1
|
Oura Y, Ishii M, Miyata H, Ikeda N, Sakurai T, Suehiro F, Komabashiri N, Nishimura M. Evaluation of the effect of platelet-derived growth factor-BB on the biological activity of human mandibular bone marrow-derived mesenchymal stem cells. Arch Oral Biol 2025; 174:106244. [PMID: 40168781 DOI: 10.1016/j.archoralbio.2025.106244] [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: 12/24/2024] [Revised: 03/06/2025] [Accepted: 03/23/2025] [Indexed: 04/03/2025]
Abstract
OBJECTIVE This study aimed to investigate the effects of platelet-derived growth factor-BB (PDGF-BB) on the biological activities of human mandibular bone marrow-derived mesenchymal stem cells (MBMSCs). DESIGN PDGF-BB (20 ng/mL) was used to treat MBMSCs, and its effects on their proliferation, osteogenic differentiation, and migration were evaluated. Cell proliferation was evaluated using a WST-1 assay. Osteogenic differentiation was evaluated by measuring the mineralization potential and alkaline phosphatase activity. Cell migration was evaluated using wound healing and Transwell chamber assays. Cytoskeletal reorganization and adhesion dynamics were evaluated using immunofluorescence staining. Changes in intracellular signaling in MBMSCs induced by PDGF-BB stimulation were evaluated using western blotting. Furthermore, we investigated Girdin signaling as the molecular mechanisms underlying the regulation of PDGF-BB-induced cell migration. RESULTS PDGF-BB treatment did not affect the proliferation or osteogenic differentiation of MBMSCs. PDGF-BB promoted the migration of MBMSCs. PDGF-BB treatment enhanced F-actin filament formation and paxillin localization at the leading edge of cells. PDGF-BB treatment activated Akt signaling in MBMSCs, and the inhibition of Akt signaling effectively suppressed PDGF-BB-induced Akt activation and migration. PDGF-BB promoted the phosphorylation of Girdin in MBMSCs, and the inhibition of Akt signaling attenuated PDGF-BB-induced Girdin activation. CONCLUSION This study demonstrated that PDGF-BB strongly induces the migration of MBMSCs without affecting their proliferation or osteogenic differentiation. Furthermore, PDGF-BB-induced migration of MBMSCs may be mediated through the Akt/Girdin signaling pathway. These findings provide important insight into the molecular mechanisms underlying PDGF-BB-induced periodontal tissue regeneration.
Collapse
Affiliation(s)
- Yurika Oura
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Masakazu Ishii
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8544, Japan.
| | - Haruka Miyata
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Nao Ikeda
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Tomoaki Sakurai
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Fumio Suehiro
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Naohiro Komabashiri
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Masahiro Nishimura
- Department of Oral and Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8544, Japan
| |
Collapse
|
2
|
Reeves J, Tournier P, Becquart P, Carton R, Tang Y, Vigilante A, Fang D, Habib SJ. Rejuvenating aged osteoprogenitors for bone repair. eLife 2024; 13:RP104068. [PMID: 39692737 DOI: 10.7554/elife.104068] [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] [Indexed: 12/19/2024] Open
Abstract
Aging is marked by a decline in tissue regeneration, posing significant challenges to an increasingly older population. Here, we investigate age-related impairments in calvarial bone healing and introduce a novel two-part rejuvenation strategy to restore youthful repair. We demonstrate that aging negatively impacts the calvarial bone structure and its osteogenic tissues, diminishing osteoprogenitor number and function and severely impairing bone formation. Notably, increasing osteogenic cell numbers locally fails to rescue repair in aged mice, identifying the presence of intrinsic cellular deficits. Our strategy combines Wnt-mediated osteoprogenitor expansion with intermittent fasting, which leads to a striking restoration of youthful levels of bone healing. We find that intermittent fasting improves osteoprogenitor function, benefits that can be recapitulated by modulating NAD+-dependent pathways or the gut microbiota, underscoring the multifaceted nature of this intervention. Mechanistically, we identify mitochondrial dysfunction as a key component in age-related decline in osteoprogenitor function and show that both cyclical nutrient deprivation and Nicotinamide mononucleotide rejuvenate mitochondrial health, enhancing osteogenesis. These findings offer a promising therapeutic avenue for restoring youthful bone repair in aged individuals, with potential implications for rejuvenating other tissues.
Collapse
Affiliation(s)
- Joshua Reeves
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Pierre Tournier
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Pierre Becquart
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Robert Carton
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Yin Tang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute Zhejiang University, Zhejiang, China
- Department of Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Alessandra Vigilante
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Dong Fang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute Zhejiang University, Zhejiang, China
- Department of Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Shukry J Habib
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
3
|
Zhang H, Hao J, Hong H, Gu W, Li Z, Sun J, Zhan H, Wei X, Zhou L. Redox signaling regulates the skeletal tissue development and regeneration. Biotechnol Genet Eng Rev 2024; 40:2308-2331. [PMID: 37043672 DOI: 10.1080/02648725.2023.2199244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023]
Abstract
Skeletal tissue development and regeneration in mammals are intricate, multistep, and highly regulated processes. Various signaling pathways have been implicated in the regulation of these processes, including redox. Redox signaling is the signal transduction by electron transfer reactions involving free radicals or related species. Redox homeostasis is essential to cell metabolic states, as the ROS not only regulates cell biological processes but also mediates physiological processes. Following a bone fracture, redox signaling is also triggered to regulate bone healing and regeneration by targeting resident stromal cells, osteoblasts, osteoclasts and endothelial cells. This review will focus on how the redox signaling impact the bone development and bone regeneration.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | - Jin Hao
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | - HaiPing Hong
- FangTa Hospital of Traditional Chinese Medicine, Songjiang Branch, Shanghai, East China, China
| | - Wei Gu
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | | | - Jun Sun
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | - Hongsheng Zhan
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | - Xiaoen Wei
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | - Lin Zhou
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| |
Collapse
|
4
|
Kukulage DSK, Samarasinghe KTG, Matarage Don NNJ, Shivamadhu MC, Shishikura K, Schiff W, Mashhadi Ramezani F, Padmavathi R, Matthews ML, Ahn YH. Protein phosphatase PP2Cα S-glutathionylation regulates cell migration. J Biol Chem 2024; 300:107784. [PMID: 39303918 PMCID: PMC11530597 DOI: 10.1016/j.jbc.2024.107784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
Redox signaling is a fundamental mechanism that controls all major biological processes partly via protein cysteine oxidations, including S-glutathionylation. Despite over 2000 cysteines identified to form S-glutathionylation in databases, the identification of redox cysteines functionally linked to a biological process of interest remains challenging. Here, we demonstrate a strategy combining glutathionylation proteomic database, bioinformatics, and biological screening, which resulted in the identification of S-glutathionylated proteins, including PP2Cα, as redox players of cell migration. We showed that PP2Cα, a prototypical magnesium-dependent serine/threonine phosphatase, is susceptible to S-glutathionylation selectively at nonconserved C314. PP2Cα glutathionylation causes increased migration and invasion of breast cancer cell lines in oxidative stress or upon hydrogen peroxide production. Mechanistically, PP2Cα glutathionylation modulates its protein-protein interactions, activating c-Jun N-terminal kinase and extracellular signal-regulated kinase pathways to elevate migration and invasion. In addition, PP2Cα glutathionylation occurs in response to epidermal growth factor, supporting a serine/threonine phosphatase PP2Cα as a new redox player in growth factor signal transduction.
