1
|
Villagrán-Andrade KM, Núñez-Carro C, Blanco FJ, de Andrés MC. Nutritional Epigenomics: Bioactive Dietary Compounds in the Epigenetic Regulation of Osteoarthritis. Pharmaceuticals (Basel) 2024; 17:1148. [PMID: 39338311 PMCID: PMC11434976 DOI: 10.3390/ph17091148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Nutritional epigenomics is exceptionally important because it describes the complex interactions among food compounds and epigenome modifications. Phytonutrients or bioactive compounds, which are secondary metabolites of plants, can protect against osteoarthritis by suppressing the expression of inflammatory and catabolic mediators, modulating epigenetic changes in DNA methylation, and the histone or chromatin remodelling of key inflammatory genes and noncoding RNAs. The combination of natural epigenetic modulators is crucial because of their additive and synergistic effects, safety and therapeutic efficacy, and lower adverse effects than conventional pharmacology in the treatment of osteoarthritis. In this review, we have summarized the chondroprotective properties of bioactive compounds used for the management, treatment, or prevention of osteoarthritis in both human and animal studies. However, further research is needed into bioactive compounds used as epigenetic modulators in osteoarthritis, in order to determine their potential value for future clinical applications in osteoarthritic patients as well as their relation with the genomic and nutritional environment, in order to personalize food and nutrition together with disease prevention.
Collapse
Affiliation(s)
- Karla Mariuxi Villagrán-Andrade
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Carmen Núñez-Carro
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Francisco J Blanco
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
- Grupo de Investigación en Reumatología y Salud, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Campus de Oza, Universidade da Coruña (UDC), 15008 A Coruña, Spain
| | - María C de Andrés
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| |
Collapse
|
2
|
Louka P, Orriss IR, Pitsillides AA. Stable Sulforaphane Targets the Early Stages of Osteoclast Formation to Engender a Lasting Functional Blockade of Osteoclastogenesis. Cells 2024; 13:165. [PMID: 38247857 PMCID: PMC10814088 DOI: 10.3390/cells13020165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Sulforaphane, the native but unstable form of SFX-01, is an antioxidant that activates the NRF2 and inhibits the NF-KB pathways to achieve its actions. Resolving the mechanism(s) by which SFX-01 serves to control the various osteoclastogenic stages may expose pathways that could be explored for therapeutic use. Here we seek to identify the stage of osteoclastogenesis targeted by SFX-01 and explore whether, like SFN, it exerts its actions via the NRF2 and NF-KB pathways. Osteoclasts generated from the bone marrow (BM) of mice were cultured with SFX-01 at different timepoints to examine each phase of osteoclastogenesis separately. This showed that SFX-01 exerted actions throughout the process of osteoclastogenesis, but had its largest effects in the early osteoclast precursor differentiation stage. Thus, treatment with SFX-01 for the duration of culture, for the initial 3 days differentiation or for as little as the first 24 h was sufficient for effective inhibition. This aligned with data suggesting that SFX-01 reduced DC-STAMP levels, osteoclast nuclear number and modified cytoskeletal architecture. Pharmacological regulation of the NRF2 pathways, via selective inhibitors/activators, supported the anti-osteoclastogenic roles of an SFX-01-mediated by NRF2 activation, as well as the need for tight NF-KB pathway regulation in osteoclast formation/function.
Collapse
Affiliation(s)
| | | | - Andrew A. Pitsillides
- Skeletal Biology Group, Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK; (P.L.); (I.R.O.)
| |
Collapse
|
3
|
Wu Z, Yang Z, Liu L, Xiao Y. Natural compounds protect against the pathogenesis of osteoarthritis by mediating the NRF2/ARE signaling. Front Pharmacol 2023; 14:1188215. [PMID: 37324450 PMCID: PMC10266108 DOI: 10.3389/fphar.2023.1188215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023] Open
Abstract
Osteoarthritis (OA), a chronic joint cartilage disease, is characterized by the imbalanced homeostasis between anabolism and catabolism. Oxidative stress contributes to inflammatory responses, extracellular matrix (ECM) degradation, and chondrocyte apoptosis and promotes the pathogenesis of OA. Nuclear factor erythroid 2-related factor 2 (NRF2) is a central regulator of intracellular redox homeostasis. Activation of the NRF2/ARE signaling may effectively suppress oxidative stress, attenuate ECM degradation, and inhibit chondrocyte apoptosis. Increasing evidence suggests that the NRF2/ARE signaling has become a potential target for the therapeutic management of OA. Natural compounds, such as polyphenols and terpenoids, have been explored to protect against OA cartilage degeneration by activating the NRF2/ARE pathway. Specifically, flavonoids may function as NRF2 activators and exhibit chondroprotective activity. In conclusion, natural compounds provide rich resources to explore the therapeutic management of OA by activating NRF2/ARE signaling.
