1
|
Kakarla R, Karuturi P, Siakabinga Q, Kasi Viswanath M, Dumala N, Guntupalli C, Nalluri BN, Venkateswarlu K, Prasanna VS, Gutti G, Yadagiri G, Gujjari L. Current understanding and future directions of cruciferous vegetables and their phytochemicals to combat neurological diseases. Phytother Res 2024; 38:1381-1399. [PMID: 38217095 DOI: 10.1002/ptr.8122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/15/2023] [Accepted: 12/27/2023] [Indexed: 01/15/2024]
Abstract
Neurological disorders incidences are increasing drastically due to complex pathophysiology, and the nonavailability of disease-modifying agents. Several attempts have been made to identify new potential chemicals to combat these neurological abnormalities. At present, complete abolishment of neurological diseases is not attainable except for symptomatic relief. However, dietary recommendations to help brain development or improvement have increased over the years. In recent times, cruciferous vegetables and their phytochemicals have been identified from preclinical and clinical investigations as potential neuroprotective agents. The present review highlights the beneficial effects and molecular mechanisms of phytochemicals such as indole-3-carbinol, diindolylmethane, sulforaphane, kaempferol, selenium, lutein, zeaxanthin, and vitamins of cruciferous vegetables against neurological diseases including Parkinson's disease, Alzheimer's disease, stroke, Huntington's disease, autism spectra disorders, anxiety, depression, and pain. Most of these cruciferous phytochemicals protect the brain by eliciting antioxidant, anti-inflammatory, and antiapoptotic properties. Regular dietary intake of cruciferous vegetables may benefit the prevention and treatment of neurological diseases. The present review suggests that there is a lacuna in identifying the clinical efficacy of these phytochemicals. Therefore, high-quality future studies should firmly establish the efficacy of the above-mentioned cruciferous phytochemicals in clinical settings.
Collapse
Affiliation(s)
- Ramakrishna Kakarla
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Guntur, India
| | - Praditha Karuturi
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Guntur, India
| | - Queen Siakabinga
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Guntur, India
| | | | - Naresh Dumala
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Guntur, India
| | | | - Buchi N Nalluri
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Guntur, India
| | - Kojja Venkateswarlu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Varanasi, India
| | - Vani Sai Prasanna
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, India
| | - Gopichand Gutti
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Ganesh Yadagiri
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Lohitha Gujjari
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
2
|
Ang HL, Mohan CD, Shanmugam MK, Leong HC, Makvandi P, Rangappa KS, Bishayee A, Kumar AP, Sethi G. Mechanism of epithelial-mesenchymal transition in cancer and its regulation by natural compounds. Med Res Rev 2023. [PMID: 36929669 DOI: 10.1002/med.21948] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 12/19/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a complex process with a primordial role in cellular transformation whereby an epithelial cell transforms and acquires a mesenchymal phenotype. This transformation plays a pivotal role in tumor progression and self-renewal, and exacerbates resistance to apoptosis and chemotherapy. EMT can be initiated and promoted by deregulated oncogenic signaling pathways, hypoxia, and cells in the tumor microenvironment, resulting in a loss-of-epithelial cell polarity, cell-cell adhesion, and enhanced invasive/migratory properties. Numerous transcriptional regulators, such as Snail, Slug, Twist, and ZEB1/ZEB2 induce EMT through the downregulation of epithelial markers and gain-of-expression of the mesenchymal markers. Additionally, signaling cascades such as Wnt/β-catenin, Notch, Sonic hedgehog, nuclear factor kappa B, receptor tyrosine kinases, PI3K/AKT/mTOR, Hippo, and transforming growth factor-β pathways regulate EMT whereas they are often deregulated in cancers leading to aberrant EMT. Furthermore, noncoding RNAs, tumor-derived exosomes, and epigenetic alterations are also involved in the modulation of EMT. Therefore, the regulation of EMT is a vital strategy to control the aggressive metastatic characteristics of tumor cells. Despite the vast amount of preclinical data on EMT in cancer progression, there is a lack of clinical translation at the therapeutic level. In this review, we have discussed thoroughly the role of the aforementioned transcription factors, noncoding RNAs (microRNAs, long noncoding RNA, circular RNA), signaling pathways, epigenetic modifications, and tumor-derived exosomes in the regulation of EMT in cancers. We have also emphasized the contribution of EMT to drug resistance and possible therapeutic interventions using plant-derived natural products, their semi-synthetic derivatives, and nano-formulations that are described as promising EMT blockers.