Collapse
Affiliation(s)
| | | | | | - Madhu C Shivamadhu
- Department of Chemistry, Drexel University, Philadelphia, Pennsylvania, USA
| | - Kyosuke Shishikura
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William Schiff
- Department of Chemistry, Drexel University, Philadelphia, Pennsylvania, USA
| | | | | | - Megan L Matthews
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Young-Hoon Ahn
- Department of Chemistry, Drexel University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
5
|
Jornayvaz FR, Gariani K, Somm E, Jaquet V, Bouzakri K, Szanto I. NADPH oxidases in healthy white adipose tissue and in obesity: function, regulation, and clinical implications. Obesity (Silver Spring) 2024; 32:1799-1811. [PMID: 39315402 DOI: 10.1002/oby.24113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/19/2024] [Accepted: 06/11/2024] [Indexed: 09/25/2024]
Abstract
Reactive oxygen species, when produced in a controlled manner, are physiological modulators of healthy white adipose tissue (WAT) expansion and metabolic function. By contrast, unbridled production of oxidants is associated with pathological WAT expansion and the establishment of metabolic dysfunctions, most notably insulin resistance and type 2 diabetes mellitus. NADPH oxidases (NOXs) produce oxidants in an orderly fashion and are present in adipocytes and in other diverse WAT-constituent cell types. Recent studies have established several links between aberrant NOX-derived oxidant production, adiposity, and metabolic homeostasis. The objective of this review is to highlight the physiological roles attributed to diverse NOX isoforms in healthy WAT and summarize current knowledge of the metabolic consequences related to perturbations in their adequate oxidant production. We detail WAT-related alterations in preclinical investigations conducted in NOX-deficient murine models. In addition, we review clinical studies that have employed NOX inhibitors and currently available data related to human NOX mutations in metabolic disturbances. Future investigations aimed at understanding the integration of NOX-derived oxidants in the regulation of the WAT cellular redox network are essential for designing successful redox-related precision therapies to curb obesity and attenuate obesity-associated metabolic pathologies.
Collapse
Affiliation(s)
- François R Jornayvaz
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Karim Gariani
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Emmanuel Somm
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Vincent Jaquet
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
- RE.A.D.S. Unit (Readers, Assay Development and Screening Unit), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Karim Bouzakri
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Ildiko Szanto
- Service of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Department of Internal Medicine, Geneva University Hospitals and University of Geneva Medical School, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
6
|
Kukulage DSK, Yapa Abeywardana M, Matarage Don NNJ, Hu RM, Shishikura K, Matthews ML, Ahn YH. Chemoproteomic strategy identified p120-catenin glutathionylation regulates E-cadherin degradation and cell migration. Cell Chem Biol 2023; 30:1542-1556.e9. [PMID: 37714153 PMCID: PMC10840712 DOI: 10.1016/j.chembiol.2023.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 05/30/2023] [Accepted: 08/15/2023] [Indexed: 09/17/2023]
Abstract
Identification of cysteines with high oxidation susceptibility is important for understanding redox-mediated biological processes. In this report, we report a chemical proteomic strategy that finds cysteines with high susceptibility to S-glutathionylation. Our proteomic strategy, named clickable glutathione-based isotope-coded affinity tag (G-ICAT), identified 1,518 glutathionylated cysteines while determining their relative levels of glutathionylated and reduced forms upon adding hydrogen peroxide. Among identified cysteines, we demonstrated that CTNND1 (p120) C692 has high susceptibility to glutathionylation. Also, p120 wild type (WT), compared to C692S, induces its dissociation from E-cadherin under oxidative stress, such as glucose depletion. p120 and E-cadherin dissociation correlated with E-cadherin destabilization via its proteasomal degradation. Lastly, we showed that p120 WT, compared to C692S, increases migration and invasion of MCF7 cells under glucose depletion, supporting a model that p120 C692 glutathionylation increases cell migration and invasion by destabilization of E-cadherin, a core player in cell-cell adhesion.
Collapse
Affiliation(s)
| | | | | | - Ren-Ming Hu
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyosuke Shishikura
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Megan L Matthews
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Young-Hoon Ahn
- Department of Chemistry, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
7
|
Thannickal VJ, Jandeleit‐Dahm K, Szyndralewiez C, Török NJ. Pre-clinical evidence of a dual NADPH oxidase 1/4 inhibitor (setanaxib) in liver, kidney and lung fibrosis. J Cell Mol Med 2023; 27:471-481. [PMID: 36658776 PMCID: PMC9930438 DOI: 10.1111/jcmm.17649] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/21/2022] [Accepted: 11/26/2022] [Indexed: 01/21/2023] Open
Abstract
Fibrosis describes a dysregulated tissue remodelling response to persistent cellular injury and is the final pathological consequence of many chronic diseases that affect the liver, kidney and lung. Nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase (NOX) enzymes produce reactive oxygen species (ROS) as their primary function. ROS derived from NOX1 and NOX4 are key mediators of liver, kidney and lung fibrosis. Setanaxib (GKT137831) is a first-in-class, dual inhibitor of NOX1/4 and is the first NOX inhibitor to progress to clinical trial investigation. The anti-fibrotic effects of setanaxib in liver, kidney and lung fibrosis are supported by multiple lines of pre-clinical evidence. However, despite advances in our understanding, the precise roles of NOX1/4 in fibrosis require further investigation. Additionally, there is a translational gap between the pre-clinical observations of setanaxib to date and the applicability of these to human patients within a clinical setting. This narrative review critically examines the role of NOX1/4 in liver, kidney and lung fibrosis, alongside the available evidence investigating setanaxib as a therapeutic agent in pre-clinical models of disease. We discuss the potential clinical translatability of this pre-clinical evidence, which provides rationale to explore NOX1/4 inhibition by setanaxib across various fibrotic pathologies in clinical trials involving human patients.
