1
|
Fahey JW, Liu H, Batt H, Panjwani AA, Tsuji P. Sulforaphane and Brain Health: From Pathways of Action to Effects on Specific Disorders. Nutrients 2025; 17:1353. [PMID: 40284217 PMCID: PMC12030691 DOI: 10.3390/nu17081353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
The brain accounts for about 2% of the body's weight, but it consumes about 20% of the body's energy at rest, primarily derived from ATP produced in mitochondria. The brain thus has a high mitochondrial density in its neurons because of its extensive energy demands for maintaining ion gradients, neurotransmission, and synaptic activity. The brain is also extremely susceptible to damage and dysregulation caused by inflammation (neuroinflammation) and oxidative stress. Many systemic challenges to the brain can be mitigated by the phytochemical sulforaphane (SF), which is particularly important in supporting mitochondrial function. SF or its biogenic precursor glucoraphanin, from broccoli seeds or sprouts, can confer neuroprotective and cognitive benefits via diverse physiological and biochemical mechanisms. SF is able to cross the blood-brain barrier as well as to protect it, and it mitigates the consequences of destructive neuroinflammation. It also protects against the neurotoxic effects of environmental pollutants, combats the tissue and cell damage wrought by advanced glycation end products (detoxication), and supports healthy glucose metabolism. These effects are applicable to individuals of all ages, from the developing brains in periconception and infancy, to cognitively, developmentally, and traumatically challenged brains, to those in later life as well as those who are suffering with multiple chronic conditions including Parkinson's and Alzheimer's diseases.
Collapse
Affiliation(s)
- Jed W. Fahey
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry & Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- iMIND Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute of Medicine, University of Maine, Orono, ME 04469, USA
| | - Hua Liu
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Holly Batt
- Anti-AGEs Foundation, Depew, NY 14043, USA;
| | - Anita A. Panjwani
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA;
- Center on Aging and the Life Course, Purdue University, West Lafayette, IN 47907, USA
| | - Petra Tsuji
- Department of Biological Sciences, Towson University, Towson, MD 21252, USA;
| |
Collapse
|
2
|
Chang CY, Hernández-Armengol R, Paul K, Lee JY, Nance K, Shibata T, Yue P, Stehlik C, Gibb DR. CDDO-Imidazole regulates RBC alloimmunization to the KEL antigen by activating Nrf2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.645598. [PMID: 40235992 PMCID: PMC11996576 DOI: 10.1101/2025.04.03.645598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
During red blood cell (RBC) transfusion, production of alloantibodies can promote significant hemolytic events. However, most transfusion recipients do not form anti-RBC alloantibodies. Identifying mechanisms that inhibit alloimmunization may lead to prophylactic interventions. One potential regulatory mechanism is activation of the transcription factor, nuclear factor erythroid-derived 2-like 2 (Nrf2), a master regulatory of antioxidant pathways. Pharmacologic Nrf2 activators improve sequelae of sickle cell disease in pre-clinical models. The Nrf2 activator, 1-[2-cyano-3-,12-dioxooleana-1,9(11)-dien-28-oyl]imidazole (CDDO-Im), suppresses production of inflammatory cytokines including type 1 interferons (IFNα/β), which have been implicated in promoting RBC alloimmunization in transfusion models. Thus, we tested the hypothesis that the Nrf2 activator, CDDO-Im, regulates RBC alloimmunization. Here, we report that CDDO-Im induced Nrf2 activated gene expression and suppressed poly(I:C)-induced IFNα/β-stimulated gene (ISG) expression in human macrophages and murine blood leukocytes. In addition, following transfusion of wildtype mice with RBCs expressing the KEL antigen, CDDO-Im treatment inhibited poly(I:C)-induced anti-KEL IgG production and promoted post-transfusion recovery of KEL+ RBCs, but failed to do so in Nrf2 -/- mice. Results indicate that activation of the Nrf2 antioxidant pathway regulates RBC alloimmunization to the KEL antigen in a pre-clinical model. If findings translate to other models and human studies, Nrf2 activators may represent a potential prophylactic intervention to inhibit alloimmunization. Key Points The antioxidant pathway, Nrf2, inhibits anti-RBC alloantibody responses in a pre-clinical transfusion model.Nrf2 activation may represent a prophylactic strategy to inhibit RBC alloimmunization in transfusion recipients.
Collapse
|
3
|
Cuadrado A, Cazalla E, Bach A, Bathish B, Naidu SD, DeNicola GM, Dinkova-Kostova AT, Fernández-Ginés R, Grochot-Przeczek A, Hayes JD, Kensler TW, León R, Liby KT, López MG, Manda G, Shivakumar AK, Hakomäki H, Moerland JA, Motohashi H, Rojo AI, Sykiotis GP, Taguchi K, Valverde ÁM, Yamamoto M, Levonen AL. Health position paper and redox perspectives - Bench to bedside transition for pharmacological regulation of NRF2 in noncommunicable diseases. Redox Biol 2025; 81:103569. [PMID: 40059038 PMCID: PMC11970334 DOI: 10.1016/j.redox.2025.103569] [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: 12/21/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a redox-activated transcription factor regulating cellular defense against oxidative stress, thereby playing a pivotal role in maintaining cellular homeostasis. Its dysregulation is implicated in the progression of a wide array of human diseases, making NRF2 a compelling target for therapeutic interventions. However, challenges persist in drug discovery and safe targeting of NRF2, as unresolved questions remain especially regarding its context-specific role in diseases and off-target effects. This comprehensive review discusses the dualistic role of NRF2 in disease pathophysiology, covering its protective and/or destructive roles in autoimmune, respiratory, cardiovascular, and metabolic diseases, as well as diseases of the digestive system and cancer. Additionally, we also review the development of drugs that either activate or inhibit NRF2, discuss main barriers in translating NRF2-based therapies from bench to bedside, and consider the ways to monitor NRF2 activation in vivo.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Eduardo Cazalla
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anders Bach
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Gina M DeNicola
- Department of Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Raquel Fernández-Ginés
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28007, Madrid, Spain
| | - Karen T Liby
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Manuela G López
- Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain; Instituto Teófilo Hernando, Madrid, Spain
| | - Gina Manda
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | | | - Henriikka Hakomäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jessica A Moerland
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Hozumi Motohashi
- Department of Medical Biochemistry, Graduate School of Medicine Tohoku University, Sendai, Japan; Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Keiko Taguchi
- Laboratory of Food Chemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan; Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas "Sols-Morreale" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
4
|
Chen JG, Zhu YR, Qian GS, Wang JB, Lu JH, Kensler TW, Jacobson LP, Muñoz A, Groopman JD. Fifty Years of Aflatoxin Research in Qidong, China: A Celebration of Team Science to Improve Public Health. Toxins (Basel) 2025; 17:79. [PMID: 39998096 PMCID: PMC11860843 DOI: 10.3390/toxins17020079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
The Qidong Liver Cancer Institute (QDLCI) and the Qidong Cancer Registry were established in 1972 with input from doctors, other medical practitioners, and non-medical investigators arriving from urban centers such as Shanghai and Nanjing. Medical teams were established to quantify the extent of primary liver cancer in Qidong, a corn-growing peninsula on the north side of the Yangtze River. High rates of liver cancer were documented and linked to several etiologic agents, including aflatoxins. Local corn, the primary dietary staple, was found to be consistently contaminated with high levels of aflatoxins, and bioassays using this corn established its carcinogenicity in ducks and rats. Observational studies noted a positive association between levels of aflatoxin in corn and incidence of liver cancer across townships. Biomarker studies measuring aflatoxin B1 and its metabolite aflatoxin M1 in biofluids reflected the exposures. Approaches to decontamination of corn from aflatoxins were also studied. In 1993, investigators from Johns Hopkins University were invited to visit the QDLCI to discuss chemoprevention studies in some townships. A series of placebo-controlled clinical trials were conducted using oltipraz (a repurposed drug), chlorophyllin (an over-the-counter drug), and beverages prepared from 3-day-old broccoli sprouts (rich in the precursor phytochemical for sulforaphane). Modulation of biomarkers of aflatoxin DNA and albumin adducts established proof of principle for the efficacy of these agents in enhancing aflatoxin detoxication. Serendipitously, by 2012, aflatoxin exposures quantified using biomarker measurements documented a many hundred-fold reduction. In turn, the Cancer Registry documents that the age-standardized incidence rate of liver cancer is now 75% lower than that seen in the 1970s. This reduction is seen in Qidongese who have never received the hepatitis B vaccination. Aflatoxin mitigation driven by economic changes switched the dietary staple of contaminated corn to rice coupled with subsequent dietary diversity leading to lower aflatoxin exposures. This 50-year effort to understand the etiology of liver cancer in Qidong provides the strongest evidence for aflatoxin mitigation as a public health strategy for reducing liver cancer burden in exposed, high-risk populations. Also highlighted are the challenges and successes of international team science to solve pressing public health issues.
