial growth factor expression. Taken together, these results suggest that an LM substrate is more conducive to myoblast growth and differentiation in response to E-stim in vitro.
Collapse
Affiliation(s)
- Anjali Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Sara Vendrell-Gonzalez
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Gabriel Haas
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Madison Marcinczyk
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Natalia Ziemkiewicz
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Muhamed Talovic
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| | - Jonathan S Fisher
- Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, Missouri
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, Missouri
| |
Collapse
|
38
|
Dhamodharan U, Karan A, Sireesh D, Vaishnavi A, Somasundar A, Rajesh K, Ramkumar KM. Tissue-specific role of Nrf2 in the treatment of diabetic foot ulcers during hyperbaric oxygen therapy. Free Radic Biol Med 2019; 138:53-62. [PMID: 31035003 DOI: 10.1016/j.freeradbiomed.2019.04.031] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/18/2019] [Accepted: 04/25/2019] [Indexed: 12/20/2022]
Abstract
Hyperbaric oxygen (HBO) therapy is proven to be very successful for diabetic foot ulcer (DFU) treatment due to its antimicrobial effect, increased angiogenesis and enhanced collagen synthesis. The molecular mechanism underlying HBO therapy particularly the involvement of Nrf2 in the wound healing process was investigated in the present study. In addition, we have studied the levels of angiogenic markers in ulcer tissues and their correlation with Nrf2 during HBO therapy compared with standard therapy (Non-HBO) for DFU. A total of 32 Patients were recruited and randomized to standard wound care procedure alone (n = 17) or HBO therapy in combination with standard wound care procedure (n = 15) for 20 days. Our results showed that the tissue levels of Nrf2 along with its downstream targets were significantly increased in patients who underwent HBO therapy when compared to Non-HBO therapy. Further, HBO therapy induced angiogenesis as assessed by increased levels of angiogenesis markers such as EGF, VEGF, PDGF, FGF-2 and CXCL10 in the tissue samples. The expressions of eNOS and nitrite concentrations were also significantly increased in HBO therapy when compared to Non-HBO therapy subjects. Moreover, HBO therapy sensitises the macrophages to release FGF-2 and EGF thereby promotes angiogenesis. Further, it increased the levels of neutrophil attractant CXCL-8 thereby promotes the release of chemokine CCL2, a well-known mediator of neovascularization. The Pearson correlation showed that Nrf2 has a positive correlation with EGF, VEGF and PDGF. In conclusion, the findings of the present study suggest that HBO therapy promotes wound healing by increasing oxygen supply and distribution to damaged tissues, stimulating angiogenesis, decreasing inflammation, and increasing the nitrite levels. Increased levels of Nrf2 transiently regulate the expression of angiogenic genes in wound biopsies, which may result in accelerated healing of chronic wounds.
Collapse
Affiliation(s)
- Umapathy Dhamodharan
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamilnadu, India
| | - Amin Karan
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamilnadu, India
| | - Dornadula Sireesh
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamilnadu, India
| | - Alladi Vaishnavi
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamilnadu, India
| | - Arumugam Somasundar
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamilnadu, India
| | - Kesavan Rajesh
- Department of Podiatry, Hycare Super Speciality Hospital, MMDA Colony, Arumbakkam, Chennai, 600 106, Tamilnadu, India.
| | - Kunka Mohanram Ramkumar
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamilnadu, India; Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamilnadu, India.