Collapse
Affiliation(s)
- Victor J. Thannickal
- John W. Deming Department of MedicineTulane University School of MedicineNew OrleansLouisianaUSA
- Southeast Louisiana Veterans Healthcare SystemNew OrleansLouisianaUSA
| | - Karin Jandeleit‐Dahm
- Department of Diabetes, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Cédric Szyndralewiez
- Calliditas Therapeutics Suisse SAGenevaSwitzerland
- Present address:
Pherecydes PharmaNantesFrance
| | - Natalie J. Török
- Division of Gastroenterology and Hepatology, Department of MedicineStanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
8
|
Li M, Tian W, Wang F, Yang C, Zhang L, Tang Q, Liu S, Wang F. Nicotinamide nucleotide transhydrogenase mutation analysis in Chinese patients with thyroid dysgenesis. Am J Med Genet A 2021; 188:89-98. [PMID: 34545694 DOI: 10.1002/ajmg.a.62493] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/27/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022]
Abstract
Thyroid dysgenesis (TD) accounts for 80% cases of congenital hypothyroidism, which is the most common neonatal disorder. Until now, the gene mutations have been reported associated with TD can only account for 5% cases, suggesting the genetic heterogeneity of the pathology. Nicotinamide nucleotide transhydrogenase (NNT) plays a crucial role in regulating redox homeostasis, patients carrying NNT mutations have been described with a clinical phenotype of hypothyroidism. As TD risk is increased in the context of several syndromes and redox homeostasis is vital for thyroid development and function, NNT might be a candidate gene involved in syndromic TD. Therefore, we performed target sequencing (TS) in 289 TD patients for causative mutations in NNT and conducted functional analysis of the gene mutations. TS and Sanger sequence were used to screen the novel mutations. For functional analysis, we performed western blot, measurement of NADPH/NADPtotal and H2 O2 generation, cell proliferation, and wounding healing assay. As a result, three presumably pathogenic mutations (c.811G > A, p.Ala271Ser; c.2078G > A, p.Arg693His; and c.2581G > A, p.Val861Met) in NNT had been identified. Our results showed the damaging effect of NNT mutations on stability and catalytic activity of proteins and redox balance of cells. In conclusion, our findings provided novel insights into the role of the NNT isotype in thyroid physiopathology and broaden the spectrum of pathogenic genes associated with TD. However, the pathogenic mechanism of NNT in TD is still need to be investigated in further study.
Collapse
Affiliation(s)
- Miaomiao Li
- The Affiliated Hospital of Qingdao University, Medical Genetic Department, Prenatal Diagnosis Center, Qingdao, China
| | - Weibing Tian
- Weifang Maternal and Child Health Hospital, Newborn Screening Center, Weifang, China
| | - Fengqi Wang
- The Affiliated Hospital of Qingdao University, Medical Genetic Department, Prenatal Diagnosis Center, Qingdao, China
| | - Chengyu Yang
- The Affiliated Hospital of Qingdao University, Medical Genetic Department, Prenatal Diagnosis Center, Qingdao, China
| | - Lu Zhang
- The Affiliated Hospital of Qingdao University, Medical Genetic Department, Prenatal Diagnosis Center, Qingdao, China
| | - Qian Tang
- The Affiliated Hospital of Qingdao University, Medical Genetic Department, Prenatal Diagnosis Center, Qingdao, China
| | - Shiguo Liu
- The Affiliated Hospital of Qingdao University, Medical Genetic Department, Prenatal Diagnosis Center, Qingdao, China
| | - Fang Wang
- The Affiliated Hospital of Qingdao University, Department of Endocrinology, Qingdao, China
| |
Collapse
|
9
|
Maraldi T, Angeloni C, Prata C, Hrelia S. NADPH Oxidases: Redox Regulators of Stem Cell Fate and Function. Antioxidants (Basel) 2021; 10:973. [PMID: 34204425 PMCID: PMC8234808 DOI: 10.3390/antiox10060973] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
One of the major sources of reactive oxygen species (ROS) generated within stem cells is the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family of enzymes (NOXs), which are critical determinants of the redox state beside antioxidant defense mechanisms. This balance is involved in another one that regulates stem cell fate: indeed, self-renewal, proliferation, and differentiation are decisive steps for stem cells during embryo development, adult tissue renovation, and cell therapy application. Ex vivo culture-expanded stem cells are being investigated for tissue repair and immune modulation, but events such as aging, senescence, and oxidative stress reduce their ex vivo proliferation, which is crucial for their clinical applications. Here, we review the role of NOX-derived ROS in stem cell biology and functions, focusing on positive and negative effects triggered by the activity of different NOX isoforms. We report recent findings on downstream molecular targets of NOX-ROS signaling that can modulate stem cell homeostasis and lineage commitment and discuss the implications in ex vivo expansion and in vivo engraftment, function, and longevity. This review highlights the role of NOX as a pivotal regulator of several stem cell populations, and we conclude that these aspects have important implications in the clinical utility of stem cells, but further studies on the effects of pharmacological modulation of NOX in human stem cells are imperative.