Collapse
Affiliation(s)
- Jian-Guo Chen
- Department of Epidemiology, Qidong Liver Cancer Institute, Qidong People’s Hospital, Affiliated Qidong Hospital of Nantong University, Qidong 226200, China; (Y.-R.Z.); (J.-B.W.); (J.-H.L.)
| | - Yuan-Rong Zhu
- Department of Epidemiology, Qidong Liver Cancer Institute, Qidong People’s Hospital, Affiliated Qidong Hospital of Nantong University, Qidong 226200, China; (Y.-R.Z.); (J.-B.W.); (J.-H.L.)
| | - Geng-Sun Qian
- Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai 200032, China;
| | - Jin-Bing Wang
- Department of Epidemiology, Qidong Liver Cancer Institute, Qidong People’s Hospital, Affiliated Qidong Hospital of Nantong University, Qidong 226200, China; (Y.-R.Z.); (J.-B.W.); (J.-H.L.)
| | - Jian-Hua Lu
- Department of Epidemiology, Qidong Liver Cancer Institute, Qidong People’s Hospital, Affiliated Qidong Hospital of Nantong University, Qidong 226200, China; (Y.-R.Z.); (J.-B.W.); (J.-H.L.)
| | - Thomas W. Kensler
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Lisa P. Jacobson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (L.P.J.); (A.M.)
| | - Alvaro Muñoz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (L.P.J.); (A.M.)
| | - John D. Groopman
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| |
Collapse
|
5
|
Peñata-Taborda A, Espitia-Pérez P, Espitia-Pérez L, Coneo-Pretelt A, Brango H, Ricardo-Caldera D, Arteaga-Arroyo G, Jiménez-Vidal L, Galeano-Páez C, Pastor-Sierra K, Humanez-Alvarez A, Bru-Cordero O, Jones-Cifuentes N, Rincón-Orozco B, Mendez-Sanchez S, Negrette-Guzmán M. Combination of Low-Dose Sulforaphane and Docetaxel on Mitochondrial Function and Metabolic Reprogramming in Prostate Cancer Cell Lines. Int J Mol Sci 2025; 26:1013. [PMID: 39940782 PMCID: PMC11817897 DOI: 10.3390/ijms26031013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 02/16/2025] Open
Abstract
Considering the limitations of monotherapies due to chemoresistance and side effects, this research aimed to determine whether low doses of sulforaphane (SFN) combined with docetaxel (DCT) could enhance therapeutic efficacy. Prostate cancer cell lines LNCaP and PC-3 were treated with individual IC50 doses of SFN and DCT and half-reduced IC50 values for the SFN:DCT combination. Metabolic markers, including glucose consumption, lactate production, reactive oxygen species (ROS), mitochondrial mass, and caspase activity, were assessed. In LNCaP cells, the SFN:DCT combination reduced cell viability to 50%, comparable to DCT monotherapy (48%). Caspase 3 activation was also higher with SFN:DCT (2.4 ± 0.75 RFU) than DCT alone (2.1 ± 0.47 RFU), while caspase 8 activation remained comparable, indicating equivalent effectiveness at lower concentrations. In PC-3 cells, the combination induced caspase 3 activation (1.16 ± 0.0484 RFU) at levels slightly lower than DCT (1.51 ± 0.2062 RFU) but achieved greater reductions in mitochondrial mass, reflecting its ability to target metabolic vulnerabilities in aggressive phenotypes. Our findings suggest that the SFN:DCT combination is a promising strategy for early-stage prostate cancer. By achieving comparable efficacy to DCT monotherapy at low doses, the SFN:DCT combination maintains the therapeutic impact, mitigating the adverse effects of conventional DCT treatment.
Collapse
Affiliation(s)
- Ana Peñata-Taborda
- Grupo de Investigación Biomédicas y Biología Molecular, Universidad del Sinú E.B.Z., Montería 230001, Colombia; (A.P.-T.); (P.E.-P.); (A.C.-P.); (G.A.-A.); (L.J.-V.); (C.G.-P.); (K.P.-S.); (A.H.-A.)
| | - Pedro Espitia-Pérez
- Grupo de Investigación Biomédicas y Biología Molecular, Universidad del Sinú E.B.Z., Montería 230001, Colombia; (A.P.-T.); (P.E.-P.); (A.C.-P.); (G.A.-A.); (L.J.-V.); (C.G.-P.); (K.P.-S.); (A.H.-A.)
| | - Lyda Espitia-Pérez
- Grupo de Investigación Biomédicas y Biología Molecular, Universidad del Sinú E.B.Z., Montería 230001, Colombia; (A.P.-T.); (P.E.-P.); (A.C.-P.); (G.A.-A.); (L.J.-V.); (C.G.-P.); (K.P.-S.); (A.H.-A.)
| | - Andrés Coneo-Pretelt
- Grupo de Investigación Biomédicas y Biología Molecular, Universidad del Sinú E.B.Z., Montería 230001, Colombia; (A.P.-T.); (P.E.-P.); (A.C.-P.); (G.A.-A.); (L.J.-V.); (C.G.-P.); (K.P.-S.); (A.H.-A.)
| | - Hugo Brango
- Facultad de Educación y Ciencias, Departamento de Matemáticas, Universidad de Sucre, Sincelejo 700003, Colombia;
| | - Dina Ricardo-Caldera
- Grupo de Investigación Enfermedades Tropicales y Resistencia Bacteriana, Universidad del Sinú E.B.Z., Montería 230001, Colombia;
| | - Gean Arteaga-Arroyo
- Grupo de Investigación Biomédicas y Biología Molecular, Universidad del Sinú E.B.Z., Montería 230001, Colombia; (A.P.-T.); (P.E.-P.); (A.C.-P.); (G.A.-A.); (L.J.-V.); (C.G.-P.); (K.P.-S.); (A.H.-A.)