| |
Collapse
|
39
|
Cox AG, Gurusinghe S, Abd Rahman R, Leaw B, Chan ST, Mockler JC, Murthi P, Marshall SA, Lim R, Wallace EM. Sulforaphane improves endothelial function and reduces placental oxidative stress in vitro. Pregnancy Hypertens 2019; 16:1-10. [PMID: 31056142 DOI: 10.1016/j.preghy.2019.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/21/2019] [Accepted: 02/08/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The maternal endothelial dysfunction characteristic of preeclampsia arises, in part, from excessive placental production of anti-angiogenic factors, including soluble Flt-1, soluble endoglin and activin A, inducing oxidative stress. We assessed whether the antioxidant and NRF2-activator sulforaphane could mitigate endothelial and trophoblast dysfunction in vitro. METHODS We induced dysfunction in human umbilical vein endothelial cells (HUVECs) with TNF-α, assessing endothelial activation and dysfunction (endothelin-1, vascular cell adhesion molecule; VCAM1, intracellular adhesion molecule; ICAM1, e-selectin and endothelial permeability) in the presence or absence of sulforaphane. We also assessed the effects of sulforaphane in mitigating hypoxic and hyperoxic injury in term placental explants by measuring secretion of anti-angiogenic factors. To assess the role of NRF2 we silenced NRF2 in HUVECs and primary trophoblast cells. RESULTS Sulforaphane reduced TNF-α mediated HUVEC secretion of endothelin-1, VCAM1, ICAM1 and E-selectin, and prevented increased endothelial permeability. In placental explants, sulforaphane reduced the secretion of soluble Flt-1, soluble endoglin and activin A. Sulforaphane induced activation and nuclear translocation of NRF2 in HUVECs, inducing heme oxygenase 1. NRF2 silencing blocked some but not all of sulforaphane's effects in HUVECs. NRF2 silencing did not prevent sulforaphane's inhibition of trophobast secretion of soluble Flt-1 or activin A. CONCLUSION In reducing placental and endothelial oxidative stress, sulforaphane may offer a new adjuvant therapeutic approach for the treatment of preeclampsia.
Collapse
Affiliation(s)
- Annie G Cox
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| | - Seshini Gurusinghe
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Rahana Abd Rahman
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| | - Bryan Leaw
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
| | - Siow T Chan
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
| | - Joanne C Mockler
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| | - Padma Murthi
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| | - Sarah A Marshall
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia.
| | - Rebecca Lim
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
| | - Euan M Wallace
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
| |
Collapse
|
40
|
Chen D, Li Z, Bao P, Chen M, Zhang M, Yan F, Xu Y, Ji C, Hu X, Sanchis D, Zhang Y, Ye J. Nrf2 deficiency aggravates Angiotensin II-induced cardiac injury by increasing hypertrophy and enhancing IL-6/STAT3-dependent inflammation. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1253-1264. [PMID: 30668979 DOI: 10.1016/j.bbadis.2019.01.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/07/2019] [Accepted: 01/16/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND NF-E2-related factor 2 (Nrf2) is a transcription factor playing cytoprotective effects in various pathological processes including oxidative stress and cardiac hypertrophy. Despite being a potential therapeutic target to treat several cardiomyopathies, the signaling underlying Nrf2-dependent cardioprotective action remains largely uncharacterized. AIM This study aimed to explore the signaling mediating the role of Nrf2 in the development of hypertensive cardiac pathogenesis by analyzing the response to Angiotensin II (Ang II) in the presence or absence of Nrf2 expression, both in vivo and in vitro. RESULTS Our results indicated that Nrf2 deficiency exacerbated cardiac damage triggered by Ang II infusion. Mechanistically, our study shows that Ang II-triggered hypertrophy and inflammation is exacerbated in the absence of Nrf2 expression and points to the involvement of the IL-6/STAT3 signaling pathway in this event. Indeed, our results show that IL-6 abundance triggered by Ang II is increased in the absence of Nrf2 and demonstrate the requirement of IL-6 in STAT3 activation and cardiac inflammation induced by Ang II. CONCLUSION Our results show that Nrf2 is important for the protection of the heart against Ang II-induced cardiac hypertrophy and inflammation by mechanisms involving the regulation of IL-6/STAT3-dependent signaling.