Collapse
Affiliation(s)
- Zhenyu Wu
- First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- First Clinical Medical College of Gannan Medical University, Ganzhou, China
| | - Zhouxin Yang
- First Clinical Medical College of Gannan Medical University, Ganzhou, China
| | - Luying Liu
- First Clinical Medical College of Gannan Medical University, Ganzhou, China
| | - Yong Xiao
- Jiangxi University of Traditional Chinese Medicine, Nanchang, China
- Xiaoyong Traditional Chinese Medicine Clinic in Yudu, Ganzhou, China
| |
Collapse
|
4
|
Royce SG, Licciardi PV, Beh RC, Bourke JE, Donovan C, Hung A, Khurana I, Liang JJ, Maxwell S, Mazarakis N, Pitsillou E, Siow YY, Snibson KJ, Tobin MJ, Ververis K, Vongsvivut J, Ziemann M, Samuel CS, Tang MLK, El-Osta A, Karagiannis TC. Sulforaphane prevents and reverses allergic airways disease in mice via anti-inflammatory, antioxidant, and epigenetic mechanisms. Cell Mol Life Sci 2022; 79:579. [PMID: 36319916 PMCID: PMC11803010 DOI: 10.1007/s00018-022-04609-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 11/30/2022]
Abstract
Sulforaphane has been investigated in human pathologies and preclinical models of airway diseases. To provide further mechanistic insights, we explored L-sulforaphane (LSF) in the ovalbumin (OVA)-induced chronic allergic airways murine model, with key hallmarks of asthma. Histological analysis indicated that LSF prevented or reversed OVA-induced epithelial thickening, collagen deposition, goblet cell metaplasia, and inflammation. Well-known antioxidant and anti-inflammatory mechanisms contribute to the beneficial effects of LSF. Fourier transform infrared microspectroscopy revealed altered composition of macromolecules, following OVA sensitization, which were restored by LSF. RNA sequencing in human peripheral blood mononuclear cells highlighted the anti-inflammatory signature of LSF. Findings indicated that LSF may alter gene expression via an epigenetic mechanism which involves regulation of protein acetylation status. LSF resulted in histone and α-tubulin hyperacetylation in vivo, and cellular and enzymatic assays indicated decreased expression and modest histone deacetylase (HDAC) inhibition activity, in comparison with the well-known pan-HDAC inhibitor suberoylanilide hydroxamic acid (SAHA). Molecular modeling confirmed interaction of LSF and LSF metabolites with the catalytic domain of metal-dependent HDAC enzymes. More generally, this study confirmed known mechanisms and identified potential epigenetic pathways accounting for the protective effects and provide support for the potential clinical utility of LSF in allergic airways disease.
Collapse
Affiliation(s)
- Simon G Royce
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, 3052, Australia
| | - Paul V Licciardi
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Raymond C Beh
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jane E Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, 2305, Australia
- Centre for Inflammation, Centenary Institute, Camperdown, NSW, 2050, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Andrew Hung
- School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | - Ishant Khurana
- Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Julia J Liang
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | - Scott Maxwell
- Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Nadia Mazarakis
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Eleni Pitsillou
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | - Ya Yun Siow
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Kenneth J Snibson
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Mark J Tobin
- ANSTO-Australian Synchrotron, Clayton, VIC, 3168, Australia
| | - Katherine Ververis
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia
| | | | - Mark Ziemann
- Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Warrnambool, VIC, 3216, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Mimi L K Tang
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Population Allergy Group, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Allergy and Immunology, Royal Children's Hospital, Parkville, VIC, 3052, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Tom C Karagiannis
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
5
|
Louka P, Orriss IR, Pitsillides AA. High bone mass in mice can be linked to lower osteoclast formation, resorptive capacity, and restricted in vitro sensitivity to inhibition by stable sulforaphane. Cell Biochem Funct 2022; 40:683-693. [PMID: 35924674 DOI: 10.1002/cbf.3734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/05/2022] [Accepted: 07/22/2022] [Indexed: 01/07/2023]
Abstract
Mouse strains can have divergent basal bone mass, yet this phenotype is seldom reflected in the design of studies seeking to identify new modulators of bone resorption by osteoclasts. Sulforaphane exerts inhibitory effects on in vitro osteoclastogenesis in cells from C57BL/6 mice. Here, we explore whether a divergent basal bone mass in different mouse strains is linked both to in vitro osteoclastogenic potential and to SFX-01 sensitivity. Accordingly, osteoclasts isolated from the bone marrow (BM) of C57BL/6, STR/Ort and CBA mice with low, high, and intermediate bone mass, respectively, were cultured under conditions to promote osteoclast differentiation and resorption; they were also treated with chemically stabilised sulforaphane (SFX-01) and respective sensitivity to inhibition evaluated by counting osteoclast number/resorption activity on dentine discs. We observed that osteoclastogenesis exhibited different macrophage colony-stimulating factor/receptor activator of nuclear factor kappa-Β ligand sensitivity in these mouse strains, with cells from C57BL/6 and CBA generating higher osteoclast numbers than STR/Ort; the latter formed only half as many mature osteoclasts. We found that 100 nM SFX-01 exerted a potent and significant reduction in osteoclast number and resorptive activity in cells derived from C57BL/6 mice. In contrast, 10-fold higher SFX-01 concentrations were required for similar inhibition in CBA-derived cells and, strikingly, a further 2.5-fold greater concentration was required for significant restriction of osteoclast formation/function in STR/Ort. These data are consistent with the notion that the BM osteoclast precursor population contributes to the relative differences in mouse bone mass and that mice with higher bone mass exhibit lower in vitro osteoclastogenic potential as well as reduced sensitivity to inhibition by SFX-01.