Collapse
Affiliation(s)
- Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hin Chong Leong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia Centre for Materials Interface, Pontedera, Pisa, Italy
| | | | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
3
|
Tucci P, Brown I, Bewick GS, Pertwee RG, Marini P. The Plant Derived 3-3'-Diindolylmethane (DIM) Behaves as CB 2 Receptor Agonist in Prostate Cancer Cellular Models. Int J Mol Sci 2023; 24:ijms24043620. [PMID: 36835033 PMCID: PMC9962283 DOI: 10.3390/ijms24043620] [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: 11/28/2022] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
3-3'-Diindolylmethane (DIM) is a biologically active dimer derived from the endogenous conversion of indole-3-carbinol (I3C), a naturally occurring glucosinolate found in many cruciferous vegetables (i.e., Brassicaceae). DIM was the first pure androgen receptor antagonist isolated from the Brassicaceae family and has been recently investigated for its potential pharmacological use in prostate cancer prevention and treatment. Interestingly, there is evidence that DIM can also interact with cannabinoid receptors. In this context, by considering the well-known involvement of the endocannabinoid system in prostate cancer, we have pharmacologically characterized the properties of DIM on both CB1 and CB2 cannabinoid receptors in two human prostate cancer cell lines: PC3 (androgen-independent/androgen receptor negative) and LNCaP (androgen-dependent). In the PC3 cell line, DIM was able to activate CB2 receptors and potentially associated apoptotic pathways. On the other hand, although DIM was also able to activate CB2 receptors in the LNCaP cell line, no apoptotic effects were observed. Our evidence confirms that DIM is a CB2 receptor ligand and, moreover, it has a potential anti-proliferative effect on androgen-independent/androgen receptor-negative prostate cancer cells.
Collapse
Affiliation(s)
- Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
- Correspondence:
| | - Iain Brown
- Division of Applied Medicine, School of Medicine and Dentistry, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Guy S. Bewick
- The Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Roger G. Pertwee
- The Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Pietro Marini
- Institute of Education in Healthcare and Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| |
Collapse
|
4
|
Williams DE. Indoles Derived From Glucobrassicin: Cancer Chemoprevention by Indole-3-Carbinol and 3,3'-Diindolylmethane. Front Nutr 2021; 8:734334. [PMID: 34660663 PMCID: PMC8517077 DOI: 10.3389/fnut.2021.734334] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/27/2021] [Indexed: 12/22/2022] Open
Abstract
Hydrolysis of glucobrassicin by plant or bacterial myrosinase produces multiple indoles predominantly indole-3-carbinol (I3C). I3C and its major in vivo product, 3,3'-diindolylmethane (DIM), are effective cancer chemopreventive agents in pre-clinical models and show promise in clinical trials. The pharmacokinetics/pharmacodynamics of DIM have been studied in both rodents and humans and urinary DIM is a proposed biomarker of dietary intake of cruciferous vegetables. Recent clinical studies at Oregon State University show surprisingly robust metabolism of DIM in vivo with mono- and di-hydroxylation followed by conjugation with sulfate or glucuronic acid. DIM has multiple mechanisms of action, the most well-characterized is modulation of aryl hydrocarbon receptor (AHR) signaling. In rainbow trout dose-dependent cancer chemoprevention by dietary I3C is achieved when given prior to or concurrent with aflatoxin B1, polycyclic aromatic hydrocarbons, nitrosamines or direct acting carcinogens such as N-methyl-N'-nitro-nitrosoguanidine. Feeding pregnant mice I3C inhibits transplacental carcinogenesis. In humans much of the focus has been on chemoprevention of breast and prostate cancer. Alteration of cytochrome P450-dependent estrogen metabolism is hypothesized to be an important driver of DIM-dependent breast cancer prevention. The few studies done to date comparing glucobrassicin-rich crucifers such as Brussels sprouts with I3C/DIM supplements have shown the greater impact of the latter is due to dose. Daily ingestion of kg quantities of Brussels sprouts is required to produce in vivo levels of DIM achievable by supplementation. In clinical trials these supplement doses have elicited few if any adverse effects. Sulforaphane from glucoraphanin can act synergistically with glucobrassicin-derived DIM and this may lead to opportunities for combinatorial approaches (supplement and food-based) in the clinic.