Collapse
Affiliation(s)
- Tullia Maraldi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy;
| | - Cristina Angeloni
- School of Pharmacy, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy;
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum—University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy;
| |
Collapse
|
10
|
Moldogazieva NT, Mokhosoev IM, Mel'nikova TI, Zavadskiy SP, Kuz'menko AN, Terentiev AA. Dual Character of Reactive Oxygen, Nitrogen, and Halogen Species: Endogenous Sources, Interconversions and Neutralization. BIOCHEMISTRY (MOSCOW) 2020; 85:S56-S78. [PMID: 32087054 DOI: 10.1134/s0006297920140047] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oxidative stress resulting from accumulation of reactive oxygen, nitrogen, and halogen species (ROS, RNS, and RHS, respectively) causes the damage of cells and biomolecules. However, over the long evolutionary time, living organisms have developed the mechanisms for adaptation to oxidative stress conditions including the activity of the antioxidant system (AOS), which maintains low intracellular levels of RONS (ROS and RNS) and RHS. Moreover, living organisms have adapted to use low concentrations of these electrophiles for the regulation of cell functions through the reversible post-translational chemical modifications of redox-sensitive amino acid residues in intracellular effectors of signal transduction pathways (protein kinases and protein phosphatases), transcription factors, etc. An important fine-tuning mechanism that ensures involvement of RONS and RHS in the regulation of physiological processes is interconversion between different reactive species. This review focuses on the complex networks of interacting RONS and RHS types and their endogenous sources, such as NOX family of NADPH oxidases, complexes I and III of the mitochondrial electron transport chain, NO synthases, cytochrome P450-containing monooxygenase system, xanthine oxidoreductase, and myeloperoxidases. We highlight that kinetic parameters of reactions involving RONS and RHS determine the effects of these reactive species on cell functions. We also describe the functioning of enzymatic and non-enzymatic AOS components and the mechanisms of RONS and RHS scavenging under physiological conditions. We believe that analysis of interactions between RONS and relationships between different endogenous sources of these compounds will contribute to better understanding of their role in the maintenance of cell redox homeostasis as well as initiation and progression of diseases.
Collapse
Affiliation(s)
- N T Moldogazieva
- Sechenov First Moscow State Medical University, Moscow, 119991, Russia.
| | - I M Mokhosoev
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia.
| | - T I Mel'nikova
- Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - S P Zavadskiy
- Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - A N Kuz'menko
- Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - A A Terentiev
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
11
|
Lyons OT, Saha P, Smith A. Redox dysregulation in the pathogenesis of chronic venous ulceration. Free Radic Biol Med 2020; 149:23-29. [PMID: 31560951 DOI: 10.1016/j.freeradbiomed.2019.09.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/04/2019] [Accepted: 09/20/2019] [Indexed: 12/23/2022]
Abstract
In chronic venous ulcers (CVUs), which account for up to 75% of leg ulcers, the inflammatory stage of wound healing fails to down-regulate, preventing progression to proliferation, remodeling and eventual epithelialisation. The roles of reactive oxygen species (ROS) in the oxidative burst and pathogen killing are well known, but ROS also have important functions in extra-cellular and intra-cellular signalling. Iron deposition, resulting from venous reflux, primes macrophages towards a persistent inflammatory response, with ongoing stimulation by bacteria potentially playing a role. Generation of excessive ROS by activated inflammatory cells causes tissue destruction and disintegration of the dermis, and then at later stages, a failure to heal. Here, we review the evidence for ROS in CVU formation and in normal and delayed healing. We also discuss how ROS modulation might be used to influence the healing of these complex wounds, which cause long-term morbidity and are associated with a significant financial burden to healthcare systems.
Collapse
Affiliation(s)
- Oliver Ta Lyons
- Academic Department of Vascular Surgery, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, St Thomas' Hospital, United Kingdom; Basildon and Thurrock University Hospitals NHS Foundation Trust, United Kingdom
| | - Prakash Saha
- Academic Department of Vascular Surgery, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, St Thomas' Hospital, United Kingdom
| | - Alberto Smith
- Academic Department of Vascular Surgery, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, St Thomas' Hospital, United Kingdom.
| |
Collapse
|
12
|
Burtenshaw D, Kitching M, Redmond EM, Megson IL, Cahill PA. Reactive Oxygen Species (ROS), Intimal Thickening, and Subclinical Atherosclerotic Disease. Front Cardiovasc Med 2019; 6:89. [PMID: 31428618 PMCID: PMC6688526 DOI: 10.3389/fcvm.2019.00089] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
Arteriosclerosis causes significant morbidity and mortality worldwide. Central to this process is the development of subclinical non-atherosclerotic intimal lesions before the appearance of pathologic intimal thickening and advanced atherosclerotic plaques. Intimal thickening is associated with several risk factors, including oxidative stress due to reactive oxygen species (ROS), inflammatory cytokines and lipid. The main ROS producing systems in-vivo are reduced nicotinamide dinucleotide phosphate (NADPH) oxidase (NOX). ROS effects are context specific. Exogenous ROS induces apoptosis and senescence, whereas intracellular ROS promotes stem cell differentiation, proliferation, and migration. Lineage tracing studies using murine models of subclinical atherosclerosis have revealed the contributory role of medial smooth muscle cells (SMCs), resident vascular stem cells, circulating bone-marrow progenitors and endothelial cells that undergo endothelial-mesenchymal-transition (EndMT). This review will address the putative physiological and patho-physiological roles of ROS in controlling vascular cell fate and ROS contribution to vascular regeneration and disease progression.
Collapse
Affiliation(s)
- Denise Burtenshaw
- Vascular Biology & Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| | | | - Eileen M Redmond
- Department of Surgery, University of Rochester, Rochester, NY, United States
| | - Ian L Megson
- Centre for Health Science, UHI Institute of Health Research and Innovation, Inverness, United Kingdom
| | - Paul A Cahill
- Vascular Biology & Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
13
|
Blázquez R, Sánchez-Margallo FM, Reinecke J, Álvarez V, López E, Marinaro F, Casado JG. Conditioned Serum Enhances the Chondrogenic and Immunomodulatory Behavior of Mesenchymal Stem Cells. Front Pharmacol 2019; 10:699. [PMID: 31316380 PMCID: PMC6609570 DOI: 10.3389/fphar.2019.00699] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/29/2019] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis is one of the most common chronic health conditions associated with pain and disability. Advanced therapies based on mesenchymal stem cells have become valuable options for the treatment of these pathologies. Conditioned serum (CS, “Orthokine”) has been used intra-articularly for osteoarthritic patients. In this work, we hypothesized that the rich content on anti-inflammatory proteins and growth factors of CS may exert a beneficial effect on the biological activity of human adipose-derived mesenchymal stem cells (hAdMSCs). In vitro studies were designed using hAdMSCs cocultured with CS at different concentrations (2.5, 5, and 10%). Chondrogenic differentiation assays and immunomodulatory experiments using in vitro-stimulated lymphocytes were performed. Our results demonstrated that CS significantly enhanced the differentiation of hAdMSCs toward chondrocytes. Moreover, hAdMSCs pre-sensitized with CS reduced the lymphocyte proliferation as well as their differentiation toward activated lymphocytes. These results suggest that in vivo coadministration of CS and hAdMSCs may have a beneficial effect on the therapeutic potential of hAdMSCs. Moreover, these results indicate that intra-articular administration of CS might influence the biological behavior of resident stem cells increasing their chondrogenic differentiation and inherent immunomodulatory activity. To our knowledge, this is the first in vitro study reporting this combination.