| | - Luisa Jiménez-Vidal
- Grupo de Investigación Biomédicas y Biología Molecular, Universidad del Sinú E.B.Z., Montería 230001, Colombia; (A.P.-T.); (P.E.-P.); (A.C.-P.); (G.A.-A.); (L.J.-V.); (C.G.-P.); (K.P.-S.); (A.H.-A.)
| | - Claudia Galeano-Páez
- Grupo de Investigación Biomédicas y Biología Molecular, Universidad del Sinú E.B.Z., Montería 230001, Colombia; (A.P.-T.); (P.E.-P.); (A.C.-P.); (G.A.-A.); (L.J.-V.); (C.G.-P.); (K.P.-S.); (A.H.-A.)
| | - Karina Pastor-Sierra
- Grupo de Investigación Biomédicas y Biología Molecular, Universidad del Sinú E.B.Z., Montería 230001, Colombia; (A.P.-T.); (P.E.-P.); (A.C.-P.); (G.A.-A.); (L.J.-V.); (C.G.-P.); (K.P.-S.); (A.H.-A.)
| | - Alicia Humanez-Alvarez
- Grupo de Investigación Biomédicas y Biología Molecular, Universidad del Sinú E.B.Z., Montería 230001, Colombia; (A.P.-T.); (P.E.-P.); (A.C.-P.); (G.A.-A.); (L.J.-V.); (C.G.-P.); (K.P.-S.); (A.H.-A.)
| | - Osnamir Bru-Cordero
- Dirección Académica, Universidad Nacional de Colombia, Kilómetro 9, Vía Valledupar-La Paz, La Paz 202010, Colombia;
| | - Nathalia Jones-Cifuentes
- Departamento de Ciencias Básicas, Escuela de Medicina, Universidad Industrial de Santander, Bucaramanga 680002, Colombia; (N.J.-C.); (B.R.-O.); (M.N.-G.)
| | - Bladimiro Rincón-Orozco
- Departamento de Ciencias Básicas, Escuela de Medicina, Universidad Industrial de Santander, Bucaramanga 680002, Colombia; (N.J.-C.); (B.R.-O.); (M.N.-G.)
| | - Stelia Mendez-Sanchez
- Escuela de Química, Universidad Industrial de Santander, Bucaramanga 680002, Colombia;
| | - Mario Negrette-Guzmán
- Departamento de Ciencias Básicas, Escuela de Medicina, Universidad Industrial de Santander, Bucaramanga 680002, Colombia; (N.J.-C.); (B.R.-O.); (M.N.-G.)
| |
Collapse
|
6
|
Krisanits BA, Kaur B, Fahey JW, Turner DP. The Anti-AGEing and RAGEing Potential of Isothiocyanates. Molecules 2024; 29:5986. [PMID: 39770075 PMCID: PMC11677037 DOI: 10.3390/molecules29245986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/03/2025] Open
Abstract
Isothiocyanates (ITCs), found in edible plants such as cruciferous vegetables, are a group of reactive organo-sulfur phytochemicals produced by the hydrolysis of precursors known as glucosinolates. ITCs have been studied extensively both in vivo and in vitro to define their therapeutic potential for the treatment of chronic health conditions. Therapeutically, they have shown an intrinsic ability to inhibit oxidative and inflammatory phenotypes to support enhanced health. This review summarizes the current evidence supporting the observation that the antioxidant and anti-inflammatory activities of ITCs temper the pathogenic effects of a group of reactive metabolites called advanced glycation end products (AGEs). AGE exposure has significantly increased across the lifespan due to health risk factors that include dietary intake, a sedentary lifestyle, and comorbid conditions. By contributing to a chronic cycle of inflammatory stress through the aberrant activation of the transmembrane receptor for AGE (RAGE), increased AGE bioavailability is associated with chronic disease onset, progression, and severity. This review debates the potential molecular mechanisms by which ITCs may inhibit AGE bioavailability to reduce RAGE-mediated pro-oxidant and pro-inflammatory phenotypes. Bringing to light the molecular impact that ITCs may have on AGE biogenesis may stimulate novel intervention strategies for reversing or preventing the impact of lifestyle factors on chronic disease risk.
Collapse
Affiliation(s)
- Bradley A. Krisanits
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA; (B.A.K.); (B.K.)
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Bhoomika Kaur
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA; (B.A.K.); (B.K.)
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Jed W. Fahey
- Departments of Medicine, Pharmacology & Molecular Sciences, Psychiatry & Behavioral Sciences, and iMIND Hopkins, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Institute of Medicine, University of Maine, Orono, ME 04469, USA
| | - David P. Turner
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA; (B.A.K.); (B.K.)
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
7
|
Giron J, Smiarowski L, Katz J. The effect of sulforaphane on markers of inflammation and metabolism in virally suppressed HIV patients. Front Nutr 2024; 11:1357906. [PMID: 39539366 PMCID: PMC11557404 DOI: 10.3389/fnut.2024.1357906] [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: 12/19/2023] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
There are currently 1.2 million people living with HIV (Human Immunodeficiency Virus) in the United States. Virally suppressed HIV patients commonly experience chronic inflammation which increases the risk for other chronic conditions. This inflammation can be quantified with a variety of biomarkers. Some current antiretroviral compounds bring about metabolic abnormalities and promote weight gain often associated with increases in visceral adipose tissue (VAT) and an increase in the risk of diabetes mellitus and cardiovascular disease. Sulforaphane, an isothiocyanate found in cruciferous vegetables, has shown efficacy in animal models by reducing lipid levels, lowering inflammatory markers, and decreasing fat mass. A double-blind randomized controlled pilot study with 14 virally suppressed HIV patients was conducted to evaluate the effects of 40 mg (225 μmol) of sulforaphane, once daily, over 12 weeks, followed by a 4-week washout period. There was a significant decrease in C-reactive protein compared to the control group (p = 0.019). Sulforaphane has been studied in a multitude of conditions and diseases, but this is the first study in a human population of patients living with HIV.
Collapse
Affiliation(s)
- Jose Giron
- Sunshine Specialty Health Care, Orlando, FL, United States
- Department of Medicine, Florida State University, College of Medicine, Tallahassee, FL, United States
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Lauren Smiarowski
- Department of Medicine, Florida State University, College of Medicine, Tallahassee, FL, United States
- OrlandoHealth- Arnold Palmer Hospital for Children, Orlando, FL, United States
| | - Johannah Katz
- Florida Center for Hormones and Wellness, Orlando, FL, United States
| |
Collapse
|
8
|
Yang Y, Eguchi A, Mori C, Hashimoto K. Dietary sulforaphane glucosinolate mitigates depression-like behaviors in mice with hepatic ischemia/reperfusion injury: A role of the gut-liver-brain axis. J Psychiatr Res 2024; 176:129-139. [PMID: 38857554 DOI: 10.1016/j.jpsychires.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Nutrition has been increasingly recognized for its use in mental health. Depression is commonly observed in patients with chronic liver disease (CLD). Building on our recent findings of depression-like behaviors in mice with hepatic ischemia/reperfusion (HI/R) injury, mediated by the gut-liver-brain axis, this study explored the potential influence of dietary sulforaphane glucosinolate (SGS) on these behaviors. Behavioral assessments for depression-like behaviors were conducted 7 days post either sham or HI/R injury surgery. Dietary intake of SGS significantly prevented splenomegaly, systemic inflammation, depression-like behaviors, and downregulation of synaptic proteins in the prefrontal cortex (PFC) of HI/R-injured mice. Through 16S rRNA analysis and untargeted metabolomic analyses, distinct bacterial profiles and metabolites were identified between control + HI/R group and SGS + HI/R group. Correlations were observed between the relative abundance of gut microbiota and both behavioral outcomes and blood metabolites. These findings suggest that SGS intake could mitigate depression-like phenotypes in mice with HI/R injury, potentially through the gut-liver-brain axis. Additionally, SGS, found in crucial vegetables like broccoli, could offer prophylactic nutritional benefits for depression in patients with CLD.
