Naringin commonly acts via hormesis.
暂无分享,去创建一个
E. Calabrese | A. Hayes | V. Calabrese | E. Agathokleous | R. Kapoor | G. Dhawan | E. Agathokleous | P. Manes | Peter Pressman | Evgenios Agathokleous
[1] E. Calabrese,et al. Lithium and hormesis: Enhancement of adaptive responses and biological performance via hormetic mechanisms. , 2023, Journal of trace elements in medicine and biology : organ of the Society for Minerals and Trace Elements.
[2] E. Calabrese,et al. Boron enhances adaptive responses and biological performance via hormetic mechanisms. , 2023, Chemico-biological interactions.
[3] Ning Zhang,et al. Naringin Protects against Tau Hyperphosphorylation in Aβ25–35-Injured PC12 Cells through Modulation of ER, PI3K/AKT, and GSK-3β Signaling Pathways , 2023, Behavioural neurology.
[4] Zengli Yu,et al. Naringin Alleviates Glucose-Induced Aging by Reducing Fat Accumulation and Promoting Autophagy in Caenorhabditis elegans , 2023, Nutrients.
[5] Qianzhou Jiang,et al. The role of PIWI-interacting RNA in naringin pro-angiogenesis by targeting HUVECs. , 2023, Chemico-biological interactions.
[6] T. Saleh,et al. VANL-100 Attenuates Beta-Amyloid-Induced Toxicity in SH-SY5Y Cells , 2022, International journal of molecular sciences.
[7] E. Calabrese,et al. Hormesis and Epidermal Stem Cells , 2022, Dose-response : a publication of International Hormesis Society.
[8] Lei Wang,et al. Hormesis in Caenorhabditis elegans exposed to pollutants , 2022, Current Opinion in Environmental Science & Health.
[9] Yuehua Jiang,et al. Naringin protects human nucleus pulposus cells against TNF-α-induced inflammation, oxidative stress, and loss of cellular homeostasis by enhancing autophagic flux via AMPK/SIRT1 activation , 2022, Oxidative medicine and cellular longevity.
[10] Zhenzhen Yuan,et al. The Effect of Naringin on the Apoptosis of Degenerative Nucleus Pulposus Cells: A Study on the Function and Mechanism , 2022, Drug Design, Development and Therapy.
[11] E. Cacan,et al. A collective analysis of lifespan-extending compounds in diverse model organisms, and of species whose lifespan can be extended the most by the application of compounds , 2021, Biogerontology.
[12] E. Calabrese,et al. Luteolin and hormesis , 2021, Mechanisms of Ageing and Development.
[13] E. Calabrese,et al. Ferulic acid and hormesis: Biomedical and environmental implications , 2021, Mechanisms of Ageing and Development.
[14] Yang Zeng,et al. Naringenin promotes SDF-1/CXCR4 signaling pathway in BMSCs osteogenic differentiation. , 2021, Folia histochemica et cytobiologica.
[15] Yansen Xu,et al. Nonlinear responses of foliar phenylpropanoids to increasing O3 exposure: Ecological implications in a Populus model system. , 2020, The Science of the total environment.
[16] J. Shang. Naringenin protects cerebral ischemic injury by inhibiting mitochondria-mediated neuronal apoptosis in acute ischemic stroke rats , 2020, Acta Poloniae Pharmaceutica - Drug Research.
[17] Xiao-Gang Zhou,et al. A Dihydroflavonoid Naringin Extends the Lifespan of C. elegans and Delays the Progression of Aging-Related Diseases in PD/AD Models via DAF-16 , 2020, Oxidative medicine and cellular longevity.
[18] A. Tsatsakis,et al. Does Green Tea Induce Hormesis? , 2020, Dose-response : a publication of International Hormesis Society.
[19] E. Calabrese. Hormesis and Ginseng: Ginseng Mixtures and Individual Constituents Commonly Display Hormesis Dose Responses, Especially for Neuroprotective Effects , 2020, Molecules.
[20] A. Tsatsakis,et al. Hormesis and Ginkgo biloba (GB): Numerous biological effects of GB are mediated via hormesis , 2020, Ageing Research Reviews.
[21] Di Ran,et al. Naringin alleviates H2O2-induced apoptosis via the PI3K/Akt pathway in rat nucleus pulposus-derived mesenchymal stem cells , 2019, Connective tissue research.
[22] M. Mattson,et al. Curcumin and hormesis with particular emphasis on neural cells. , 2019, Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association.
[23] Sridevi Muruhan,et al. Naringin prevents ultraviolet‐B radiation‐induced oxidative damage and inflammation through activation of peroxisome proliferator‐activated receptor γ in mouse embryonic fibroblast (NIH‐3T3) cells , 2018, Journal of biochemical and molecular toxicology.
[24] Yu Cao,et al. Effects of Naringin on the Proliferation, Migration and Osteogenesis of Two Human Osteoblast-Like Cell Lines , 2018, Journal of Biomaterials and Tissue Engineering.
[25] Jian-xiong Ma,et al. Naringin enhances endothelial progenitor cell (EPC) proliferation and tube formation capacity through the CXCL12/CXCR4/PI3K/Akt signaling pathway. , 2018, Chemico-biological interactions.
[26] Alex Zhavoronkov,et al. The Evaluation of Geroprotective Effects of Selected Flavonoids in Drosophila melanogaster and Caenorhabditis elegans , 2017, Front. Pharmacol..