Collapse
Affiliation(s)
- David E. Williams
- Department of Environmental and Molecular Toxicology, Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
5
|
Vermillion Maier ML, Siddens LK, Uesugi SL, Choi J, Leonard SW, Pennington JM, Tilton SC, Smith JN, Ho E, Chow HHS, Nguyen BD, Kolluri SK, Williams DE. 3,3'-Diindolylmethane Exhibits Significant Metabolism after Oral Dosing in Humans. Drug Metab Dispos 2021; 49:694-705. [PMID: 34035125 PMCID: PMC8407664 DOI: 10.1124/dmd.120.000346] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 05/07/2021] [Indexed: 01/07/2023] Open
Abstract
3,3'-Diindolylmethane (DIM), a major phytochemical derived from ingestion of cruciferous vegetables, is also a dietary supplement. In preclinical models, DIM is an effective cancer chemopreventive agent and has been studied in a number of clinical trials. Previous pharmacokinetic studies in preclinical and clinical models have not reported DIM metabolites in plasma or urine after oral dosing, and the pharmacological actions of DIM on target tissues is assumed to be solely via the parent compound. Seven subjects (6 males and 1 female) ranging from 26-65 years of age, on a cruciferous vegetable-restricted diet prior to and during the study, took 2 BioResponse DIM 150-mg capsules (45.3 mg DIM/capsule) every evening for one week with a final dose the morning of the first blood draw. A complete time course was performed with plasma and urine collected over 48 hours and analyzed by UPLC-MS/MS. In addition to parent DIM, two monohydroxylated metabolites and 1 dihydroxylated metabolite, along with their sulfate and glucuronide conjugates, were present in both plasma and urine. Results reported here are indicative of significant phase 1 and phase 2 metabolism and differ from previous pharmacokinetic studies in rodents and humans, which reported only parent DIM present after oral administration. 3-((1H-indole-3-yl)methyl)indolin-2-one, identified as one of the monohydroxylated products, exhibited greater potency and efficacy as an aryl hydrocarbon receptor agonist when tested in a xenobiotic response element-luciferase reporter assay using Hepa1 cells. In addition to competitive phytochemical-drug adverse reactions, additional metabolites may exhibit pharmacological activity highlighting the importance of further characterization of DIM metabolism in humans. SIGNIFICANCE STATEMENT: 3,3'-Diindolylmethane (DIM), derived from indole-3-carbinol in cruciferous vegetables, is an effective cancer chemopreventive agent in preclinical models and a popular dietary supplement currently in clinical trials. Pharmacokinetic studies to date have found little or no metabolites of DIM in plasma or urine. In marked contrast, we demonstrate rapid appearance of mono- and dihydroxylated metabolites in human plasma and urine as well as their sulfate and glucuronide conjugates. The 3-((1H-indole-3-yl)methyl)indolin-2-one metabolite exhibited significant aryl hydrocarbon receptor agonist activity, emphasizing the need for further characterization of the pharmacological properties of DIM metabolites.
Collapse
Affiliation(s)
- Monica L Vermillion Maier
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - Lisbeth K Siddens
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - Sandra L Uesugi
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - Jaewoo Choi
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - Scott W Leonard
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - Jamie M Pennington
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - Susan C Tilton
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - Jordan N Smith
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - Emily Ho
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - H H Sherry Chow
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - Bach D Nguyen
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - Siva K Kolluri
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| | - David E Williams
- Department of Environmental and Molecular Toxicology (M.L.V.M., L.K.S., S.C.T., B.D.N., S.K.K., D.E.W.), the Linus Pauling Institute (M.L.V.M., S.L.U., J.C., S.W.L., J.M.P., E.H., D.E.W.), School of Biological and Population Health Sciences (E.H.), Oregon State University, Corvallis, OR; Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, Richland, WA (J.N.S.); and Cancer Center, University of Arizona, Tucson, AZ (H.H.S.C.)