Collapse
Affiliation(s)
- Rebeca Blázquez
- Stem Cell Therapy Unit, "Jesús Usón" Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - Francisco Miguel Sánchez-Margallo
- Stem Cell Therapy Unit, "Jesús Usón" Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - Julio Reinecke
- Research and Development Department, ORTHOGEN AG, Düsseldorf, Germany
| | - Verónica Álvarez
- Stem Cell Therapy Unit, "Jesús Usón" Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Esther López
- Stem Cell Therapy Unit, "Jesús Usón" Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Federica Marinaro
- Stem Cell Therapy Unit, "Jesús Usón" Minimally Invasive Surgery Centre, Cáceres, Spain
| | - Javier G Casado
- Stem Cell Therapy Unit, "Jesús Usón" Minimally Invasive Surgery Centre, Cáceres, Spain.,CIBER de Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| |
Collapse
|
14
|
Rharass T, Lucas S. High Glucose Level Impairs Human Mature Bone Marrow Adipocyte Function Through Increased ROS Production. Front Endocrinol (Lausanne) 2019; 10:607. [PMID: 31551934 PMCID: PMC6746912 DOI: 10.3389/fendo.2019.00607] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022] Open
Abstract
Bone marrow adipocytes (BMAds) accumulate in aging, menopause, and metabolic diseases such as Type 2 diabetes. These osteoporotic conditions are associated with oxidative stress and hyperglycemia which are both considered as critical factors underlying bone fragility. Glucose excess and reactive oxygen species (ROS) are known to favor adipogenesis over osteoblastogenesis. In this study, we investigated whether high glucose exposure could determine dysfunction of mature BMAds, specifically through ROS production. The effects of low (LG, 5 mM) or high glucose (HG, 25 mM) concentrations were examined using human bone mesenchymal stromal cells (hBMSCs) in the time course of differentiation, and, up to 21 days once adipocytes were mature. HG did not alter the adipocyte differentiation process of hBMSCs. Yet, after 21 days under HG exposure, PPARG, CEBPA, and adiponectin mRNA expressions were decreased. These alterations were also observed following adipogenic inducer withdrawal as well as in adipocytes fully differentiated in LG then cultured in HG for the last 11 days. Without inducers, HG condition also led to decreased leptin mRNA level. Importantly, intracellular and extracellular ROS concentrations measured using Amplex Red were significantly raised by 50% under HG exposure. This rise was observed once adipocytes ended differentiation and was reproduced within the different cell culture settings without any cytotoxicity. Among genes involved in ROS metabolism, the mRNA level of the H2O2 generating enzyme NOX4 was found upregulated in the presence of HG. Following cell separation, mature BMAds were shown to overproduce ROS and to display the gene alterations in contrast to non-lipid-laden cells. Finally, a non-lethal treatment with a pro-oxidant agent under LG condition reduces the mRNA levels of PPARG, adiponectin, and leptin as the HG condition does in the absence of inducers, and amplifies the effect of glucose excess on gene expression. HG concentration drives mature BMAds toward altered expression of the main adipokines and transcriptional factors. These perturbations are associated with a rise in ROS generation likely mediated through enhanced expression of NOX4. Mature BMAds are thus responsive to changes in glucose and ROS concentrations, which is relevant regarding with their phenotype and function in age- or metabolic disease-related osteoporosis.
Collapse
|
15
|
Achyranthes bidentata polypeptides promotes migration of Schwann cells via NOX4/DUOX2-dependent ROS production in rats. Neurosci Lett 2018; 696:99-107. [PMID: 30572102 DOI: 10.1016/j.neulet.2018.12.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/29/2018] [Accepted: 12/16/2018] [Indexed: 12/30/2022]
Abstract
Achyranthes bidentata polypeptides (ABPP), an active polypeptides isolated from the aqueous extract of Achyranthes bidentata Blume, contributes to the regeneration of injured peripheral nerves by promoting migration of Schwann cells (SCs). In this study, we aimed to investigate the possible mechanism underlying the ABPP-induced migration of primary cultured rat SCs. Transwell migration assays indicated that ABPP promoted SCs migration in a concentration-dependent manner by inducing production of NADPH-oxidase (NOX)-derived reactive oxygen species (ROS). Inhibition of ROS production by NOXs inhibitor apocynin (APO) or diphenyleneiodonium (DPI) partially blocked ABPP-mediated SCs migration. Furthermore, by using real-time polymerase chain reaction analysis and siRNA interference technique, we verified the participation of NOX subunit 4 (NOX4) and dual oxidase 2 (DUOX2) in ABPP-induced ROS production and consequential SCs migration. Taken together, these results demonstrated that ABPP promoted SCs migration via NOX4/DUOX2-activated ROS in SCs.
Collapse
|
16
|
Rashid MI, Ali A, Andleeb S. Functional Annotation and Analysis of Dual Oxidase 1 (DUOX1): a Potential Anti-pyocyanin Immune Component. Interdiscip Sci 2018; 11:597-610. [PMID: 30483939 DOI: 10.1007/s12539-018-0308-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 10/24/2018] [Accepted: 10/24/2018] [Indexed: 11/27/2022]
Abstract
Dual Oxidase 1 (DUOX1) is a prominent immune system component primarily expressed in esophagus, lungs, skin, and urinary bladder including others. DUOX1 is involved in lactoperoxidase-mediated innate immunity at mucosal surfaces by generation of antimicrobial hypothiocyanite at the apical surface of epithelial lining. Upon detection of bacterial pathogens mainly Pseudomonas aeruginosa, DUOX1 is activated in bronchial epithelial cells. Both the host and pathogen enter a redox dual with DUOX1 and hypothiocyanite from host and Pyocyanin (PCN) as a redox active virulence factor from P. aeruginosa. The synergy of the both enzymes permanently oxidizes PCN and thus holds the potential to prevent PCN-induced virulence, which otherwise paves the way for establishment of persistent chronic infection. In this study, we structurally and functionally annotated the DUOX1, predicted its 3d structure, physio-chemical properties, post-translational modifications, and genetic polymorphism analysis with subsequent disease-associated single-nucleotide variations and their impact on DUOX1 functionality by employing in silico approaches. DUOX1 holds greater homology with gorilla and chimpanzee than other primates. The localization signal peptide was present at the beginning of the peptide with cleavage site at 22 aa position. Three distinct functional domains were observed based on homology: An_peroxidase, FRQ1, and oxido-reductase domains. Polymorphism analysis revealed > 60 SNPs associated with different cancers with probable damaging effects. No cancer-associated methylated island was observed for DUOX1. Three-dimensional structure was developed via homology modeling strategy. The proper annotation will help in characterization of DUOX1 and enhance our knowledge of its functionality and biological roles.