Collapse
Affiliation(s)
- Polymnia Louka
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Isabel R Orriss
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Andrew A Pitsillides
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| |
Collapse
|
6
|
Gambari L, Grigolo B, Grassi F. Dietary organosulfur compounds: Emerging players in the regulation of bone homeostasis by plant-derived molecules. Front Endocrinol (Lausanne) 2022; 13:937956. [PMID: 36187121 PMCID: PMC9521401 DOI: 10.3389/fendo.2022.937956] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
The progressive decline of bone mass and the deterioration of bone microarchitecture are hallmarks of the bone aging. The resulting increase in bone fragility is the leading cause of bone fractures, a major cause of disability. As the frontline pharmacological treatments for osteoporosis suffer from low patients' adherence and occasional side effects, the importance of diet regimens for the prevention of excessive bone fragility has been increasingly recognized. Indeed, certain diet components have been already associated to a reduced fracture risk. Organosulfur compounds are a broad class of molecules containing sulfur. Among them, several molecules of potential therapeutic interest are found in edible plants belonging to the Allium and Brassica botanical genera. Polysulfides derived from Alliaceae and isothiocyanates derived from Brassicaceae hold remarkable nutraceutical potential as anti-inflammatory, antioxidants, vasorelaxant and hypolipemic. Some of these effects are linked to the ability to release the gasotrasmitter hydrogen sulfide (H2S). Recent preclinical studies have investigated the effect of organosulfur compounds in bone wasting and metabolic bone diseases, revealing a strong potential to preserve skeletal health by exerting cytoprotection and stimulating the bone forming activity by osteoblasts and attenuating bone resorption by osteoclasts. This review is intended for revising evidence from preclinical and epidemiological studies on the skeletal effects of organosulfur molecules of dietary origin, with emphasis on the direct regulation of bone cells by plant-derived polysulfides, glucosinolates and isothiocyanates. Moreover, we highlight the potential molecular mechanisms underlying the biological role of these compounds and revise the importance of the so-called 'H2S-system' on the regulation of bone homeostasis.
Collapse
|
7
|
Connolly EL, Sim M, Travica N, Marx W, Beasy G, Lynch GS, Bondonno CP, Lewis JR, Hodgson JM, Blekkenhorst LC. Glucosinolates From Cruciferous Vegetables and Their Potential Role in Chronic Disease: Investigating the Preclinical and Clinical Evidence. Front Pharmacol 2021; 12:767975. [PMID: 34764875 PMCID: PMC8575925 DOI: 10.3389/fphar.2021.767975] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/11/2021] [Indexed: 01/04/2023] Open
Abstract
An increasing body of evidence highlights the strong potential for a diet rich in fruit and vegetables to delay, and often prevent, the onset of chronic diseases, including cardiometabolic, neurological, and musculoskeletal conditions, and certain cancers. A possible protective component, glucosinolates, which are phytochemicals found almost exclusively in cruciferous vegetables, have been identified from preclinical and clinical studies. Current research suggests that glucosinolates (and isothiocyanates) act via several mechanisms, ultimately exhibiting anti-inflammatory, antioxidant, and chemo-protective effects. This review summarizes the current knowledge surrounding cruciferous vegetables and their glucosinolates in relation to the specified health conditions. Although there is evidence that consumption of a high glucosinolate diet is linked with reduced incidence of chronic diseases, future large-scale placebo-controlled human trials including standardized glucosinolate supplements are needed.
Collapse
Affiliation(s)
- Emma L Connolly
- Institute for Nutrition Research, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Marc Sim
- Institute for Nutrition Research, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia.,Medical School, Royal Perth Hospital Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Nikolaj Travica
- IMPACT-The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, VIC, Australia
| | - Wolfgang Marx
- IMPACT-The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, VIC, Australia
| | - Gemma Beasy
- Quadram Institute Bioscience, Norwich, United Kingdom
| | - Gordon S Lynch
- Department of Anatomy and Physiology, Centre for Muscle Research, School of Biomedical Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Catherine P Bondonno
- Institute for Nutrition Research, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia.,Medical School, Royal Perth Hospital Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Joshua R Lewis
- Institute for Nutrition Research, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia.,Medical School, Royal Perth Hospital Research Foundation, The University of Western Australia, Perth, WA, Australia.,Centre for Kidney Research, Children's Hospital at Westmead, School of Public Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Jonathan M Hodgson
- Institute for Nutrition Research, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia.,Medical School, Royal Perth Hospital Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Lauren C Blekkenhorst
- Institute for Nutrition Research, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia.,Medical School, Royal Perth Hospital Research Foundation, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
8
|
Sikorska-Zimny K, Beneduce L. The Metabolism of Glucosinolates by Gut Microbiota. Nutrients 2021; 13:2750. [PMID: 34444909 PMCID: PMC8401010 DOI: 10.3390/nu13082750] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/30/2021] [Accepted: 08/08/2021] [Indexed: 02/08/2023] Open
Abstract
Glucosinolates (GLS) and their derivatives are secondary plant metabolites abundant in Brassicaceae. Due to the enzymatic reaction between GLS and myrosinase enzyme, characteristic compounds with a pungent taste are formed, used by plants to defend themselves against insect herbivores. These GLS derivatives have an important impact on human health, including anti-inflammation and anti-cancer effects. However, GLS derivatives' formation needs previous enzymatic reactions catalyzed by myrosinase enzyme. Many of the brassica-based foods are processed at a high temperature that inactivates enzymes, hindering its bioavailability. In the last decade, several studies showed that the human gut microbiome can provide myrosinase activity that potentially can raise the beneficial effects of consumption of vegetables rich in GLS. The variability of the human gut microbiome (HGM) in human populations and the diverse intake of GLS through the diet may lead to greater variability of the real dose of pro-healthy compounds absorbed by the human body. The exploitation of the genetic and biochemical potential of HGM and correct ecological studies of both isolated strains and mixed population are of great interest. This review focuses on the most recent advances in this field.