Collapse
Affiliation(s)
- Yong Yang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan; Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Akifumi Eguchi
- Department of Sustainable Health Science, Chiba University Center for Preventive Medical Sciences, Chiba, 263-8522, Japan
| | - Chisato Mori
- Department of Sustainable Health Science, Chiba University Center for Preventive Medical Sciences, Chiba, 263-8522, Japan; Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
9
|
Jacquier EF, Kassis A, Marcu D, Contractor N, Hong J, Hu C, Kuehn M, Lenderink C, Rajgopal A. Phytonutrients in the promotion of healthspan: a new perspective. Front Nutr 2024; 11:1409339. [PMID: 39070259 PMCID: PMC11272662 DOI: 10.3389/fnut.2024.1409339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Considering a growing, aging population, the need for interventions to improve the healthspan in aging are tantamount. Diet and nutrition are important determinants of the aging trajectory. Plant-based diets that provide bioactive phytonutrients may contribute to offsetting hallmarks of aging and reducing the risk of chronic disease. Researchers now advocate moving toward a positive model of aging which focuses on the preservation of functional abilities, rather than an emphasis on the absence of disease. This narrative review discusses the modulatory effect of nutrition on aging, with an emphasis on promising phytonutrients, and their potential to influence cellular, organ and functional parameters in aging. The literature is discussed against the backdrop of a recent conceptual framework which describes vitality, intrinsic capacity and expressed capacities in aging. This aims to better elucidate the role of phytonutrients on vitality and intrinsic capacity in aging adults. Such a review contributes to this new scientific perspective-namely-how nutrition might help to preserve functional abilities in aging, rather than purely offsetting the risk of chronic disease.
Collapse
Affiliation(s)
| | | | - Diana Marcu
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Jina Hong
- Amway Innovation and Science, Ada, MI, United States
| | - Chun Hu
- Amway Innovation and Science, Ada, MI, United States
| | - Marissa Kuehn
- Amway Innovation and Science, Ada, MI, United States
| | | | - Arun Rajgopal
- Amway Innovation and Science, Ada, MI, United States
| |
Collapse
|
10
|
Ross SA, Emenaker NJ, Kumar A, Riscuta G, Biswas K, Gupta S, Mohammed A, Shoemaker RH. Green Cancer Prevention and Beyond. Cancer Prev Res (Phila) 2024; 17:107-118. [PMID: 38251904 PMCID: PMC10911807 DOI: 10.1158/1940-6207.capr-23-0308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/13/2023] [Accepted: 01/18/2024] [Indexed: 01/23/2024]
Abstract
The concept of green chemoprevention was introduced in 2012 by Drs. Jed Fahey and Thomas Kensler as whole-plant foods and/or extract-based interventions demonstrating cancer prevention activity. Refining concepts and research demonstrating proof-of-principle approaches are highlighted within this review. Early approaches included extensively investigated whole foods, including broccoli sprouts and black raspberries showing dose-responsive effects across a range of activities in both animals and humans with minimal or no apparent toxicity. A recent randomized crossover trial evaluating the detoxification of tobacco carcinogens by a broccoli seed and sprout extract in the high-risk cohort of current smokers highlights the use of a dietary supplement as a potential next-generation green chemoprevention or green cancer prevention approach. Challenges are addressed, including the selection of dose, duration and mode of delivery, choice of control group, and standardization of the plant food or extract. Identification and characterization of molecular targets and careful selection of high-risk cohorts for study are additional important considerations when designing studies. Goals for precision green cancer prevention include acquiring robust evidence from carefully controlled human studies linking plant foods, extracts, and compounds to modulation of targets for cancer risk reduction in individual cancer types.
Collapse
Affiliation(s)
- Sharon A. Ross
- Division of Cancer Prevention, Nutritional Sciences Research Group, National Cancer Institute, Rockville, Maryland
| | - Nancy J. Emenaker
- Division of Cancer Prevention, Nutritional Sciences Research Group, National Cancer Institute, Rockville, Maryland
| | - Amit Kumar
- Division of Cancer Prevention, Nutritional Sciences Research Group, National Cancer Institute, Rockville, Maryland
| | - Gabriela Riscuta
- Division of Cancer Prevention, Nutritional Sciences Research Group, National Cancer Institute, Rockville, Maryland
| | - Kajal Biswas
- Division of Cancer Prevention, Chemopreventive Agent Development Research Group, National Cancer Institute, Rockville, Maryland
| | - Shanker Gupta
- Division of Cancer Prevention, Chemopreventive Agent Development Research Group, National Cancer Institute, Rockville, Maryland
| | - Altaf Mohammed
- Division of Cancer Prevention, Chemopreventive Agent Development Research Group, National Cancer Institute, Rockville, Maryland
| | - Robert H. Shoemaker
- Division of Cancer Prevention, Chemopreventive Agent Development Research Group, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
11
|
Bouranis JA, Beaver LM, Wong CP, Choi J, Hamer S, Davis EW, Brown KS, Jiang D, Sharpton TJ, Stevens JF, Ho E. Sulforaphane and Sulforaphane-Nitrile Metabolism in Humans Following Broccoli Sprout Consumption: Inter-individual Variation, Association with Gut Microbiome Composition, and Differential Bioactivity. Mol Nutr Food Res 2024; 68:e2300286. [PMID: 38143283 PMCID: PMC10922398 DOI: 10.1002/mnfr.202300286] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/14/2023] [Indexed: 12/26/2023]
Abstract
SCOPE The glucosinolate glucoraphanin from broccoli is converted to sulforaphane (SFN) or sulforaphane-nitrile (SFN-NIT) by plant enzymes or the gut microbiome. Human feeding studies typically observe high inter-individual variation in absorption and excretion of SFN, however, the source of this variation is not fully known. To address this, a human feeding trial to comprehensively evaluate inter-individual variation in the absorption and excretion of all known SFN metabolites in urine, plasma, and stool, and tested the hypothesis that gut microbiome composition influences inter-individual variation in total SFN excretion has been conducted. METHODS AND RESULTS Participants (n = 55) consumed a single serving of broccoli or alfalfa sprouts and plasma, stool, and total urine are collected over 72 h for quantification of SFN metabolites and gut microbiome profiling using 16S gene sequencing. SFN-NIT excretion is markedly slower than SFN excretion (72 h vs 24 h). Members of genus Bifidobacterium, Dorea, and Ruminococcus torques are positively associated with SFN metabolite excretion while members of genus Alistipes and Blautia has a negative association. CONCLUSION This is the first report of SFN-NIT metabolite levels in human plasma, urine, and stool following consumption of broccoli sprouts. The results help explain factors driving inter-individual variation in SFN metabolism and are relevant for precision nutrition.