Collapse
Affiliation(s)
- Dandan Chen
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Zhe Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular research Institute, Wuhan University, Wuhan 430060, China; Hubei key Laboratory of Cardiology, Wuhan 430060, China
| | - Peiqing Bao
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Miao Chen
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Miao Zhang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Yitao Xu
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London W120NN, United Kingdom
| | - Caoyu Ji
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Xinyue Hu
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Daniel Sanchis
- Institut de Recerca Biomedica de Lleida (IRBLLEIDA), Universitat de Lleida, Edifici Biomedicina-I. Av. Rovira Roure, 80, 25198 Lleida, Spain.
| | - Yubin Zhang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China.
| | - Junmei Ye
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China.
| |
Collapse
|
41
|
Sagrillo-Fagundes L, Bienvenue-Pariseault J, Vaillancourt C. Melatonin: The smart molecule that differentially modulates autophagy in tumor and normal placental cells. PLoS One 2019; 14:e0202458. [PMID: 30629581 PMCID: PMC6328125 DOI: 10.1371/journal.pone.0202458] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023] Open
Abstract
Melatonin has protective roles in normal cells and cytotoxic actions in cancer cells, with effects involving autophagy and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) transcription factor pathways. Hypoxia/reoxygenation (H/R) induces oxidative damage and apoptosis. These consequences activate autophagy, which degrades damaged cellular content, as well as activates Nrf2 the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) transcription factor, and thereby the expression of protective genes. Melatonin has protective roles in normal cells and cytotoxic actions in cancer cells, with effects involving autophagy and Nrf2 pathways. The current study shows melatonin to differentially modulate autophagy and Nrf2 pathways in tumor and normal placental cells exposed to H/R. BeWo, a human placental choriocarcinoma cell line, and primary villous cytotrophoblasts isolated from normal term placenta, were maintained in normoxia (8% O2) for 24 h or exposed to hypoxia (0.5% of O2 for 4 h) followed by 20 h of normoxia, creating a situation of H/R, in the presence or absence of 1 mM melatonin. Melatonin induced a 7-fold increase in the activation of 5' adenosine monophosphate-activated protein kinase (AMPK)α, an upstream modulator of autophagy, rising to a 16-fold increase in BeWo cells co-exposed to H/R and melatonin, compared to controls. H/R induced autophagosome formation via the increased expression of Beclin-1 (by 94%) and ATG7 (by 97%) in BeWo cells. Moreover, H/R also induced autophagic activity, indicated by the by the 630% increase in P62, and increased Nrf2 by 314% in BeWo cells. In H/R conditions, melatonin reduced autophagic activity by 74% and Nrf2 expression activation by 300%, leading to BeWo cell apoptosis. In contrast, In human primary villous cytotrophoblasts, H/R induced autophagy and Nrf2, which melatonin further potentiated, thereby affording protection against H/R. This study demonstrates that melatonin differentially modulates autophagy and the Nrf2 pathway in normal vs. tumor trophoblast cells, being cytoprotective in normal cells whilst increasing apoptosis in tumoral trophoblast cells.
Collapse
Affiliation(s)
- Lucas Sagrillo-Fagundes
- INRS-Institut Armand-Frappier and BioMed Research Centre, Laval, QC, Canada
- Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Université du Québec à Montréal, Montréal, QC, Canada
| | - Josianne Bienvenue-Pariseault
- INRS-Institut Armand-Frappier and BioMed Research Centre, Laval, QC, Canada
- Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Université du Québec à Montréal, Montréal, QC, Canada
| | - Cathy Vaillancourt
- INRS-Institut Armand-Frappier and BioMed Research Centre, Laval, QC, Canada
- Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Université du Québec à Montréal, Montréal, QC, Canada
- * E-mail:
| |
Collapse
|
42
|
Zheng L, Jiang WD, Feng L, Wu P, Tang L, Kuang SY, Zeng YY, Zhou XQ, Liu Y. Selenium deficiency impaired structural integrity of the head kidney, spleen and skin in young grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2018; 82:408-420. [PMID: 30142391 DOI: 10.1016/j.fsi.2018.08.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/06/2018] [Accepted: 08/17/2018] [Indexed: 06/08/2023]