Collapse
Affiliation(s)
- Muhammad Ibrahim Rashid
- Department of Industrial Biotechnology, Atta ur Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Amjad Ali
- Department of Industrial Biotechnology, Atta ur Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Saadia Andleeb
- Department of Industrial Biotechnology, Atta ur Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan.
| |
Collapse
|
17
|
Tyurin-Kuzmin PA, Dyikanov DT, Fadeeva JI, Sysoeva VY, Kalinina NI. Flow cytometry analysis of adrenoceptors expression in human adipose-derived mesenchymal stem/stromal cells. Sci Data 2018; 5:180196. [PMID: 30277480 PMCID: PMC6167953 DOI: 10.1038/sdata.2018.196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/02/2018] [Indexed: 01/23/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) were identified in most tissues of an adult organism. MSCs mediate physiological renewal, as well as regulation of tissue homeostasis, reparation and regeneration. Functions of MSCs are regulated by endocrine and neuronal signals, and noradrenaline is one of the most important MSC regulators. We provided flow cytometry analysis of expression of adrenergic receptors on the surface of human MSCs isolated from ten different donors. We have found that the expression profile of adrenergic receptors in MSCs vary significantly between donors. We also showed that alpha1A-adrenoceptor expression is upregulated under the action of noradrenaline. We share our flow cytometry raw data, as well as processing of these data on a flow cytometry repository for freely downloading.
Collapse
Affiliation(s)
- Pyotr A. Tyurin-Kuzmin
- Department of Biochemistry and Molecular Biology, Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Daniyar T. Dyikanov
- Department of Biochemistry and Molecular Biology, Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Julia I. Fadeeva
- Department of Biochemistry and Molecular Biology, Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Veronika Yu. Sysoeva
- Department of Biochemistry and Molecular Biology, Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Natalia I. Kalinina
- Department of Biochemistry and Molecular Biology, Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
18
|
Feng D, Zhang L, Ding F, Yang F, Ma W, Han Z, Hua B, Wang X, Yu Y, Huang Q, Lei L, Pan Z, Cai B. Blocking Nox2 improves mesenchymal stem cells therapy in myocardial infarction via antagonizing oxidant and promoting survival. J Cell Physiol 2018; 233:7004-7015. [PMID: 29744879 DOI: 10.1002/jcp.26623] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 03/28/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Dan Feng
- Department of Pharmacy The Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang Province China
- Shanxian Central Hospital Heze Shandong Province China
| | - Lai Zhang
- Department of Pharmacy The Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang Province China
- Department of Pharmacology (Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy Harbin Medical University Harbin Heilongjiang Province China
| | - Fengzhi Ding
- Department of Pharmacy The Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang Province China
- Department of Pharmacology (Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy Harbin Medical University Harbin Heilongjiang Province China
| | - Fan Yang
- Department of Pharmacy The Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang Province China
- Department of Pharmacology (Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy Harbin Medical University Harbin Heilongjiang Province China
| | - Wenya Ma
- Department of Pharmacy The Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang Province China
- Department of Pharmacology (Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy Harbin Medical University Harbin Heilongjiang Province China
| | - Zhenbo Han
- Department of Pharmacy The Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang Province China
- Department of Pharmacology (Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy Harbin Medical University Harbin Heilongjiang Province China
| | - Bingjie Hua
- Department of Pharmacy The Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang Province China
- Department of Pharmacology (Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy Harbin Medical University Harbin Heilongjiang Province China
| | - Xiuxiu Wang
- Department of Pharmacy The Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang Province China
- Department of Pharmacology (Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy Harbin Medical University Harbin Heilongjiang Province China
| | - Ying Yu
- Department of Pharmacy The Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang Province China
- Department of Pharmacology (Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy Harbin Medical University Harbin Heilongjiang Province China
| | - Qi Huang
- Department of Pharmacy The Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang Province China
- Department of Pharmacology (Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy Harbin Medical University Harbin Heilongjiang Province China
| | - Lei Lei
- Department of Histology and Embryology Harbin Medical University Harbin Heilongjiang Province China
| | - Zhenwei Pan
- Department of Pharmacology (Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy Harbin Medical University Harbin Heilongjiang Province China
| | - Benzhi Cai
- Department of Pharmacy The Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang Province China
- Department of Pharmacology (Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy Harbin Medical University Harbin Heilongjiang Province China
- Institute of Clinical Pharmacy, the Heilongjiang Province Key Laboratory of Drug Research Harbin Medical University Harbin Heilongjiang Province China
| |
Collapse
|
19
|
Moldogazieva NT, Mokhosoev IM, Feldman NB, Lutsenko SV. ROS and RNS signalling: adaptive redox switches through oxidative/nitrosative protein modifications. Free Radic Res 2018; 52:507-543. [PMID: 29589770 DOI: 10.1080/10715762.2018.1457217] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Over the last decade, a dual character of cell response to oxidative stress, eustress versus distress, has become increasingly recognized. A growing body of evidence indicates that under physiological conditions, low concentrations of reactive oxygen and nitrogen species (RONS) maintained by the activity of endogenous antioxidant system (AOS) allow reversible oxidative/nitrosative modifications of key redox-sensitive residues in regulatory proteins. The reversibility of redox modifications such as Cys S-sulphenylation/S-glutathionylation/S-nitrosylation/S-persulphidation and disulphide bond formation, or Tyr nitration, which occur through electrophilic attack of RONS to nucleophilic groups in amino acid residues provides redox switches in the activities of signalling proteins. Key requirement for the involvement of the redox modifications in RONS signalling including ROS-MAPK, ROS-PI3K/Akt, and RNS-TNF-α/NF-kB signalling is their specificity provided by a residue microenvironment and reaction kinetics. Glutathione, glutathione peroxidases, peroxiredoxins, thioredoxin, glutathione reductases, and glutaredoxins modulate RONS level and cell signalling, while some of the modulators (glutathione, glutathione peroxidases and peroxiredoxins) are themselves targets for redox modifications. Additionally, gene expression, activities of transcription factors, and epigenetic pathways are also under redox regulation. The present review focuses on RONS sources (NADPH-oxidases, mitochondrial electron-transportation chain (ETC), nitric oxide synthase (NOS), etc.), and their cross-talks, which influence reversible redox modifications of proteins as physiological phenomenon attained by living cells during the evolution to control cell signalling in the oxygen-enriched environment. We discussed recent advances in investigation of mechanisms of protein redox modifications and adaptive redox switches such as MAPK/PI3K/PTEN, Nrf2/Keap1, and NF-κB/IκB, powerful regulators of numerous physiological processes, also implicated in various diseases.