Collapse
Affiliation(s)
- John A Bouranis
- College of Health, Oregon State University, Corvallis, Oregon, USA
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
| | - Laura M Beaver
- College of Health, Oregon State University, Corvallis, Oregon, USA
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
| | - Carmen P Wong
- College of Health, Oregon State University, Corvallis, Oregon, USA
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
| | - Sean Hamer
- College of Health, Oregon State University, Corvallis, Oregon, USA
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
| | - Ed W Davis
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
- Center for Quantitative Life Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Kevin S Brown
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Duo Jiang
- Department of Statistics, Oregon State University, Corvallis, Oregon, USA
| | - Thomas J Sharpton
- Department of Statistics, Oregon State University, Corvallis, Oregon, USA
- Department of Microbiology, Oregon State University, Corvallis, Oregon, USA
| | - Jan F Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Emily Ho
- College of Health, Oregon State University, Corvallis, Oregon, USA
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
| |
Collapse
|
12
|
Gasparello J, Marzaro G, Papi C, Gentili V, Rizzo R, Zurlo M, Scapoli C, Finotti A, Gambari R. Effects of Sulforaphane on SARS‑CoV‑2 infection and NF‑κB dependent expression of genes involved in the COVID‑19 'cytokine storm'. Int J Mol Med 2023; 52:76. [PMID: 37477130 PMCID: PMC10555481 DOI: 10.3892/ijmm.2023.5279] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 06/09/2023] [Indexed: 07/22/2023] Open
Abstract
Since its spread at the beginning of 2020, the coronavirus disease 2019 (COVID‑19) pandemic represents one of the major health problems. Despite the approval, testing, and worldwide distribution of anti‑severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) vaccines, the development of specific antiviral agents targeting the SARS‑CoV‑2 life cycle with high efficiency, and/or interfering with the associated 'cytokine storm', is highly required. A recent study, conducted by the authors' group indicated that sulforaphane (SFN) inhibits the expression of IL‑6 and IL‑8 genes induced by the treatment of IB3‑1 bronchial cells with a recombinant spike protein of SARS‑CoV‑2. In the present study, the ability of SFN to inhibit SARS‑CoV‑2 replication and the expression of pro‑inflammatory genes encoding proteins of the COVID‑19 'cytokine storm' was evaluated. SARS‑CoV‑2 replication was assessed in bronchial epithelial Calu‑3 cells. Moreover, SARS‑CoV‑2 replication and expression of pro‑inflammatory genes was evaluated by reverse transcription quantitative droplet digital PCR. The effects on the expression levels of NF‑κB were assessed by western blotting. Molecular dynamics simulations of NF‑kB/SFN interactions were conducted with Gromacs 2021.1 software under the Martini 2 CG force field. Computational studies indicated that i) SFN was stably bound with the NF‑κB monomer; ii) a ternary NF‑kB/SFN/DNA complex was formed; iii) the SFN interacted with both the protein and the nucleic acid molecules modifying the binding mode of the latter, and impairing the full interaction between the NF‑κB protein and the DNA molecule. This finally stabilized the inactive complex. Molecular studies demonstrated that SFN i) inhibits the SARS‑CoV‑2 replication in infected Calu‑3 cells, decreasing the production of the N‑protein coding RNA sequences, ii) decreased NF‑κB content in SARS‑CoV‑2 infected cells and inhibited the expression of NF‑kB‑dependent IL‑1β and IL‑8 gene expression. The data obtained in the present study demonstrated inhibitory effects of SFN on the SARS‑CoV‑2 life cycle and on the expression levels of the pro‑inflammatory genes, sustaining the possible use of SFN in the management of patients with COVID‑19.
Collapse
Affiliation(s)
- Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, I-35131 Padova
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Valentina Gentili
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, I-44121 Ferrara, Italy
| | - Roberta Rizzo
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, I-44121 Ferrara, Italy
| | - Matteo Zurlo
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Chiara Scapoli
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara
| |
Collapse
|
13
|
Mordecai J, Ullah S, Ahmad I. Sulforaphane and Its Protective Role in Prostate Cancer: A Mechanistic Approach. Int J Mol Sci 2023; 24:ijms24086979. [PMID: 37108142 PMCID: PMC10138336 DOI: 10.3390/ijms24086979] [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: 03/16/2023] [Revised: 04/08/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
The increasing incidence of prostate cancer worldwide has spurred research into novel therapeutics for its treatment and prevention. Sulforaphane, derived from broccoli and other members of the Brassica genus, is a phytochemical shown to have anticancer properties. Numerous studies have shown that sulforaphane prevents the development and progression of prostatic tumors. This review evaluates the most recent published reports on prevention of the progression of prostate cancer by sulforaphane in vitro, in vivo and in clinical settings. A detailed description of the proposed mechanisms of action of sulforaphane on prostatic cells is provided. Furthermore, we discuss the challenges, limitations and future prospects of using sulforaphane as a therapeutic agent in treatment of prostate cancer.
Collapse
Affiliation(s)
- James Mordecai
- Department of Bioengineering, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia
| | - Saleem Ullah
- Department of Transdisciplinary Science and Engineering, School of Environment and Society, Tokyo Institute of Technology, Nagatsuta, Midori-ku, Yokohama 226-8503, Japan
| | - Irshad Ahmad
- Department of Bioengineering, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Membranes and Water Security, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia
| |
Collapse
|
14
|
Manai F, Govoni S, Amadio M. The Challenge of Dimethyl Fumarate Repurposing in Eye Pathologies. Cells 2022; 11:cells11244061. [PMID: 36552824 PMCID: PMC9777082 DOI: 10.3390/cells11244061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/28/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Dimethyl fumarate (DMF) is a small molecule currently approved and used in the treatment of psoriasis and multiple sclerosis due to its immuno-modulatory, anti-inflammatory, and antioxidant properties. As an Nrf2 activator through Keap1 protein inhibition, DMF unveils a potential therapeutical use that is much broader than expected so far. In this comprehensive review we discuss the state-of-art and future perspectives regarding the potential repositioning of this molecule in the panorama of eye pathologies, including Age-related Macular Degeneration (AMD). The DMF's mechanism of action, an extensive analysis of the in vitro and in vivo evidence of its beneficial effects, together with a search of the current clinical trials, are here reported. Altogether, this evidence gives an overview of the new potential applications of this molecule in the context of ophthalmological diseases characterized by inflammation and oxidative stress, with a special focus on AMD, for which our gene-disease (KEAP1-AMD) database search, followed by a protein-protein interaction analysis, further supports the rationale of DMF use. The necessity to find a topical route of DMF administration to the eye is also discussed. In conclusion, the challenge of DMF repurposing in eye pathologies is feasible and worth scientific attention and well-focused research efforts.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Stefano Govoni
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
| | - Marialaura Amadio
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy
- Correspondence: ; Tel.: +39-0382-987888
| |
Collapse
|
15
|
Microorganisms-An Effective Tool to Intensify the Utilization of Sulforaphane. Foods 2022; 11:foods11233775. [PMID: 36496582 PMCID: PMC9737538 DOI: 10.3390/foods11233775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Sulforaphane (SFN) was generated by the hydrolysis of glucoraphanin under the action of myrosinase. However, due to the instability of SFN, the bioavailability of SFN was limited. Meanwhile, the gut flora obtained the ability to synthesize myrosinase and glucoraphanin, which could be converted into SFN in the intestine. However, the ability of microorganisms to synthesize myrosinase in the gut was limited. Therefore, microorganisms with myrosinase synthesis ability need to be supplemented. With the development of research, microorganisms with high levels of myrosinase synthesis could be obtained by artificial selection and gene modification. Researchers found the SFN production rate of the transformed microorganisms could be significantly improved. However, despite applying transformation technology and regulating nutrients to microorganisms, it still could not provide the best efficiency during generating SFN and could not accomplish colonization in the intestine. Due to the great effect of microencapsulation on improving the colonization ability of microorganisms, microencapsulation is currently an important way to deliver microorganisms into the gut. This article mainly analyzed the possibility of obtaining SFN-producing microorganisms through gene modification and delivering them to the gut via microencapsulation to improve the utilization rate of SFN. It could provide a theoretical basis for expanding the application scope of SFN.