Abstract
This study focused on the effects of dietary selenium deficiency on structural integrity of the head kidney, spleen and skin in young grass carp (Ctenopharyngodon idella). A total of 540 healthy grass carp (mean weight 226.48 ± 0.68 g) were randomly divided into six groups and fed six separate diets with graded dietary levels of selenium (0.025-1.049 mg/kg diet) for 80 days. Results showed that selenium deficiency (1) caused oxidative damage in part by reducing the activities of antioxidant enzymes (such as SOD, CAT, GPx, GST and GR) and glutathione (GSH) content, down-regulating the transcript abundances of antioxidant enzymes (except GSTp1) partly related to Kelch-like-ECH-associated protein 1a (Keap1a)/NF-E2-related factor 2 (Nrf2) signalling; (2) aggravated apoptosis in part by up-regulating the mRNA levels of caspase-2, -3, -7, -8 and -9, which were partially related to p38MAPK/FasL/caspase-8 signalling and JNK/(BAX, Bcl-2, Mcl-1b, IAP)/(Apaf1, caspase-9) signalling; (3) damaged the tight junctions in part by down-regulating the mRNA levels of ZO-1 (except spleen), ZO-2 (except spleen), claudin-c, -f, -7, -11 and claudin-15, and up-regulating the mRNA levels of claudin-12, which were partially related to myosin light chain kinase (MLCK) signalling. Interesting, selenium deficiency failed to affect the expression of GSTp1, Keap1a, occludin, claudin-b, claudin-3c, ZO-1 (spleen only) and ZO-2 (spleen only) in the head kidney, spleen and skin of grass carp. Finally, based on the activities of glutathione peroxidase (GPx) and reactive oxygen species (ROS) content in the head kidney, spleen and skin, the dietary selenium requirements for young grass carp were estimated to be 0.558-0.588 mg/kg diet.
Collapse
Affiliation(s)
- Lin Zheng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Yun-Yun Zeng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
43
|
Feng R, Morine Y, Ikemoto T, Imura S, Iwahashi S, Saito Y, Shimada M. Nrf2 activation drive macrophages polarization and cancer cell epithelial-mesenchymal transition during interaction. Cell Commun Signal 2018; 16:54. [PMID: 30180849 PMCID: PMC6122794 DOI: 10.1186/s12964-018-0262-x] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/14/2018] [Indexed: 02/07/2023] Open
Abstract
Background The M2 phenotype of tumor-associated macrophages (TAM) inhibits the anti-tumor inflammation, increases angiogenesis and promotes tumor progression. The transcription factor Nuclear Factor (erythroid-derived 2)-Like 2 (Nrf2) not only modulates the angiogenesis but also plays the anti-inflammatory role through inhibiting pro-inflammatory cytokines expression; however, the role of Nrf2 in the cancer cell and macrophages interaction is not clear. Methods Hepatocellular carcinoma cells (Hep G2 and Huh 7) and pancreatic cancer cells (SUIT2 and Panc-1) were co-cultured with monocytes cells (THP-1) or peripheral blood monocytes derived macrophages, then the phenotype changes of macrophages and epithelial-mesenchymal transition of cancer cells were detected. Also, the role of Nrf2 in cancer cells and macrophages interaction were investigated. Results In this study, we found that cancer cells could induce an M2-like macrophage characterized by up-regulation of CD163 and Arg1, and down-regulation of IL-1b and IL-6 through Nrf2 activation. Also, Nrf2 activation of macrophages promoted VEGF expression. The Nrf2 activation of macrophages correlated with the reactive oxygen species induced by cancer cells derived lactate. Cancer cells educated macrophages could activate Nrf2 of the cancer cells, in turn, to increase cancer cells epithelial-mesenchymal transition (EMT) through paracrine VEGF. These findings suggested that Nrf2 played the important role in the cancer cells and macrophages interaction. Conclusions Macrophage Nrf2 activation by cancer cell-derived lactate skews macrophages polarization towards an M2-like phenotype and educated macrophages activate Nrf2 of the cancer cells to promote EMT of cancer cells. This study provides a new understanding of the role of Nrf2 in the cancer cell and TAM interaction and suggests a potential therapeutic target. Electronic supplementary material The online version of this article (10.1186/s12964-018-0262-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rui Feng
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University of Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Yuji Morine
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University of Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan.