| |
Collapse
|
6
|
Amare DE. Anti-Cancer and Other Biological Effects of a Dietary Compound 3,3ʹ-Diindolylmethane Supplementation: A Systematic Review of Human Clinical Trials . NUTRITION AND DIETARY SUPPLEMENTS 2020. [DOI: 10.2147/nds.s261577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
7
|
Giacomini A, Grillo E, Rezzola S, Ribatti D, Rusnati M, Ronca R, Presta M. The FGF/FGFR system in the physiopathology of the prostate gland. Physiol Rev 2020; 101:569-610. [PMID: 32730114 DOI: 10.1152/physrev.00005.2020] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factors (FGFs) are a family of proteins possessing paracrine, autocrine, or endocrine functions in a variety of biological processes, including embryonic development, angiogenesis, tissue homeostasis, wound repair, and cancer. Canonical FGFs bind and activate tyrosine kinase FGF receptors (FGFRs), triggering intracellular signaling cascades that mediate their biological activity. Experimental evidence indicates that FGFs play a complex role in the physiopathology of the prostate gland that ranges from essential functions during embryonic development to modulation of neoplastic transformation. The use of ligand- and receptor-deleted mouse models has highlighted the requirement for FGF signaling in the normal development of the prostate gland. In adult prostate, the maintenance of a functional FGF/FGFR signaling axis is critical for organ homeostasis and function, as its disruption leads to prostate hyperplasia and may contribute to cancer progression and metastatic dissemination. Dissection of the molecular landscape modulated by the FGF family will facilitate ongoing translational efforts directed toward prostate cancer therapy.
Collapse
Affiliation(s)
- Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Domenico Ribatti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Marco Rusnati
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; and Italian Consortium for Biotechnology, Unit of Brescia, Brescia, Italy
| |
Collapse
|
8
|
Quirante-Moya S, García-Ibañez P, Quirante-Moya F, Villaño D, Moreno DA. The Role of Brassica Bioactives on Human Health: Are We Studying It the Right Way? Molecules 2020; 25:E1591. [PMID: 32235638 PMCID: PMC7180841 DOI: 10.3390/molecules25071591] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 12/11/2022] Open
Abstract
Brassica vegetables and their components, the glucosinolates, have been suggested as good candidates as dietary coadjutants to improve health in non-communicable diseases (NCDs). Different preclinical and clinical studies have been performed in the last decade; however, some concerns have been posed on the lack of established and standardized protocols. The different concentration of bioactive compounds used, time of intervention or sample size, and the lack of blinding are some factors that may influence the studies' outcomes. This review aims to analyze the critical points of the studies performed with Brassica-related biomolecules and propose some bases for future trials in order to avoid biases.
Collapse
Affiliation(s)
- Sarai Quirante-Moya
- Centro de Salud Callosa del Segura, Paseo Enrique Tierno Galvan, 19, E-03360 Alicante, Spain;
| | - Paula García-Ibañez
- CEBAS-CSIC, Department of Plant Nutrition. Aquaporins Group, Campus Universitario de Espinardo-25, E-30100 Murcia, Spain; (P.G.-I.); (F.Q.-M.)
- CEBAS-CSIC, Department of Food Science and Technology, Phytochemistry and Healthy Foods Lab, Research Group on Quality, Safety and Bioactivity of Plant Foods, Campus de Espinardo-25, E-30100 Espinardo, Murcia, Spain
| | - Francisco Quirante-Moya
- CEBAS-CSIC, Department of Plant Nutrition. Aquaporins Group, Campus Universitario de Espinardo-25, E-30100 Murcia, Spain; (P.G.-I.); (F.Q.-M.)