Collapse
|
16
|
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
|
17
|
Paśko P, Zagrodzki P, Okoń K, Prochownik E, Krośniak M, Galanty A. Broccoli Sprouts and Their Influence on Thyroid Function in Different In Vitro and In Vivo Models. PLANTS (BASEL, SWITZERLAND) 2022; 11:2750. [PMID: 36297774 PMCID: PMC9610815 DOI: 10.3390/plants11202750] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/05/2022] [Accepted: 10/15/2022] [Indexed: 06/16/2023]
Abstract
Broccoli sprouts are a super vegetable; however, they have possible negative effects on thyroid function, which is especially important for patients with hypothyroidism. As the data on this issue are scarce, this study aimed to determine the safety and possible beneficial effect of broccoli sprouts both in vitro and in vivo. The in vitro model comprised the evaluation of the impact of broccoli sprouts on normal and neoplastic thyroid cells and the determination of their anti-inflammatory and antioxidant (IL-6, TNF-alpha, NO, and SOD) potential in macrophages. The in vivo model concerned the histopathological analysis of thyroid glands in healthy rats and rats with hypothyroidism (induced by iodine deficiency or sulfadimethoxine ingestion) fed with broccoli sprouts. The results of our study indicated that broccoli sprouts decreased the viability of thyroid cancer cells and prevented inflammation. The results also confirmed the satisfactory safety profile of the sprouts, both in vitro and in vivo; however, a further in-depth evaluation of this problem is still needed. Information on the influence of brassica vegetables on thyroid function is of great importance in terms of public health, particularly when taking into account that the risk of iodine deficiency, hypothyroidism, and thyroid cancer in the global population is still increasing.
Collapse
Affiliation(s)
- Paweł Paśko
- Department of Food Chemistry and Nutrition, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Paweł Zagrodzki
- Department of Food Chemistry and Nutrition, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Krzysztof Okoń
- Department of Pathomorphology, Jagiellonian University Medical College, Grzegórzecka 16, 31-531 Kraków, Poland
| | - Ewelina Prochownik
- Department of Food Chemistry and Nutrition, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Mirosław Krośniak
- Department of Food Chemistry and Nutrition, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Agnieszka Galanty
- Department of Pharmacognosy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| |
Collapse
|
18
|
Chartoumpekis DV, Ziros PG, Habeos IG, Sykiotis GP. Emerging roles of Keap1/Nrf2 signaling in the thyroid gland and perspectives for bench-to-bedside translation. Free Radic Biol Med 2022; 190:276-283. [PMID: 35988853 DOI: 10.1016/j.freeradbiomed.2022.08.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 12/14/2022]
Abstract
The signaling pathway centered on the transcription factor nuclear erythroid factor 2-like 2 (Nrf2) has emerged during the last 15 years as a target for the prevention and treatment of diseases broadly related with oxidative stress such as cancer, neurodegenerative and metabolic diseases. The roles of Nrf2 are expanding beyond general cytoprotection, and they encompass its crosstalk with other pathways as well as tissue-specific functions. The thyroid gland relies on reactive oxygen species for its main physiological function, the synthesis and secretion of thyroid hormones. A few years ago, Nrf2 was characterized as a central regulator of the antioxidant response in the thyroid, as well as of the transcription and processing of thyroglobulin, the major thyroidal protein that serves as the substrate for thyroid hormone synthesis. Herein, we summarize the current knowledge about the roles of Nrf2 in thyroid physiology, pathophysiology and disease. We focus specifically on the most recent publications in the field, and we discuss the implications for the preclinical and clinical use of Nrf2 modulators.
Collapse
Affiliation(s)
- Dionysios V Chartoumpekis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, CH-1011, Lausanne, Switzerland
| | - Panos G Ziros
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, CH-1011, Lausanne, Switzerland
| | - Ioannis G Habeos
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, GR-26504, Patras, Greece
| | - Gerasimos P Sykiotis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, CH-1011, Lausanne, Switzerland.
| |
Collapse
|
19
|
Long-lasting beneficial effects of maternal intake of sulforaphane glucosinolate on gut microbiota in adult offspring. J Nutr Biochem 2022; 109:109098. [PMID: 35788394 DOI: 10.1016/j.jnutbio.2022.109098] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 03/01/2022] [Accepted: 06/06/2022] [Indexed: 11/24/2022]
Abstract
Mounting evidence suggests the impact of maternal diet on the health of offspring. We reported that maternal diet of sulforaphane glucosinolate (SGS) could prevent behavioral abnormalities in offspring after maternal immune activation. The present study was designed to investigate whether the dietary intake of SGS during pregnancy and lactation influences the composition of gut microbiota in the offspring. The dietary intake of SGS during pregnancy and lactation caused significant changes in the α-diversity and β-diversity of gut microbiota in 3-week-old offspring (SGS-3W group) and 10-week-old offspring (SGS-10W group). The LEfSe algorithm identified several microbes as important phylotypes in the SGS-3W or SGS-10W groups. Predictive functional metagenomes showed that the maternal intake of SGS caused several KEGG pathways alterations with respect to the genetic information processing and metabolism. Furthermore, the plasma levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in the SGS-10W group after the injection of lipopolysaccharide (LPS: 0.5 mg/kg) were significantly lower than those of the CON-10W group. It is noteworthy that there were positive correlations between the relative abundance of the genus Blautia and IL-6 (or TNF-α) in adult offspring. Moreover, there were sex differences of gut microbiota composition in offspring. In conclusion, these data suggest that the dietary intake of SGS during pregnancy and lactation might produce long-lasting beneficial effects in adult offspring through the persistent modulation of gut microbiota. It is likely that the modulation of gut microbiota by maternal nutrition may confer resilience versus vulnerability to stress-related psychiatric disorders in the offspring.