| | - Tetsuya Ikemoto
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University of Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Satoru Imura
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University of Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Shuichi Iwahashi
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University of Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Yu Saito
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University of Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Mitsuo Shimada
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University of Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| |
Collapse
|
44
|
Embryotropic effects of vascular endothelial growth factor on porcine embryos produced by in vitro fertilization. Theriogenology 2018; 120:147-156. [PMID: 30121547 DOI: 10.1016/j.theriogenology.2018.07.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 12/18/2022]
Abstract
Current research suggests that supplementing in vitro culture (IVC) media with vascular endothelial growth factor (VEGF) may have beneficial effects on the development of porcine embryos in vitro. However, the molecular signaling mechanisms underlying this effect are unclear. Therefore, we aimed to investigate the effects of VEGF on molecular signaling events during in vitro embryonic development of porcine embryos. Porcine oocytes matured in vitro were fertilized, and the resultant zygotes were cultured with 5 ng/mL of VEGF supplemented with or without fetal bovine serum from day 4 till day 7. Without VEGF and/or FBS served as the control group. Real-time quantitative PCR was used to detect expression patterns of apoptosis- and oxidative stress-related genes in day 7 blastocysts (BLs). Early-stage apoptosis was detected by annexin-V assays in day 2 and day 7 embryos. We found that the addition of VEGF throughout the culture period with or without FBS supplementation significantly improved embryo survival and development. Supplementation with VEGF in the IVC medium significantly increased early BL formation (p < 0.05), although addition of FBS on day 4 significantly increased hatched BL formation (p < 0.05) regardless of VEGF supplementation. However, supplementation of media with both VEGF and FBS increased the formation of expanded BLs synergistically. The average total cell numbers per BL were significantly (p < 0.05) higher in embryos supplemented with VEGF and FBS than in those supplemented with either VEGF or FBS alone. We also found that accumulation of reactive oxygen species in VEGF-treated embryos was significantly lower (p < 0.05) than that in untreated embryos. The mRNA levels of caspase-3 were significantly lower (p < 0.05), and those of Bcl-2 and Nrf-2 were significantly higher (p < 0.05) in embryos grown in VEGF-supplemented media than in embryos grown in non-supplemented media. Furthermore, on day 2, the numbers of viable embryos (44.06 ± 3.94%) and blastomeres (67.18 ± 3.60%) were significantly higher (p < 0.05), and the numbers of early apoptotic embryos (55.94 ± 3.94) and blastomeres (23.23 ± 4.22) were significantly lower (p < 0.05) in VEGF-treated BLs than in controls. Furthermore, the numbers of early apoptotic cells in BLs on day 7 were also significantly lower (p < 0.05) in VEGF-treated BLs than in controls. Overall, our results indicate that supplementing IVC media with VEGF during in vitro culture of porcine embryos increases their developmental potential.
Collapse
|
45
|
Kikuchi K, Iida M, Ikeda N, Moriyama S, Hamada M, Takahashi S, Kitamura H, Watanabe T, Hasegawa Y, Hase K, Fukuhara T, Sato H, Kobayashi EH, Suzuki T, Yamamoto M, Tanaka M, Asano K. Macrophages Switch Their Phenotype by Regulating Maf Expression during Different Phases of Inflammation. THE JOURNAL OF IMMUNOLOGY 2018; 201:635-651. [PMID: 29907708 DOI: 10.4049/jimmunol.1800040] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/30/2018] [Indexed: 12/14/2022]
Abstract
Macrophages manifest distinct phenotype according to the organs in which they reside. In addition, they flexibly switch their character in adaptation to the changing environment. However, the molecular basis that explains the conversion of the macrophage phenotype has so far been unexplored. We find that CD169+ macrophages change their phenotype by regulating the level of a transcription factor Maf both in vitro and in vivo in C57BL/6J mice. When CD169+ macrophages were exposed to bacterial components, they expressed an array of acute inflammatory response genes in Maf-dependent manner and simultaneously start to downregulate Maf. This Maf suppression is dependent on accelerated degradation through proteasome pathway and microRNA-mediated silencing. The downregulation of Maf unlocks the NF-E2-related factor 2-dominant, cytoprotective/antioxidative program in the same macrophages. The present study provides new insights into the previously unanswered question of how macrophages initiate proinflammatory responses while retaining their capacity to repair injured tissues during inflammation.