Collapse
Affiliation(s)
- Kalina Sikorska-Zimny
- Fruit and Vegetables Storage and Processing Department, Division of Fruit and Vegetable Storage and Postharvest Physiology, The National Institute of Horticultural Research, Pomologiczna 13a Street, 96-100 Skierniewice, Poland
- Medical, Natural and Technical College, Institute of Health Sciences, Stefan Batory State University, Batorego 64c Street, 96-100 Skierniewice, Poland
| | - Luciano Beneduce
- Department of the Sciences of Agriculture, Food, Natural Resources, and Engineering (DAFNE) the University of Foggia, Via Napoli 25, 71122 Foggia, Italy;
| |
Collapse
|
9
|
Chang YM, Menges S, Westhof A, Kleinschmidt-Doerr K, Brenneis C, Pitsillides AA. Systematic analysis reveals that colony housing aligns gait profiles and strengthens link between histological and micro-CT bone markers in rat models of osteoarthritis. FASEB J 2021; 35:e21451. [PMID: 33683776 DOI: 10.1096/fj.202002009r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/01/2021] [Accepted: 02/01/2021] [Indexed: 11/11/2022]
Abstract
Osteoarthritis (OA) etiopathogenesis is complex with strong environmental/lifestyle determinants that, in laboratory animals, extend to social context and stress levels. This study seeks to identify whether colony housing of rats exerts a social impact on locomotion behaviors to influence alignment between symptomatic (gait) and structural (bone micro-CT measures, cartilage morphometry, and histology) OA outcome measures. Rats were randomly allocated to conventional (type IV; n = 48) or rat colony cage (RCC; n = 30) housing, further randomized to OA surgical models (ACLT + tMx, MMT or DMM) or no surgery (control), and maintained for 19 weeks during which multiple gait recordings were made. Standard histological grading and bone micro-CT data were collected at necropsy. Principal component analysis was used to summarize the variation in gait, micro-CT or histology. Linear mixed effects model or two-way ANOVA was employed to evaluate the impact of the housing system, surgery and time on gait, or micro-CT and histology components Analyses reveal that RCC exaggerates trends in gait change via a combined effect of the housing system and surgery. Intriguingly, RCC-housed nonoperated control rats showed similar gait changes to rats subjected to surgery; the latter exhibited significant structural joint changes in both systems. Stronger correlation between histological and micro-CT bone changes were found in medial and lateral tibia joint compartments of rats housed in RCC system. This study has established that rat social housing exaggerates outcomes in traditional histological measures of OA, generates stronger links between histology and micro-CT bone changes and removes gait differences as a variable in their etiology.
Collapse
Affiliation(s)
- Yu-Mei Chang
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | | | | | | | | | - Andrew A Pitsillides
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
10
|
Moon SJ, Jhun J, Ryu J, Kwon JY, Kim SY, Jung K, Cho ML, Min JK. The anti-arthritis effect of sulforaphane, an activator of Nrf2, is associated with inhibition of both B cell differentiation and the production of inflammatory cytokines. PLoS One 2021; 16:e0245986. [PMID: 33592002 PMCID: PMC7886167 DOI: 10.1371/journal.pone.0245986] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is an important transcription factor that plays a pivotal role in cellular defense against oxidative injury. Nrf2 signaling is involved in attenuating autoimmune disorders such as rheumatoid arthritis (RA). B cells play several roles in the pathogenesis of RA, such as in autoantibody production, antigen presentation, and T-cell activation. We investigated the anti-arthritic mechanisms of sulforaphane, an activator of Nrf2, in terms of its effect on B cells. To investigate the effect of sulforaphane on collagen-induced arthritis (CIA), sulforaphane was administered intraperitoneally after CIA induction. Hematoxylin and eosin-stained sections were scored for inflammation, pannus invasion, and bone and cartilage damage. We assessed the expression levels of inflammation-related factors by real-time PCR and the levels of various IgG subclasses by enzyme-linked immunosorbent assay. Sulforaphane treatment reduced the arthritis score and the severity of histologic inflammation in CIA mice. The joints from sulforaphane-treated CIA mice showed decreased expression of interleukin (IL)-6, IL-17, tumor necrosis factor (TNF)-α, receptor activator of NF-κB ligand, and tartrate-resistant acid phosphatase. Sulforaphane-treated mice showed lower circulating levels of type-II-collagen-specific IgG, IgG1, and IgG2a. In vitro, sulforaphane treatment significantly reduced the differentiation of lipopolysaccharide-stimulated murine splenocytes into plasma B cells and germinal-center B cells. Finally, sulforaphane significantly inhibited the production of IL-6, TNF-α, and IL-17 by human peripheral blood mononuclear cells stimulated with an anti-CD3 monoclonal antibody in a dose-dependent manner. Inhibition of differentiation into plasma B and Germinal Center B cells may be the mechanism underlying the anti-arthritic effect of sulforaphane.