Collapse
|
20
|
Improvement of the Stability and Release of Sulforaphane-enriched Broccoli Sprout Extract Nanoliposomes by Co-encapsulation into Basil Seed Gum. FOOD BIOPROCESS TECH 2022. [DOI: 10.1007/s11947-022-02826-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
21
|
Wang Z, Tu C, Pratt R, Khoury T, Qu J, Fahey JW, McCann SE, Zhang Y, Wu Y, Hutson AD, Ambrosone CB, Edge SB, Cappuccino HH, Takabe K, Young JS, Tang L. A Presurgical-Window Intervention Trial of Isothiocyanate-Rich Broccoli Sprout Extract in Patients with Breast Cancer. Mol Nutr Food Res 2022; 66:e2101094. [PMID: 35475592 PMCID: PMC9925304 DOI: 10.1002/mnfr.202101094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/17/2022] [Indexed: 11/09/2022]
Abstract
SCOPE Dietary isothiocyanates (ITCs) from cruciferous vegetables have shown potent anti-breast cancer activities in preclinical models, but their anticancer effects in vivo in breast cancer patients remain elusive. A proof-of-principle, presurgical window of opportunity trial is conducted to assess the anticancer effects of dietary ITCs in breast cancer patients. METHODS AND RESULTS Thirty postmenopausal breast cancer patients are randomly assigned to receive ITC-rich broccoli sprout extract (BSE) (200 µmol ITC per day) or a placebo for 2 weeks. Expression of biomarkers related to ITCs functions are measured in breast cancer tissue specimens at pre- and post-interventions using immunohistochemistry staining. First morning urine samples are collected at both timepoints for proteomic analysis. Overall, the study shows high compliance (100%) and low toxicity (no grade 4 adverse event). Trends of increase in cleaved caspase 3 and tumor-infiltrating lymphocytes (TILs) and trends of decrease in Ki-67 and nuclear to cytoplasm ratio of estrogen receptor (ER)-α are observed in the BSE arm only, consistent with the significantly altered signaling pathways identified in urinary proteomic analysis. CONCLUSIONS Anticancer activities of ITCs are observed in breast cancer patients, supporting the potential beneficial roles of ITC-containing cruciferous vegetables in breast cancer prognosis.
Collapse
Affiliation(s)
- Zinian Wang
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Chengjian Tu
- Department of Pharmaceutical Sciences, the State University of New York at Buffalo, Buffalo, NY
| | - Rachel Pratt
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Thaer Khoury
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Jun Qu
- Department of Pharmaceutical Sciences, the State University of New York at Buffalo, Buffalo, NY
| | - Jed W. Fahey
- Departments of Medicine & Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD; Department of Nutrition and Food Studies, George Mason University, Fairfax, VA
| | - Susan E. McCann
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Yuesheng Zhang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Yue Wu
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Alan D. Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Christine B. Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Stephen B. Edge
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Helen H. Cappuccino
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Jessica S. Young
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Li Tang
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| |
Collapse
|
22
|
Regulation of BDNF transcription by Nrf2 and MeCP2 ameliorates MPTP-induced neurotoxicity. Cell Death Dis 2022; 8:267. [PMID: 35595779 PMCID: PMC9122988 DOI: 10.1038/s41420-022-01063-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 05/06/2022] [Accepted: 05/12/2022] [Indexed: 12/31/2022]
Abstract
Mounting evidence suggests the key role of brain-derived neurotrophic factor (BDNF) in the dopaminergic neurotoxicity of Parkinson’s disease (PD). Activation of NF-E2-related factor-2 (Nrf2) and inhibition of methyl CpG-binding protein 2 (MeCP2) can regulate BDNF upregulation. However, the regulation of BDNF by Nrf2 and MeCP2 in the PD pathogenesis has not been reported. Here, we revealed that Nrf2/MeCP2 coordinately regulated BDNF transcription, reversing the decreased levels of BDNF expression in 1-methyl-4-phenylpyridinium (MPP+)-treated SH-SY5Y cells and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. Repeated administration of sulforaphane (SFN, an Nrf2 activator) attenuated dopaminergic neurotoxicity in MPTP-treated mice through activation of BDNF and suppression of MeCP2 expression. Furthermore, intracerebroventricular injection of MeCP2-HDO, a DNA/RNA heteroduplex oligonucleotide (HDO) silencing MeCP2 expression, ameliorated dopaminergic neurotoxicity in MPTP-treated mice via activation of Nrf2 and BDNF expression. Moreover, we found decreased levels of Nrf2 and BDNF, and increased levels of MeCP2 protein expression in the striatum of patients with dementia with Lewy bodies (DLB). Interesting, there were correlations between BDNF and Nrf2 (or MeCP2) expression in the striatum from DLB patients. Therefore, it is likely that the activation of BDNF transcription by activation of Nrf2 and/or suppression of MeCP2 could be a new therapeutic approach for PD.
Collapse
|
23
|
Ruiz-Alcaraz AJ, Martínez-Sánchez MA, García-Peñarrubia P, Martinez-Esparza M, Ramos-Molina B, Moreno DA. Analysis of the anti-inflammatory potential of Brassica bioactive compounds in a human macrophage-like cell model derived from HL-60 cells. Biomed Pharmacother 2022; 149:112804. [PMID: 35279599 DOI: 10.1016/j.biopha.2022.112804] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Chronic inflammatory diseases are major causes of global morbidity and mortality. Acute inflammation is meant to protect the body against foreign agents, but it also plays a major role in tissue repairment. Several mediators are involved in this process, including pro-inflammatory cytokines produced by macrophages. Occasionally, if the inflammatory response is not resolved, the acute inflammatory process can evolve into a chronic inflammation. Natural compounds from vegetables are considered as an important source of active agents with potential to treat or prevent inflammatory related pathologies and could be used as an alternative of the therapeutic agents currently in use, such as non-steroidal anti-inflammatory drugs (NSAIDs), which present several side effects. METHODS In this research work we evaluated in vitro the anti-inflammatory activity of a series of ten phytochemicals present in Brassica, measured as the potential of those compounds to reduce the production of key pro-inflammatory cytokines (TNF-α, IL-6 and IL-1β) by a human macrophage-like cell model of HL-60 cells RESULTS: Most of the tested phytochemicals (including the most representative bioactive molecules of the major classes of compounds present in cruciferous foods such as glucosinolates, isothiocyanates, hydroxycinnamic acids, flavonols and anthocyanins) demonstrated significant anti-inflammatory activity at micromolar level in the absence of cytotoxic effects in this human macrophage-like cell model. CONCLUSION These data confirm that phytochemicals commonly obtained from Brassica may be potential therapeutic leads to treat or prevent human chronic inflammation and related diseases.
Collapse
Affiliation(s)
- Antonio José Ruiz-Alcaraz
- Department of Biochemistry, Molecular Biology B and Immunology, School of Medicine, University of Murcia, Regional Campus of International Excellence "Campus Mare Nostrum", 30100 Murcia, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain.
| | - María Antonia Martínez-Sánchez
- Department of Biochemistry, Molecular Biology B and Immunology, School of Medicine, University of Murcia, Regional Campus of International Excellence "Campus Mare Nostrum", 30100 Murcia, Spain; Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
| | - Pilar García-Peñarrubia
- Department of Biochemistry, Molecular Biology B and Immunology, School of Medicine, University of Murcia, Regional Campus of International Excellence "Campus Mare Nostrum", 30100 Murcia, Spain
| | - María Martinez-Esparza
- Department of Biochemistry, Molecular Biology B and Immunology, School of Medicine, University of Murcia, Regional Campus of International Excellence "Campus Mare Nostrum", 30100 Murcia, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
| | - Bruno Ramos-Molina
- Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
| | - Diego A Moreno
- Phytochemistry and Healthy Food Lab (LabFAS), Department of Food Science Technology, Centro de Edafología y Biología Aplicada del Segura (CEBAS-CSIC), Campus de Espinardo - 25, 30100 Murcia, Spain.