Collapse
Affiliation(s)
- Kenta Kikuchi
- Laboratory of Immune Regulation, The School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Mayumi Iida
- Laboratory of Immune Regulation, The School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Naoki Ikeda
- Laboratory of Immune Regulation, The School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Shigetaka Moriyama
- Laboratory of Immune Regulation, The School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Michito Hamada
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Hiroshi Kitamura
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 069-8501, Japan
| | - Takashi Watanabe
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Yoshinori Hasegawa
- Department of Research and Development, Kazusa DNA Research Institute, Kisarazu 292-0818, Japan
| | - Koji Hase
- Division of Biochemistry, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
| | - Takeshi Fukuhara
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.,Laboratory of Oncology, The School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Hideyo Sato
- Department of Medical Technology, Faculty of Medicine, Niigata University, Niigata 951-8518, Japan; and
| | - Eri H Kobayashi
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Takafumi Suzuki
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Masato Tanaka
- Laboratory of Immune Regulation, The School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan;
| | - Kenichi Asano
- Laboratory of Immune Regulation, The School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan;
| |
Collapse
|
46
|
Neves KB, Rios FJ, van der Mey L, Alves-Lopes R, Cameron AC, Volpe M, Montezano AC, Savoia C, Touyz RM. VEGFR (Vascular Endothelial Growth Factor Receptor) Inhibition Induces Cardiovascular Damage via Redox-Sensitive Processes. Hypertension 2018; 71:638-647. [DOI: 10.1161/hypertensionaha.117.10490] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/07/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022]
Abstract
Although VEGF (vascular endothelial growth factor) inhibitors (VEGFIs), are effective anticancer therapies, they cause hypertension through unknown mechanisms. We questioned whether changes in vascular redox state may be important, because VEGF signaling involves nitric oxide (NO) and reactive oxygen species. Molecular mechanisms, including NOS, NADPH oxidase (Nox)–derived reactive oxygen species, antioxidant systems, and vasoconstrictor signaling pathways, were probed in human endothelial cells and vascular smooth muscle exposed to vatalanib, a VEGFI. Vascular functional effects of VEGFI were assessed ex vivo in mouse arteries. Cardiovascular and renal in vivo effects were studied in vatalanib- or gefitinib (EGFI [epidermal growth factor inhibitor])-treated mice. In endothelial cells, vatalanib decreased eNOS (Ser
1177
) phosphorylation and reduced NO and H
2
O
2
production, responses associated with increased Nox-derived O
2
−
and ONOO
−
formation. Inhibition of Nox1/4 (GKT137831) or Nox1 (NoxA1ds), prevented vatalanib-induced effects. Nrf-2 (nuclear factor erythroid 2–related factor 2) nuclear translocation and expression of Nrf-2–regulated antioxidant enzymes were variably downregulated by vatalanib. In human vascular smooth muscles, VEGFI increased Nox activity and stimulated Ca
2+
influx and MLC
20
phosphorylation. Acetylcholine-induced vasodilatation was impaired and U46619-induced vasoconstriction was enhanced by vatalanib, effects normalized by N-acetyl-cysteine and worsened by L-NAME. In vatalanib-, but not gefitinib-treated mice vasorelaxation was reduced and media:lumen ratio of mesenteric arteries was increased with associated increased cardiovascular and renal oxidative stress, decreased Nrf-2 activity and downregulation of antioxidant genes. We demonstrate that inhibition of VEGF signaling induces vascular dysfunction through redox-sensitive processes. Our findings identify Noxs and antioxidant enzymes as novel targets underling VEGFI-induced vascular dysfunction. These molecular processes may contribute to vascular toxicity and hypertension in VEGFI-treated patients.