Collapse
Affiliation(s)
- Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, South Korea
| | - Jooyeon Jhun
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Jaeyoon Ryu
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Ji ye Kwon
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Se-Young Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | | | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
- Impact Biotech, Seoul, South Korea
- Laboratory of Immune Network, Conversant Research Consortium in Immunologic Disease, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- * E-mail: (JKM); (MLC)
| | - Jun-Ki Min
- Department of Internal Medicine, and the Clinical Medicine Research Institute of Bucheon St. Mary’s Hospital, Bucheon-si, South Korea
- * E-mail: (JKM); (MLC)
| |
Collapse
|
11
|
Schulze-Tanzil G. Experimental Therapeutics for the Treatment of Osteoarthritis. J Exp Pharmacol 2021; 13:101-125. [PMID: 33603501 PMCID: PMC7887204 DOI: 10.2147/jep.s237479] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) therapy remains a large challenge since no causative treatment options are so far available. Despite some main pathways contributing to OA are identified its pathogenesis is still rudimentary understood. A plethora of therapeutically promising agents are currently tested in experimental OA research to find an opportunity to reverse OA-associated joint damage and prevent its progression. Hence, this review aims to summarize novelly emerging experimental approaches for OA. Due to the diversity of strategies shown only main aspects could be summarized here including herbal medicines, nanoparticular compounds, growth factors, hormones, antibody-, cell- and extracellular vesicle (EV)-based approaches, optimized tools for joint viscosupplementation, genetic regulators such as si- or miRNAs and promising combinations. An abundant multitude of compounds obtained from plants, environmental, autologous or synthetic sources have been identified with anabolic, anti-inflammatory, -catabolic and anti-apoptotic properties. Some ubiquitous signaling pathways such as wingless and Integration site-1 (Wnt), Sirtuin, Toll-like receptor (TLR), mammalian target of rapamycin (mTOR), Nuclear Factor (NF)-κB and complement are involved in OA and addressed by them. Hyaluronan (HA) provided benefit in OA since many decades, and novel HA formulations have been developed now with higher HA content and long-term stability achieved by cross-linking suitable to be combined with other agents such as components from herbals or chemokines to attract regenerative cells. pH- or inflammation-sensitive nanoparticular compounds could serve as versatile slow-release systems of active compounds, for example, miRNAs. Some light has been brought into the intimate regulatory network of small RNAs in the pathogenesis of OA which might be a novel avenue for OA therapy in future. Attraction of autologous regenerative cells by chemokines and exosome-based treatment strategies could also innovate OA therapy.
Collapse
Affiliation(s)
- Gundula Schulze-Tanzil
- Department of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg, Bavaria, Germany
| |
Collapse
|
12
|
Luo T, Fu X, Liu Y, Ji Y, Shang Z. Sulforaphane Inhibits Osteoclastogenesis via Suppression of the Autophagic Pathway. Molecules 2021; 26:molecules26020347. [PMID: 33445451 PMCID: PMC7830922 DOI: 10.3390/molecules26020347] [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: 12/11/2020] [Revised: 01/01/2021] [Accepted: 01/03/2021] [Indexed: 02/06/2023] Open
Abstract
Previous studies have demonstrated that sulforaphane (SFN) is a promising agent against osteoclastic bone destruction. However, the mechanism underlying its anti-osteoclastogenic activity is still unclear. Herein, for the first time, we explored the potential role of autophagy in SFN-mediated anti-osteoclastogenesis in vitro and in vivo. We established an osteoclastogenesis model using receptor activator of nuclear factor kappa-β ligand (RANKL)-induced RAW264.7 cells and bone marrow macrophages (BMMs). Tartrate-resistant acid phosphatase (TRAP) staining showed the formation of osteoclasts. We observed autophagosomes by transmission electron microscopy (TEM). In vitro, we found that SFN inhibited osteoclastogenesis (number of osteoclasts: 22.67 ± 0.88 in the SFN (0) group vs. 20.33 ± 1.45 in the SFN (1 μM) group vs. 13.00 ± 1.00 in the SFN (2.5 μM) group vs. 6.66 ± 1.20 in the SFN (2.5 μM) group), decreased the number of autophagosomes, and suppressed the accumulation of several autophagic proteins in osteoclast precursors. The activation of autophagy by rapamycin (RAP) almost reversed the SFN-elicited anti-osteoclastogenesis (number of osteoclasts: 22.67 ± 0.88 in the control group vs. 13.00 ± 1.00 in the SFN group vs. 17.33 ± 0.33 in the SFN+RAP group). Furthermore, Western blot (WB) analysis revealed that SFN inhibited the phosphorylation of c-Jun N-terminal kinase (JNK). The JNK activator anisomycin significantly promoted autophagy, whereas the inhibitor SP600125 markedly suppressed autophagic activation in pre-osteoclasts. Microcomputed tomography (CT), immunohistochemistry (IHC), and immunofluorescence (IF) were used to analyze the results in vivo. Consistent with the in vitro results, we found that the administration of SFN could decrease the number of osteoclasts and the expression of autophagic light chain 3 (LC3) and protect against lipopolysaccharide (LPS)-induced calvarial erosion. Our findings highlight autophagy as a crucial mechanism of SFN-mediated anti-osteoclastogenesis and show that the JNK signaling pathway participates in this process.
Collapse
Affiliation(s)
- Tingting Luo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430000, China; (T.L.); (X.F.); (Y.L.)
| | - Xiazhou Fu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430000, China; (T.L.); (X.F.); (Y.L.)
| | - Yaoli Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430000, China; (T.L.); (X.F.); (Y.L.)
| | - Yaoting Ji
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430000, China; (T.L.); (X.F.); (Y.L.)
- Correspondence: (Y.J.); (Z.S.); Tel.: +86-138-8607-0344 (Y.J.); +86-27-8768-6129 (Z.S.)
| | - Zhengjun Shang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430000, China; (T.L.); (X.F.); (Y.L.)
- Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan 430000, China
- Correspondence: (Y.J.); (Z.S.); Tel.: +86-138-8607-0344 (Y.J.); +86-27-8768-6129 (Z.S.)
| |
Collapse
|
13
|
Jacobs BY, Allen KD. Factors affecting the reliability of behavioral assessments for rodent osteoarthritis models. Lab Anim 2020; 54:317-329. [PMID: 31431137 PMCID: PMC7830740 DOI: 10.1177/0023677219867715] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The translational value of osteoarthritis (OA) models is often debated because numerous studies have shown that animal models frequently fail to predict the efficacy of therapies in humans. In part, this failing may be due to the paucity of preclinical studies that include behavioral assessments in their metrics. Behavioral assessments of animal OA models can provide valuable data on the pain and disability associated with disease-sequelae of significant clinical relevance. Clinical definitions of efficacy for OA therapeutics often center on their palliative effects. Thus, the widespread inclusion of behaviors indicative of pain and disability in preclinical animal studies may contribute to greater success identifying clinically relevant interventions. Unfortunately, studies that include behavioral assays still frequently encounter pitfalls in assay selection, protocol consistency, and data/methods transparency. Targeted selection of behavioral assays, with consideration of the array of clinical OA phenotypes and the limitations of individual behavioral assays, is necessary to identify clinically relevant outcomes in OA animal models appropriately. Furthermore, to facilitate accurate comparisons across research groups and studies, it is necessary to improve the transparency of methods. Finally, establishing agreed-upon and clear definitions of behavioral data will reduce the convolution of data both within and between studies. Improvement in these areas is critical to the continued benefit of preclinical animal studies as translationally relevant data in OA research. As such, this review highlights the current state of behavioral analyses in preclinical OA models.