Collapse
Affiliation(s)
- N T Moldogazieva
- a Department of Biotechnology, I.M. Sechenov First Moscow State Medical University (Sechenov University) , Moscow , Russia
| | - I M Mokhosoev
- a Department of Biotechnology, I.M. Sechenov First Moscow State Medical University (Sechenov University) , Moscow , Russia
| | - N B Feldman
- a Department of Biotechnology, I.M. Sechenov First Moscow State Medical University (Sechenov University) , Moscow , Russia
| | - S V Lutsenko
- a Department of Biotechnology, I.M. Sechenov First Moscow State Medical University (Sechenov University) , Moscow , Russia
| |
Collapse
|
20
|
Tyurin-Kuzmin PA, Karagyaur MN, Rubtsov YP, Dyikanov DT, Vasiliev PA, Vorotnikov AV. CRISPR/Cas9-mediated modification of the extreme C-terminus impairs PDGF-stimulated activity of Duox2. Biol Chem 2018; 399:437-446. [DOI: 10.1515/hsz-2017-0229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/11/2018] [Indexed: 11/15/2022]
Abstract
Abstract
Duox2 belongs to the large family of NADPH-oxidase enzymes that are implicated in immune response, vasoregulation, hormone synthesis, cell growth and differentiation via the regulated synthesis of H2O2 and reactive oxygen species. We and others have shown that Duox2 and H2O2 are involved in platelet-derived growth factor (PDGF) induced migration of fibroblasts. Now, using the CRISPR/Cas9-mediated genome editing we demonstrate that the extreme C-terminal region of Duox2 is required for PDGF-stimulated activity of Duox2 and H2O2 production. We generated the fibroblast cells that stably co-express the wild-type or C-terminally modified Duox2 and fluorescent H2O2 probe Hyper. We found that nonsense substitution of the last 23 amino acids in Duox2 results in complete loss of PDGF stimulation of intracellular H2O2 and fibroblast migration, yet these mutations have no effects on the expression of Duox2 and other NADPH-oxidases in cells. These findings illustrate for the first time that the extreme C-terminus of Duox2 is required for the functional activity of the enzyme. Furthermore, the conservative nature of the C-terminus suggests its role for activity in other NADPH-oxidases.
Collapse
Affiliation(s)
- Pyotr A. Tyurin-Kuzmin
- Department of Biochemistry and Molecular Medicine , Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University , Lomonosovsky ave, 27-1 , Moscow 119991 , Russia
| | - Maxim N. Karagyaur
- Department of Biochemistry and Molecular Medicine , Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University , Lomonosovsky ave, 27-1 , Moscow 119991 , Russia
| | - Yury P. Rubtsov
- Department of Biochemistry and Molecular Medicine , Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University , Lomonosovsky ave, 27-1 , Moscow 119991 , Russia
| | - Daniyar T. Dyikanov
- Department of Biochemistry and Molecular Medicine , Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University , Lomonosovsky ave, 27-1 , Moscow 119991 , Russia
| | - Pyotr A. Vasiliev
- Department of Biochemistry and Molecular Medicine , Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University , Lomonosovsky ave, 27-1 , Moscow 119991 , Russia
| | - Alexander V. Vorotnikov
- M.V. Lomonosov Moscow State University Medical Center , Lomonosovsky ave, 27-10 , Moscow 119991 , Russia
- Laboratory of Cell Motility , Institute of Experimental Cardiology, National Medical Research Center of Cardiology , Moscow 121552 , Russia
| |
Collapse
|
21
|
Chen X, Xu ML, Wang CN, Zhang LZ, Zhao YH, Zhu CL, Chen Y, Wu J, Yang YM, Wang XD. A partition-type tubular scaffold loaded with PDGF-releasing microspheres for spinal cord repair facilitates the directional migration and growth of cells. Neural Regen Res 2018; 13:1231-1240. [PMID: 30028332 PMCID: PMC6065242 DOI: 10.4103/1673-5374.235061] [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] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The best tissue-engineered spinal cord grafts not only match the structural characteristics of the spinal cord but also allow the seed cells to grow and function in situ. Platelet-derived growth factor (PDGF) has been shown to promote the migration of bone marrow stromal cells; however, cytokines need to be released at a steady rate to maintain a stable concentration in vivo. Therefore, new methods are needed to maintain an optimal concentration of cytokines over an extended period of time to effectively promote seed cell localization, proliferation and differentiation. In the present study, a partition-type tubular scaffold matching the anatomical features of the thoracic 8–10 spinal cord of the rat was fabricated using chitosan and then subsequently loaded with chitosan-encapsulated PDGF-BB microspheres (PDGF-MSs). The PDGF-MS-containing scaffold was then examined in vitro for sustained-release capacity, biocompatibility, and its effect on neural progenitor cells differentiated in vitro from multilineage-differentiating stress-enduring cells (MUSE-NPCs). We found that pre-freezing for 2 hours at −20°C significantly increased the yield of partition-type tubular scaffolds, and 30 μL of 25% glutaraldehyde ensured optimal crosslinking of PDGF-MSs. The resulting PDGF-MSs cumulatively released 52% of the PDGF-BB at 4 weeks in vitro without burst release. The PDGF-MS-containing tubular scaffold showed suitable biocompatibility towards MUSE-NPCs and could promote the directional migration and growth of these cells. These findings indicate that the combination of a partition-type tubular scaffold, PDGF-MSs and MUSE-NPCs may be a promising model for the fabrication of tissue-engineered spinal cord grafts.