Collapse
Affiliation(s)
- Karla B. Neves
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Francisco J. Rios
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Lucas van der Mey
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Rheure Alves-Lopes
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Alan C. Cameron
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Massimo Volpe
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Augusto C. Montezano
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Carmine Savoia
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Rhian M. Touyz
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| |
Collapse
|
47
|
Qu HM, Qu LP, Li XY, Pan XZ. Overexpressed HO-1 is associated with reduced STAT3 activation in preeclampsia placenta and inhibits STAT3 phosphorylation in placental JEG-3 cells under hypoxia. Arch Med Sci 2018; 14:597-607. [PMID: 29765448 PMCID: PMC5949914 DOI: 10.5114/aoms.2016.63261] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 08/08/2016] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION Inadequate trophoblast invasion and placentation are widely believed to contribute to preeclampsia, and multiple lines of evidence indicate the involvement of hypoxia in preeclampsia. However, the molecular mechanisms underlying the association of placental hypoxia with preeclampsia are not clear. MATERIAL AND METHODS The present study focused on the role in preeclampsia of heme oxygenase 1 (HO-1), which is an inducible isoform of HO in response to hypoxia, via examining the expression of HO-1 and the expression and phosphorylation (Tyr705) of Signal transducer and activator of transcription (STAT) 3 in preeclamptic placentas via the immunohistochemical method, western blotting assay and RT-qPCR method. Then we investigated the regulation by HO-1 of the expression and phosphorylation of STAT3 in human placental choriocarcinoma JEG-3 cells under hypoxia. RESULTS There was upregulation of HO-1 at both mRNA (1.506 ±0.08347 (N = 37) vs. 1.000 ±0.08854 (N = 31), p < 0.0001) and protein (0.630 ±0.155 (N = 35) vs. 0.310 ±0.052, 0.630 ±0.155 (N = 35), p < 0.001) levels and a reduced level of STAT3 phosphorylation (Tyr 705) in the preeclamptic placental tissues, compared to normal placental tissues (0.143 ±0.027 (N = 35) vs. 0.194 ±0.028 (N = 35), p < 0.01). Also, in vitro experiments demonstrated that HO-1 was markedly promoted by hypoxia in human placental choriocarcinoma JEG-3 cells, 6 or 12 h post treatment (p < 0.05 or p < 0.01). However, the STAT3 phosphorylation (Tyr 705) was attenuated by sustained hypoxia (p < 0.01). Moreover, it was demonstrated that HO-1 overexpression significantly inhibited the hypoxia-promoted STAT3 phosphorylation (Tyr 705). CONCLUSIONS HO-1 was overexpressed in PE placenta, in association with reduced STAT3 phosphorylation (Tyr 705). HO-1 inhibits the STAT3 phosphorylation in placental JEG-3 cells under hypoxia. Thus, we speculate that overexpressed HO-1 might contribute to the reduced STAT3 phosphorylation (Tyr 705) and the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Hong-Mei Qu
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Li-Ping Qu
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Xiao-Yan Li
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Xian-Zhen Pan
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| |
Collapse
|
48
|
Logue OC, Mahdi F, Chapman H, George EM, Bidwell GL. A Maternally Sequestered, Biopolymer-Stabilized Vascular Endothelial Growth Factor (VEGF) Chimera for Treatment of Preeclampsia. J Am Heart Assoc 2017; 6:e007216. [PMID: 29629873 PMCID: PMC5779036 DOI: 10.1161/jaha.117.007216] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/30/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Preeclampsia is a hypertensive syndrome that complicates 3% to 5% of pregnancies in the United States. Preeclampsia originates from an improperly vascularized and ischemic placenta that releases factors that drive systemic pathophysiology. One of these factors, soluble fms-like tyrosine kinase-1, is believed to sequester vascular