Collapse
Affiliation(s)
- Brittany Y Jacobs
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Kyle D Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|
14
|
Abstract
Osteoarthritis (OA) is one of the most debilitating diseases and is associated with a high personal and socioeconomic burden. So far, there is no therapy available that effectively arrests structural deterioration of cartilage and bone or is able to successfully reverse any of the existing structural defects. Efforts to identify more tailored treatment options led to the development of strategies that enabled the classification of patient subgroups from the pool of heterogeneous phenotypes that display distinct common characteristics. To this end, the classification differentiates the structural endotypes into cartilage and bone subtypes, which are predominantly driven by structure-related degenerative events. In addition, further classifications have highlighted individuals with an increased inflammatory contribution (inflammatory phenotype) and pain-driven phenotypes as well as senescence and metabolic syndrome phenotypes. Most probably, it will not be possible to classify individuals by a single definite subtype, but it might help to identify groups of patients with a predominant pathology that would more likely benefit from a specific drug or cell-based therapy. Current clinical trials addressed mainly regeneration/repair of cartilage and bone defects or targeted pro-inflammatory mediators by intra-articular injections of drugs and antibodies. Pain was treated mostly by antagonizing nerve growth factor (NGF) activity and its receptor tropomyosin-related kinase A (TrkA). Therapies targeting metabolic disorders such as diabetes mellitus and senescence/aging-related pathologies are not specifically addressing OA. However, none of these therapies has been proven to modify disease progression significantly or successfully prevent final joint replacement in the advanced disease stage. Within this review, we discuss the recent advances in phenotype-specific treatment options and evaluate their applicability for use in personalized OA therapy.
Collapse
Affiliation(s)
- Susanne Grässel
- Department of Orthopedic Surgery, Exp. Orthopedics, ZMB/Biopark 1, Am Biopark 9, University of Regensburg, Regensburg, 93053, Germany
| | - Dominique Muschter
- Department of Orthopedic Surgery, Exp. Orthopedics, ZMB/Biopark 1, Am Biopark 9, University of Regensburg, Regensburg, 93053, Germany
| |
Collapse
|
15
|
Xu X, Wang D, Zheng C, Gao B, Fan J, Cheng P, Liu B, Yang L, Luo Z. Progerin accumulation in nucleus pulposus cells impairs mitochondrial function and induces intervertebral disc degeneration and therapeutic effects of sulforaphane. Theranostics 2019; 9:2252-2267. [PMID: 31149042 PMCID: PMC6531300 DOI: 10.7150/thno.30658] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 02/10/2019] [Indexed: 01/18/2023] Open
Abstract
Progerin, a truncated unprocessed lamin A protein, causes tissue aging and degeneration. In this study we explored the role of progerin in the pathogenesis of intervertebral disc degeneration (IDD). We also examined the effect of sulforaphane (SFN) on progerin accumulation and mitochondrial dysfunction in IDD. Methods: The role of progerin in IDD was explored using human nucleus pulposus (NP) tissues, rat NP cells, and Lmna G609G knock-in mice. Immunostaining, X-ray imaging, and Western blotting were performed to assess the phenotypes of intervertebral discs. Alterations in senescence and apoptosis were evaluated by SA-β-galactosidase, immunofluorescence, flow cytometry, and TUNEL assays. Mitochondrial function was investigated by JC-1 staining, transmission electron microscopy, and determination of the level of ATP and the activities of mitochondrial enzymes. Results: The progerin level was elevated in degenerated human NP tissues. Lmna G609G/G609G mice displayed IDD, as evidenced by increased matrix metalloproteinase-13 expression and decreased collagen II and aggrecan expression and disc height. Furthermore, progerin overexpression in rat NP cells induced mitochondrial dysfunction (decreased ATP synthesis, mitochondrial membrane potential, and activities of mitochondrial complex enzymes), morphologic abnormalities, and disrupted mitochondrial dynamic (abnormal expression of proteins involved in fission and fusion), resulting in apoptosis and senescence. SFN ameliorated the progerin-induced aging defects and mitochondrial dysfunction in NP cells and IDD in Lmna G609G/G609G mice. Conclusions: Progerin is involved in the pathogenesis of IDD. Also, SFN alleviates progerin‑induced IDD, which is associated with amelioration of aging defects and mitochondrial dysfunction. Thus, SFN shows promise for the treatment of IDD.