Collapse
Affiliation(s)
- Xue Chen
- School of Biology & Basic Medical Sciences, Soochow University, Suzhou; Department of Histology and Embryology, Medical College, Nantong University, Nantong; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Mei-Ling Xu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Cheng-Niu Wang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Lu-Zhong Zhang
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Ya-Hong Zhao
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Chang-Lai Zhu
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Ying Chen
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Jian Wu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Yu-Min Yang
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xiao-Dong Wang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
22
|
Kizys MML, Louzada RA, Mitne-Neto M, Jara JR, Furuzawa GK, de Carvalho DP, Dias-da-Silva MR, Nesi-França S, Dupuy C, Maciel RMB. DUOX2 Mutations Are Associated With Congenital Hypothyroidism With Ectopic Thyroid Gland. J Clin Endocrinol Metab 2017; 102:4060-4071. [PMID: 28666341 DOI: 10.1210/jc.2017-00832] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/22/2017] [Indexed: 12/11/2022]
Abstract
CONTEXT Thyroid dysgenesis (TD) is the leading cause of congenital hypothyroidism (CH). The etiology of TD remains unknown in ∼90% of cases, the most common form being thyroid ectopia (TE) (48% to 61%). OBJECTIVE To search for candidate genes in hypothyroid children with TE. DESIGN, SETTING, AND PARTICIPANTS We followed a cohort of 268 children with TD and performed whole-exome sequencing (WES) in three children with CH with TE (CHTE) and compared them with 18 thyroid-healthy controls. We then screened an additional 41 children with CHTE by Sanger sequencing and correlated the WES and Sanger molecular findings with in vitro functional analysis. MAIN OUTCOME MEASURES Genotyping, mutation prediction analysis, and in vitro functional analysis. RESULTS We identified seven variants in the DUOX2 gene, namely G201E, L264CfsX57, P609S, M650T, E810X, M822V, and E1017G, and eight known variations. All children carrying DUOX2 variations had high thyroid-stimulating hormone levels at neonatal diagnosis. All mutations were localized in the N-terminal segment, and three of them led to effects on cell surface targeting and reactive oxygen species generation. The DUOX2 mutants also altered the interaction with the maturation factor DUOXA2 and the formation of a stable DUOX2/DUOXA2 complex at the cell surface, thereby impairing functional enzymatic activity. We observed no mutations in the classic genes related to TD or in the DUOX1 gene. CONCLUSION Our findings suggest that, in addition to thyroid hormonogenesis, the DUOX2 N-terminal domain may play a role in thyroid development.
Collapse
Affiliation(s)
- Marina M L Kizys
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| | - Ruy A Louzada
- UMR 8200 CNRS, Villejuif, 94800, France
- Institut Gustave Roussy, Villejuif, 94800, France
- Université Paris-Saclay, Orsay, 91405, France
- Laboratory of Endocrine Physiology Doris Rosenthal, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Miguel Mitne-Neto
- Fleury Group, São Paulo 04344-070, Brazil
- Human Genome and Stem Cell Research Center, Biosciences Institute, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Jessica R Jara
- Department of Pediatrics, Universidade Federal do Paraná, Curitiba 80060-240, Brazil
| | - Gilberto K Furuzawa
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| | - Denise P de Carvalho
- Laboratory of Endocrine Physiology Doris Rosenthal, Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Magnus R Dias-da-Silva
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| | - Suzana Nesi-França
- Department of Pediatrics, Universidade Federal do Paraná, Curitiba 80060-240, Brazil
| | - Corinne Dupuy
- UMR 8200 CNRS, Villejuif, 94800, France
- Institut Gustave Roussy, Villejuif, 94800, France
- Université Paris-Saclay, Orsay, 91405, France
| | - Rui M B Maciel
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
- Fleury Group, São Paulo 04344-070, Brazil
| |
Collapse
|
23
|
Zhang GP, Zhang J, Zhu CH, Lin L, Wang J, Zhang HJ, Li J, Yu XG, Zhao ZS, Dong W, Liu GB. MicroRNA-98 regulates osteogenic differentiation of human bone mesenchymal stromal cells by targeting BMP2. J Cell Mol Med 2016; 21:254-264. [PMID: 27860183 PMCID: PMC5264139 DOI: 10.1111/jcmm.12961] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/27/2016] [Indexed: 12/20/2022] Open
Abstract
To study the effects of microRNA‐98 (miR‐98) on human bone mesenchymal stromal cells (hBMSCs). The patients undergoing hip arthroplasty were selected by inclusion/exclusion criteria for this study. The extracted hBMSCs were detected of osteogenic differentiation by alizarin red S staining, and of cell phenotype by flow cytometry. Bioinformatics, dual luciferase report, western blotting, RT‐PCR and immunoblotting were used in our study. The hBMSCs were divided into miR‐98 mimics, miR‐98 negative control (NC), miR‐98 inhibitors, Mock and miR‐98 inhibitors + siBMP2 groups. Human bone mesenchymal stromal cells were extracted and purified in vitro and had specific cytological morphology, surface markers and abilities of self‐renewal and differentiation. Compared with the NC group and Mock group, the miR‐98 mimics group showed increased miR‐98 level while the miR‐98 inhibitors group decreased miR‐98 level (both P < 0.01). Dual luciferase reporter showed BMP2 was the target gene of miR‐98. The levels of mRNA and protein expression of BMP2, protein expression of RUNX2, alkaline phosphatase activity and osteocalcin content significantly decreased in the miR‐98 mimics group while increased in the miR‐98 inhibitors group and showed no changes in the NC group and Mock group (all P < 0.05). The miR‐98 mimics group showed obviously declined stained red particles and the miR‐98 inhibitors group showed opposite result. After lowering the expression of miR‐98, osteogenic differentiation ability of hBMSCs rose, which was weakened by the transfection with siBMP2. miR‐98 may regulate osteogenic differentiation of hBMSCs by targeting BMP2.
Collapse
Affiliation(s)
- Guo-Ping Zhang
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Zhang
- Medical Physics Department of Basic Medical College of Hebei Medical University, Shijiazhuang, China
| | - Chao-Hua Zhu
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Lin
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jian Wang
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hai-Jing Zhang
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jun Li
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiao-Guang Yu
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhen-Shuan Zhao
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wei Dong
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guo-Bin Liu
- Department of Orthopedics, The First Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|