| | - Débora Villaño
- Faculty of Health Sciences, Department of Pharmacy, Universidad Católica de San Antonio de Murcia (UCAM), Campus de los Jerónimos, E-30107 Guadalupe, Murcia, Spain;
| | - Diego A. Moreno
- CEBAS-CSIC, Department of Food Science and Technology, Phytochemistry and Healthy Foods Lab, Research Group on Quality, Safety and Bioactivity of Plant Foods, Campus de Espinardo-25, E-30100 Espinardo, Murcia, Spain
| |
Collapse
|
9
|
Preparation and Characterization of Whey Protein Isolate-DIM Nanoparticles. Int J Mol Sci 2019; 20:ijms20163917. [PMID: 31408980 PMCID: PMC6721066 DOI: 10.3390/ijms20163917] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022] Open
Abstract
3,3’-Diindolylmethane (DIM) is a bioactive compound found in Cruciferous vegetables that possesses health benefits such as antioxidant, anticancer, and anti-inflammatory effects. However, hydrophobicity and photolabile limit its pharmaceutical applications. This study aims to prepare and characterize DIM-encapsulated whey protein isolate (WPI) nanoparticles mixed at different ratios of WPI and DIM using the combined heating–ultrasound method. Results showed that all the samples showed adequate physicochemical characteristics: The mean particle size of the nanoparticles could be controlled down to 96–157 nm depending on the DIM to WPI ratio used in the preparation with a low polydispersity index (<0.5), higher negative values of zeta potential (>−40 mV) as well as with greater encapsulation efficiency (>82%). Flow behavior indices showed the shear-thinning Non-Newtonian or pseudoplastic (n < 1) behavior of the nanoparticles. The thermal properties were characterized by differential scanning calorimetry (DSC), which showed that DIM was successfully entrapped in WPI nanoparticles. The secondary structure of WPI was changed after DIM incorporation; electrostatic interaction and hydrogen bonding were major facilitating forces for nanoparticles formation, confirmed by Fourier Transform Infrared Spectroscopy (FT-IR). Transmission electron microscopy (TEM) micrographs showed that all the samples had a smooth surface and spherical structure. The wall material (WPI) and encapsulation method provide effective protection to DIM against UV light and a broad range of physiologically relevant pH’s (2.5, 3.5, 4.5, 5.5, and 7). In conclusion, whey protein isolate (WPI)-based nanoparticles are a promising approach to encapsulate DIM and overcome its physicochemical limitations with improved stability.
Collapse
|
10
|
Cui K, Li X, Du Y, Tang X, Arai S, Geng Y, Xi Y, Xu H, Zhou Y, Ma W, Zhang T. Chemoprevention of prostate cancer in men with high-grade prostatic intraepithelial neoplasia (HGPIN): a systematic review and adjusted indirect treatment comparison. Oncotarget 2018; 8:36674-36684. [PMID: 28415774 PMCID: PMC5482687 DOI: 10.18632/oncotarget.16230] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 03/06/2017] [Indexed: 12/26/2022] Open
Abstract
Background High-grade prostatic intraepithelial neoplasia (HGPIN) is the precursor or premalignant form of prostate cancer. At least 30% patients with a confirmed HGPIN will develop prostate cancer within 1 year after repeated biopsy. HGPIN patients are the appropriate at-risk population for chemoprevention strategies investigation against prostate cancer. However the commonly used chemoprevention agents that targeted on hormonal imbalance or lifestyle-related factors showed varied results in HGPIN patients. Methods Literature searches were conducted in PubMed, EMBASE and Cochrane library according to Cochrane guidelines before January 31st, 2017. Direct meta-analysis were performed to summarize the efficacy of candidate chemopreventative agents Dutasteride, Flutamide, Toremifene, Selenium, Green tea components, Lycopene and natural food products combination. Adjusted indirect meta-analyses were employed to compare the relative efficacy of these candidate chemoprevention agents head-to-head. Results The overall incidence of prostate cancer in HGPIN was slightly decreased by chemoprevention agents (25.7% vs 31.5%, RR = 0.92, 95% CI: 0.83-1.03, P = 0.183), with minor heterogeneity (I2 = 22.3%, χ2 = 15.08, P = 0.237), but without statistical significance. Subgroup analysis showed that green tea catechins significantly decreased prostate cancer in HGPIN patients (7.60% vs 23.1%, RR = 0.39, 95% CI: 0.16-10.97, P P = 0.044), with moderate heterogeneity (I2 = 47.9%, χ2 = 1.92, P = 0.166). The adjusted indirect meta-analysis favored green tea catechins over other chemoprevention agents, and significantly when compared to natural food products combination (RR = 0.355, 95% CI: 0.134-0.934). Conclusion The overall efficacy of chemoprevention agents in HGPIN patients is limited. But Green tea catechins showed the superiority to decrease prostate cancer in HGPIN patients.