Collapse
|
16
|
Du C, Smith A, Avalos M, South S, Crabtree K, Wang W, Kwon YH, Vijayagopal P, Juma S. Blueberries Improve Pain, Gait Performance, and Inflammation in Individuals with Symptomatic Knee Osteoarthritis. Nutrients 2019; 11:nu11020290. [PMID: 30699971 PMCID: PMC6413191 DOI: 10.3390/nu11020290] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/17/2019] [Accepted: 01/23/2019] [Indexed: 01/11/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disorder in the world and is the most frequent cause of walking related disability among older adults in the US, which brings a significant economic burden and reduces quality of life. The initiation and development of OA typically involves degeneration or progressive loss of the structure and function of articular cartilage. Inflammation is one of the major drives of the progression of OA. Dietary polyphenols have been studied for their anti-inflammatory properties and potential anabolic effects on the cartilage cells. Blueberries are widely consumed and are high in dietary polyphenols, therefore regular consumption of blueberries may help improve OA. The purpose of the present study was to examine the effect of freeze dried whole blueberries on pain, gait performance, and inflammation in individuals with symptomatic knee OA. In a randomized, double-blind trial, adults age 45 to 79 with symptomatic knee OA, were randomized to either consume 40 g freeze-dried blueberry powder (n = 33) or placebo powder (n = 30) daily for four months. Blood draws and assessment of pain and gait were conducted at baseline, two months, and four months. Western Ontario McMaster Osteoarthritis Index (WOMAC) questionnaires were used to assess pain and GAITRite® electronic walkway was used to evaluate gait spatiotemporal parameters. WOMAC total score and sub-groups, including pain, stiffness, and difficulty to perform daily activities decreased significantly in the blueberry treatment group (p < 0.05), but improvement of WOMAC total score and difficulty to perform daily activities were not observed in the placebo group. Normal walking pace single support percentage for both limbs increased (p = or < 0.007), while double support percentage for both limbs decreased in the blueberry treatment group (p = or < 0.003). No significant changes were observed in plasma concentrations of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-10, IL-13, matrix metalloproteinases (MMP)-3, MMP-13, and monocyte chemoattractant protein-1 (MCP-1) in both treatment groups. However, an increasing trend for IL-13 concentration and a decreasing trend in MCP-1 concentration were noted in the blueberry group. The findings of this study suggest that daily incorporation of whole blueberries may reduce pain, stiffness, and difficulty to perform daily activities, while improving gait performance, and would therefore improve quality of life in individuals with symptomatic knee OA.
Collapse
Affiliation(s)
- Chen Du
- Department of Nutrition and Food Science, Texas Woman's University, Denton, TX 76204, USA.
| | - Amy Smith
- Department of Nutrition and Food Science, Texas Woman's University, Denton, TX 76204, USA.
| | - Marco Avalos
- School of Health Promotion and Kinesiology, Texas Woman's University, Denton, TX 76204, USA.
| | - Sanique South
- Department of Nutrition and Food Science, Texas Woman's University, Denton, TX 76204, USA.
| | - Keith Crabtree
- Department of Nutrition and Food Science, Texas Woman's University, Denton, TX 76204, USA.
| | - Wanyi Wang
- Center for Research Design and Analysis, Texas Woman's University, Houston, TX 77030, USA.
| | - Young-Hoo Kwon
- School of Health Promotion and Kinesiology, Texas Woman's University, Denton, TX 76204, USA.
| | - Parakat Vijayagopal
- Department of Nutrition and Food Science, Texas Woman's University, Denton, TX 76204, USA.
| | - Shanil Juma
- Department of Nutrition and Food Science, Texas Woman's University, Denton, TX 76204, USA.
| |
Collapse
|
17
|
Identifying chondroprotective diet-derived bioactives and investigating their synergism. Sci Rep 2018; 8:17173. [PMID: 30464238 PMCID: PMC6249298 DOI: 10.1038/s41598-018-35455-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/06/2018] [Indexed: 01/18/2023] Open
Abstract
Osteoarthritis (OA) is a multifactorial disease and nutrition is a modifiable factor that may contribute to disease onset or progression. A detailed understanding of mechanisms through which diet-derived bioactive molecules function and interact in OA is needed. We profiled 96 diet-derived, mainly plant-based bioactives using an in vitro model in chondrocytes, selecting four candidates for further study. We aimed to determine synergistic interactions between bioactives that affected the expression of key genes in OA. Selected bioactives, sulforaphane, apigenin, isoliquiritigenin and luteolin, inhibited one or more interleukin-1-induced metalloproteinases implicated in OA (MMP1, MMP13, ADAMTS4, ADAMTS5). Isoliquiritigenin and luteolin showed reactive oxygen species scavenging activity in chondrocytes whereas sulforaphane had no effect and apigenin showed only a weak trend. Sulforaphane inhibited the IL-1/NFκB and Wnt3a/TCF/Lef pathways and increased TGFβ/Smad2/3 and BMP6/Smad1/5/8 signalling. Apigenin showed potent inhibition of the IL-1/NFκB and TGFβ/Smad2/3 pathways, whereas luteolin showed only weak inhibition of the IL-1/NFκB pathway. All four bioactives inhibited cytokine-induced aggrecan loss from cartilage tissue explants. The combination of sulforaphane and isoliquiritigenin was synergistic for inhibiting MMP13 gene expression in chondrocytes. We conclude that dietary-derived bioactives may be important modulators of cartilage homeostasis and synergistic relationships between bioactives may have an anti-inflammatory and chondroprotective role.