| |
Collapse
|
24
|
Co-encapsulation of broccoli sprout extract nanoliposomes into basil seed gum: effects on in vitro antioxidant, antibacterial and anti-Listeria activities in ricotta cheese. Int J Food Microbiol 2022; 376:109761. [DOI: 10.1016/j.ijfoodmicro.2022.109761] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 11/22/2022]
|
25
|
Hill CR, Shafaei A, Balmer L, Lewis JR, Hodgson JM, Millar AH, Blekkenhorst LC. Sulfur compounds: From plants to humans and their role in chronic disease prevention. Crit Rev Food Sci Nutr 2022; 63:8616-8638. [PMID: 35380479 DOI: 10.1080/10408398.2022.2057915] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sulfur is essential for the health of plants and is an indispensable dietary component for human health and disease prevention. Its incorporation into our food supply is heavily reliant upon the uptake of sulfur into plant tissue and our subsequent intake. Dietary requirements for sulfur are largely calculated based upon requirements for the sulfur-containing amino acids (SAA), cysteine and methionine, to meet the demands for synthesis of proteins, enzymes, co-enzymes, vitamins, and hormones. SAA are found in abundance in animal sources and are relatively low in plants. However, some plants, particularly cruciferous and allium vegetables, produce many protective sulfur-containing secondary metabolites, such as glucosinolates and cysteine sulfoxides. The variety and quantity of these sulfur-containing metabolites are extensive and their effects on human health are wide-reaching. Many benefits appear to be related to sulfur's role in redox biochemistry, protecting against uncontrolled oxidative stress and inflammation; features consistent within cardiometabolic dysfunction and many chronic metabolic diseases of aging. This narrative explores the origins and importance of sulfur, its incorporation into our food supply and dietary sources. It also explores the overarching potential of sulfur for human health, particularly around the amelioration of oxidative stress and chronic inflammation, and subsequent chronic disease prevention.
Collapse
Affiliation(s)
- Caroline R Hill
- Nutrition & Health Innovation Research Institute, School of Medical and Health Science, Edith Cowan University, Perth, Australia
- Royal Perth Hospital Research Foundation, Perth, Australia
| | - Armaghan Shafaei
- Centre for Integrative Metabolomics and Computational Biology, School of Science, Edith Cowan University, Joondalup, Australia
| | - Lois Balmer
- Centre for Precision Health, School of Medical and Health Science, Edith Cowan University, Perth, Australia
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, The University of Western Australia, Nedlands, Australia
| | - Joshua R Lewis
- Nutrition & Health Innovation Research Institute, School of Medical and Health Science, Edith Cowan University, Perth, Australia
- Royal Perth Hospital Research Foundation, Perth, Australia
- Medical School, The University of Western Australia, Nedlands, Australia
- Centre for Kidney Research, Children's Hospital at Westmead School of Public Health, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Jonathan M Hodgson
- Nutrition & Health Innovation Research Institute, School of Medical and Health Science, Edith Cowan University, Perth, Australia
- Royal Perth Hospital Research Foundation, Perth, Australia
- Medical School, The University of Western Australia, Nedlands, Australia
| | - A Harvey Millar
- Australian Research Council Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Lauren C Blekkenhorst
- Nutrition & Health Innovation Research Institute, School of Medical and Health Science, Edith Cowan University, Perth, Australia
- Royal Perth Hospital Research Foundation, Perth, Australia
- Medical School, The University of Western Australia, Nedlands, Australia
| |
Collapse
|
26
|
Ordonez AA, Bullen CK, Villabona-Rueda AF, Thompson EA, Turner ML, Merino VF, Yan Y, Kim J, Davis SL, Komm O, Powell JD, D'Alessio FR, Yolken RH, Jain SK, Jones-Brando L. Sulforaphane exhibits antiviral activity against pandemic SARS-CoV-2 and seasonal HCoV-OC43 coronaviruses in vitro and in mice. Commun Biol 2022; 5:242. [PMID: 35304580 PMCID: PMC8933402 DOI: 10.1038/s42003-022-03189-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 02/24/2022] [Indexed: 12/31/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), has incited a global health crisis. Currently, there are limited therapeutic options for the prevention and treatment of SARS-CoV-2 infections. We evaluated the antiviral activity of sulforaphane (SFN), the principal biologically active phytochemical derived from glucoraphanin, the naturally occurring precursor present in high concentrations in cruciferous vegetables. SFN inhibited in vitro replication of six strains of SARS-CoV-2, including Delta and Omicron, as well as that of the seasonal coronavirus HCoV-OC43. Further, SFN and remdesivir interacted synergistically to inhibit coronavirus infection in vitro. Prophylactic administration of SFN to K18-hACE2 mice prior to intranasal SARS-CoV-2 infection significantly decreased the viral load in the lungs and upper respiratory tract and reduced lung injury and pulmonary pathology compared to untreated infected mice. SFN treatment diminished immune cell activation in the lungs, including significantly lower recruitment of myeloid cells and a reduction in T cell activation and cytokine production. Our results suggest that SFN should be explored as a potential agent for the prevention or treatment of coronavirus infections.
Collapse
Affiliation(s)
- Alvaro A Ordonez
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - C Korin Bullen
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andres F Villabona-Rueda
- Division of Pulmonology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth A Thompson
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mitchell L Turner
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vanessa F Merino
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu Yan
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Kim
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephanie L Davis
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Oliver Komm
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan D Powell
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Franco R D'Alessio
- Division of Pulmonology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanjay K Jain
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lorraine Jones-Brando
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
27
|
García-Caballero M, Torres-Vargas JA, Marrero AD, Martínez-Poveda B, Medina MÁ, Quesada AR. Angioprevention of Urologic Cancers by Plant-Derived Foods. Pharmaceutics 2022; 14:pharmaceutics14020256. [PMID: 35213989 PMCID: PMC8875200 DOI: 10.3390/pharmaceutics14020256] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
The number of cancer cases worldwide keeps growing unstoppably, despite the undeniable advances achieved by basic research and clinical practice. Urologic tumors, including some as prevalent as prostate, bladder or kidney tumors, are no exceptions to this rule. Moreover, the fact that many of these tumors are detected in early stages lengthens the duration of their treatment, with a significant increase in health care costs. In this scenario, prevention offers the most cost-effective long-term strategy for the global control of these diseases. Although specialized diets are not the only way to decrease the chances to develop cancer, epidemiological evidence support the role of certain plant-derived foods in the prevention of urologic cancer. In many cases, these plants are rich in antiangiogenic phytochemicals, which could be responsible for their protective or angiopreventive properties. Angiogenesis inhibition may contribute to slow down the progression of the tumor at very different stages and, for this reason, angiopreventive strategies could be implemented at different levels of chemoprevention, depending on the targeted population. In this review, epidemiological evidence supporting the role of certain plant-derived foods in urologic cancer prevention are presented, with particular emphasis on their content in bioactive phytochemicals that could be used in the angioprevention of cancer.
Collapse
Affiliation(s)
- Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - José Antonio Torres-Vargas
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - Ana Dácil Marrero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - Beatriz Martínez-Poveda
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), E-28019 Madrid, Spain
| | - Miguel Ángel Medina
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Malaga, Spain
| | - Ana R. Quesada
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Malaga, Spain
- Correspondence:
| |
Collapse
|
28
|
Fahey JW, Kensler TW. Phytochemicals: Do They Belong on our Plate for Sustaining Healthspan? FOOD FRONTIERS 2021; 2:235-239. [PMID: 36818577 PMCID: PMC9937450 DOI: 10.1002/fft2.81] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Jed W. Fahey
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD 21205,Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205,Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205,Department of Nutrition and Food Studies, College of Health and Human Services, George Mason University, Fairfax, VA 22030,Correspondence: Jed W. Fahey, , Thomas W. Kensler,
| | - Thomas W. Kensler
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109,Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205,Correspondence: Jed W. Fahey, , Thomas W. Kensler,
| |
Collapse
|