Collapse
Affiliation(s)
- Kang Cui
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiangnan Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yabing Du
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiance Tang
- Department of Medical Affairs, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Seiji Arai
- Department of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States.,Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yiwei Geng
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Xi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Han Xu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yue Zhou
- Deparmtent of B-Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wang Ma
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tengfei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
11
|
Abstract
CONTEXT - Precursor lesions of urologic malignancies are established histopathologic entities, which are important not only to recognize for clinical purposes, but also to further investigate at the molecular level in order to gain a better understanding of the pathogenesis of these malignancies. OBJECTIVE - To provide a brief overview of precursor lesions to the most common malignancies that develop within the genitourinary tract with a focus on their clinical implications, histologic features, and molecular characteristics. DATA SOURCES - Literature review from PubMed, urologic pathology textbooks, and the 4th edition of the World Health Organization Classification of Tumours of the Urinary System and Male Genital Organs. All photomicrographs were taken from cases seen at Weill Cornell Medicine or from the authors' personal slide collections. CONCLUSIONS - The clinical importance and histologic criteria are well established for the known precursor lesions of the most common malignancies throughout the genitourinary tract, but further investigation is warranted at the molecular level to better understand the pathogenesis of these lesions. Such investigation may lead to better risk stratification of patients and potentially novel treatments.
Collapse
|
12
|
Xie VK, Li Z, Yan Y, Jia Z, Zuo X, Ju Z, Wang J, Du J, Xie D, Xie K, Wei D. DNA-Methyltransferase 1 Induces Dedifferentiation of Pancreatic Cancer Cells through Silencing of Krüppel-Like Factor 4 Expression. Clin Cancer Res 2017; 23:5585-5597. [PMID: 28659310 DOI: 10.1158/1078-0432.ccr-17-0387] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/19/2017] [Accepted: 06/19/2017] [Indexed: 01/22/2023]
Abstract
Purpose: The dismal prognosis of pancreatic cancer has been linked to poor tumor differentiation. However, molecular basis of pancreatic cancer differentiation and potential therapeutic value of the underlying molecules remain unknown. We investigated the mechanistic underexpression of Krüppel-like factor 4 (KLF4) in pancreatic cancer and defined a novel epigenetic pathway of its activation for pancreatic cancer differentiation and treatment.Experimental Design: Expressions of KLF4 and DNMT1 in pancreatic cancer tissues were determined by IHC and the genetic and epigenetic alterations of KLF4 in and KLF4's impact on differentiation of pancreatic cancer were examined using molecular biology techniques. The function of dietary 3,3'-diindolylmethane (DIM) on miR-152/DNMT1/KLF4 signaling in pancreatic cancer was evaluated using both cell culture and animal models.Results: Overexpression of DNMT1 and promoter hypermethylation contributed to decreased KLF4 expression in and associated with poor differentiation of pancreatic cancer. Manipulation of KLF4 expression significantly affected differentiation marker expressions in pancreatic cancer cells. DIM treatment significantly induced miR-152 expression, which blocked DNMT1 protein expression and its binding to KLF4 promoter region, and consequently reduced promoter DNA methylation and activated KLF4 expression in pancreatic cancer cells. In addition, DIM treatment caused significant inhibition of cell growth in vitro and tumorigenesis in animal models of pancreatic cancer.Conclusions: This is the first demonstration that dysregulated KLF4 expression associates with poor differentiation of pancreatic cancer. Epigenetic activation of miR-152/DNMT1/KLF4 signaling pathway by dietary DIM causes differentiation and significant growth inhibition of pancreatic cancer cells, highlighting its translational implications for pancreatic and other cancers. Clin Cancer Res; 23(18); 5585-97. ©2017 AACR.
Collapse
Affiliation(s)
- Victoria K Xie
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhiwei Li
- Department Gastroenterology Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yongmin Yan
- Department Gastroenterology Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhiliang Jia
- Department Gastroenterology Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiangsheng Zuo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiawei Du
- Department of Oncology, Shanghai Tongji University East Hospital, Shanghai, P.R. China
| | - Dacheng Xie
- Department of Oncology, Shanghai Tongji University East Hospital, Shanghai, P.R. China
| | - Keping Xie
- Department Gastroenterology Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Daoyan Wei
- Department Gastroenterology Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|