Collapse
|
18
|
Patel B, Mann GE, Chapple SJ. Concerted redox modulation by sulforaphane alleviates diabetes and cardiometabolic syndrome. Free Radic Biol Med 2018; 122:150-160. [PMID: 29427794 DOI: 10.1016/j.freeradbiomed.2018.02.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 02/07/2023]
Abstract
Diabetes and cardiometabolic disorders such as hypertension and obesity are major risk factors for the development of cardiovascular disease, with a wealth of evidence suggesting that oxidative stress is linked to the initiation and pathogenesis of these disease processes. With yearly increases in the global incidence of cardiovascular diseases (CVD) and diabetes, numerous studies have focused on characterizing whether upregulating antioxidant defenses through exogenous antioxidants (e.g. vitamin E, vitamin C) or activation of endogenous defenses (e.g. the Nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant defense pathway) may be of benefit. The dietary isothiocyanate sulforaphane (SFN) is currently the subject of several clinical trials for a variety of disease states, including the evaluation of its therapeutic potential to ameliorate diabetic and cardiometabolic complications. SFN is a well characterized and potent Nrf2 inducer, however recent studies suggest its protective actions may be in part mediated by its modulation of various pro-inflammatory (e.g. Nuclear factor-kappa B (NFκB)) and metabolic (e.g. Peroxisome Proliferator-Activator Receptor Gamma (PPARγ)) signaling pathways. The focus of this review is to provide a detailed analysis of the known mechanisms by which SFN modulates Nrf2, NFκB and PPARγ signaling and crosstalk and to provide a critical evaluation of the evidence linking these transcriptional pathways with diabetic and cardiometabolic complications and SFN mediated cytoprotection. To allow comparison between rodent and human studies, we discuss the published bioavailability of SFN metabolites achieved in rodents and man in the context of Nrf2, NFκB and PPARγ signaling. Furthermore, we provide an update on the functional outcomes and implicated signaling pathways reported in recent clinical trials with SFN in Type 2 diabetic patients.
Collapse
Affiliation(s)
- Bijal Patel
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Giovanni E Mann
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Sarah J Chapple
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom.
| |
Collapse
|
19
|
Sexually dimorphic tibia shape is linked to natural osteoarthritis in STR/Ort mice. Osteoarthritis Cartilage 2018; 26:807-817. [PMID: 29604337 PMCID: PMC5987380 DOI: 10.1016/j.joca.2018.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/14/2018] [Accepted: 03/19/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Human osteoarthritis (OA) is detected only at late stages. Male STR/Ort mice develop knee OA spontaneously with known longitudinal trajectory, offering scope to identify OA predisposing factors. We exploit the lack of overt OA in female STR/Ort and in both sexes of parental, control CBA mice to explore whether early divergence in tibial bone mass or shape are linked to emergent OA. METHOD We undertook detailed micro-CT comparisons of trabecular and cortical bone, multiple structural/architectural parameters and finite element modelling (FEM) of the tibia from male and female STR/Ort and CBA mice at 8-10 (pre-OA), 18-20 (OA onset) and 40 + weeks (advanced OA) of age. RESULTS We found higher trabecular bone mass in female STR/Ort than in either OA-prone male STR/Ort or non-prone CBA mice. Cortical bone, as expected, showed greater cross-sectional area in male than female CBA, which surprisingly was reversed in STR/Ort mice. STR/Ort also exhibited higher cortical bone mass than CBA mice. Our analyses revealed similar tibial ellipticity, yet greater predicted resistance to torsion in male than female CBA mice. In contrast, male STR/Ort exhibited greater ellipticity than both female STR/Ort and CBA mice at specific cortical sites. Longitudinal analysis revealed greater tibia curvature and shape deviations in male STR/Ort mice that coincided with onset and were more pronounced in late OA. CONCLUSION Generalised higher bone mass in STR/Ort mice is more marked in non OA-prone females, but pre-OA divergence in bone shape is restricted to male STR/Ort mice in which OA develops spontaneously.
Collapse
|
20
|
Javaheri B, Carriero A, Wood M, De Souza R, Lee PD, Shefelbine S, Pitsillides AA. Transient peak-strain matching partially recovers the age-impaired mechanoadaptive cortical bone response. Sci Rep 2018; 8:6636. [PMID: 29703931 PMCID: PMC5924380 DOI: 10.1038/s41598-018-25084-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/16/2018] [Indexed: 12/14/2022] Open
Abstract
Mechanoadaptation maintains bone mass and architecture; its failure underlies age-related decline in bone strength. It is unclear whether this is due to failure of osteocytes to sense strain, osteoblasts to form bone or insufficient mechanical stimulus. Mechanoadaptation can be restored to aged bone by surgical neurectomy, suggesting that changes in loading history can rescue mechanoadaptation. We use non-biased, whole-bone tibial analyses, along with characterisation of surface strains and ensuing mechanoadaptive responses in mice at a range of ages, to explore whether sufficient load magnitude can activate mechanoadaptation in aged bone. We find that younger mice adapt when imposed strains are lower than in mature and aged bone. Intriguingly, imposition of short-term, high magnitude loading effectively primes cortical but not trabecular bone of aged mice to respond. This response was regionally-matched to highest strains measured by digital image correlation and to osteocytic mechanoactivation. These data indicate that aged bone’s loading response can be partially recovered, non-invasively by transient, focal high strain regions. Our results indicate that old murine bone does respond to load when the loading is of sufficient magnitude, and bones’ age-related adaptation failure may be due to insufficient mechanical stimulus to trigger mechanoadaptation.
Collapse
Affiliation(s)
- Behzad Javaheri
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK.
| | - Alessandra Carriero
- The City College of New York, Department of Biomedical Engineering, 160 Convent Avenue, New York, NY, 10031, USA
| | - Maria Wood
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Roberto De Souza
- Universidade Federal de Mato Grosso (UFMT), Departamento de Clínica, Av. Fernando Corrêa da Costa, 2367 - Boa Esperança, Cuiabá, 78060-900, Brazil
| | - Peter D Lee
- Manchester X-Ray Imaging Facility, University of Manchester, Research Complex at Harwell, RAL, Didcot, OX11 0FA, UK
| | - Sandra Shefelbine
- Department of Mechanical and Industrial Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Andrew A Pitsillides
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| |
Collapse
|