endothelial growth factor (VEGF), leading to systemic endothelial dysfunction and hypertension. With the goal of targeting soluble fms-like tyrosine kinase-1 while simultaneously preventing fetal exposure to VEGF, we fused VEGF to elastin-like polypeptide, a biopolymer carrier that does not cross the placental barrier (ELP-VEGF). METHODS AND RESULTS ELP-VEGF restored in vitro endothelial cell tube formation in the presence of plasma from placental ischemic rats. Long-term administered ELP-VEGF in pregnant rats accumulated in maternal kidneys, aorta, liver, and placenta, but the protein was undetectable in the pups when administered at therapeutic doses in dams. Long-term administration of ELP-VEGF in a placental ischemia rat model achieved dose-dependent attenuation of hypertension, with blood pressure equal to sham controls at a dose of 5 mg/kg per day. ELP-VEGF infusion increased total plasma soluble fms-like tyrosine kinase-1 levels but dramatically reduced free plasma soluble fms-like tyrosine kinase-1 and induced urinary excretion of nitrate/nitrite, indicating enhanced renal nitric oxide signaling. ELP-VEGF at up to 5 mg/kg per day had no deleterious effect on maternal or fetal body weight. However, dose-dependent adverse events were observed, including ascites production and neovascular tissue encapsulation around the minipump. CONCLUSIONS ELP-VEGF has the potential to treat the preeclampsia maternal syndrome, but careful dosing and optimization of the delivery route are necessary.
Collapse
Affiliation(s)
- Omar C Logue
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
| | - Fakhri Mahdi
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
| | - Heather Chapman
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
| | - Eric M George
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS
| | - Gene L Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
49
|
Sönmez TT, Bayer A, Cremer T, Hock JVP, Lethaus B, Kweider N, Wruck CJ, Drescher W, Jahr H, Lippross S, Pufe T, Tohidnezhad M. The protective effect of platelet released growth factors and bone augmentation (Bio-Oss ® ) on ethanol impaired osteoblasts. Ann Anat 2017; 214:36-42. [DOI: 10.1016/j.aanat.2017.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/17/2017] [Accepted: 07/12/2017] [Indexed: 01/02/2023]
|
50
|
Antioxidant and pro-angiogenic effects of corilagin in rat cerebral ischemia via Nrf2 activation. Oncotarget 2017; 8:114816-114828. [PMID: 29383122 PMCID: PMC5777734 DOI: 10.18632/oncotarget.22023] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 09/23/2017] [Indexed: 01/07/2023] Open
Abstract
The nuclear factor erythroid-2-related factor 2 (Nrf2) pathway has been considered as a potential target for neuroprotection in stroke. The aim of present study was to determine whether corilagin, a novel Nrf2 activator, can protect against ischemia-reperfusion injury and explore the underlying mechanism involved. In vivo, rats exposed to middle cerebral artery occlusion were applied to establish an ischemic stroke model. Posttreatment of corilagin significantly reduced infarct volumes and apoptotic cells as well as improved neurologic score after reperfusion, together with increased vascular density in the ischemic penumbra. Meanwhile, posttreatment with corilagin in MCAO rats significantly decreased malondialdehyde levels, restored the superoxide dismutase and glutathione activity, elevating the Nrf2, heme oxygenase-1, the vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2) expression. However, consecutive intrathecal injection of short interference RNAs targeting Nrf2 at 24-h intervals 72 h before ischemia reduced the beneficial effects of corilagin. In primary cultured neurons, corilagin dose-dependently protected against oxygen and glucose deprivation-induced insult, but the protective effect of corilagin was attenuated by knockdown of Nrf2. In conclusion, these findings indicate that corilagin exerts protective effects against cerebral ischemic injury by attenuating oxidative stress and enhancing angiogenesis via activation of Nrf2 signaling pathway.
Collapse
|