[27] Hanbin Ouyang,et al. Naringin protects myocardial cells from doxorubicin-induced apoptosis partially by inhibiting the p38MAPK pathway , 2017, Molecular medicine reports.
[28] T. Saleh,et al. A co‐drug conjugate of naringenin and lipoic acid mediates neuroprotection in a rat model of oxidative stress , 2017, Clinical and experimental pharmacology & physiology.
[29] Shubhankar Das,et al. Naringin abates adverse effects of cadmium‐mediated hepatotoxicity: An experimental study using HepG2 cells , 2017, Journal of biochemical and molecular toxicology.
[30] Shangtian Yang,et al. Effects of naringin on the proliferation and osteogenic differentiation of human amniotic fluid‐derived stem cells , 2017, Journal of tissue engineering and regenerative medicine.
[31] Xue-Dong Li,et al. Naringin promotes osteogenic differentiation of bone marrow stromal cells by up-regulating Foxc2 expression via the IHH signaling pathway. , 2016, American journal of translational research.
[32] Nianhu Li,et al. Therapeutic effects of naringin on degenerative human nucleus pulposus cells for discogenic low back pain. , 2016, The spine journal : official journal of the North American Spine Society.
[33] E. Calabrese. Preconditioning is hormesis part I: Documentation, dose-response features and mechanistic foundations. , 2016, Pharmacological Research.
[34] E. Calabrese. Preconditioning is hormesis part II: How the conditioning dose mediates protection: Dose optimization within temporal and mechanistic frameworks. , 2016, Pharmacological research.
[35] H. Wu,et al. Protective effects of naringin against gp120-induced injury mediated by P2X7 receptors in BV2 microglial cells. , 2016, Genetics and molecular research : GMR.
[36] K. Thirumurugan,et al. Context- and dose-dependent modulatory effects of naringenin on survival and development of Drosophilamelanogaster , 2016, Biogerontology.
[37] Xue-Dong Li,et al. Naringin Stimulates Osteogenic Differentiation of Rat Bone Marrow Stromal Cells via Activation of the Notch Signaling Pathway , 2016, Stem cells international.
[38] S. Ganapasam,et al. Neuroprotective efficacy of naringin on 3-nitropropionic acid-induced mitochondrial dysfunction through the modulation of Nrf2 signaling pathway in PC12 cells , 2015, Molecular and Cellular Biochemistry.
[39] M. Barbosa,et al. Inflammation in intervertebral disc degeneration and regeneration , 2015, Journal of The Royal Society Interface.
[40] Bo Sun,et al. Stimulation of Wnt/β-Catenin Signaling to Improve Bone Development by Naringin via Interacting with AMPK and Akt , 2015, Cellular Physiology and Biochemistry.
[41] Wei Zhang,et al. Effects of Naringin on Proliferation and Osteogenic Differentiation of Human Periodontal Ligament Stem Cells In Vitro and In Vivo , 2015, Stem cells international.
[42] E. Calabrese. Dose–Response: A Fundamental Concept in Toxicology , 2014 .
[43] A. Kandhare,et al. Naringin, a flavanone glycoside, promotes angiogenesis and inhibits endothelial apoptosis through modulation of inflammatory and growth factor expression in diabetic foot ulcer in rats. , 2014, Chemico-biological interactions.
[44] G. Gutiérrez‐Venegas,et al. Flavonoids inhibit iNOS production via mitogen activated proteins in lipoteichoic acid stimulated cardiomyoblasts. , 2014, International immunopharmacology.
[45] S. Sarker,et al. Effect of citrus flavonoids, naringin and naringenin, on metabolic syndrome and their mechanisms of action. , 2014, Advances in nutrition.
[46] Runmin Guo,et al. Naringin Inhibits ROS‐activated MAPK Pathway in High Glucose‐induced Injuries in H9c2 Cardiac Cells , 2014, Basic & clinical pharmacology & toxicology.
[47] E. Calabrese. Hormetic mechanisms , 2013, Critical reviews in toxicology.
[48] P. Wooley,et al. Naringin promotes osteoblast differentiation and effectively reverses ovariectomy-associated osteoporosis , 2013, Journal of orthopaedic science : official journal of the Japanese Orthopaedic Association.
[49] F. Boege,et al. Adaptive and maladaptive responses in skin: mild heat exposure protects against UVB-induced photoaging in mice. , 2013, The Journal of investigative dermatology.
[50] Q. Ma. Role of nrf2 in oxidative stress and toxicity. , 2013, Annual review of pharmacology and toxicology.
[51] G. Sudhandiran,et al. Naringin modulates oxidative stress and inflammation in 3-nitropropionic acid-induced neurodegeneration through the activation of nuclear factor-erythroid 2-related factor-2 signalling pathway , 2012, Neuroscience.
[52] M. Nagata,et al. Curcumin prevents diabetic cardiomyopathy in streptozotocin-induced diabetic rats: possible involvement of PKC-MAPK signaling pathway. , 2012, European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences.
[53] J. Giddings,et al. Preventive Effects of Hesperidin, Glucosyl Hesperidin and Naringin on Hypertension and Cerebral Thrombosis in Stroke‐prone Spontaneously Hypertensive Rats , 2012, Phytotherapy research : PTR.
[54] A. Palmeri,et al. Hormetic effect of amyloid-beta peptide in synaptic plasticity and memory , 2012, Neurobiology of Aging.
[55] Edward J Calabrese,et al. Toxicology rewrites its history and rethinks its future: Giving equal focus to both harmful and beneficial effects , 2011, Environmental toxicology and chemistry.
[56] Edward J. Calabrese,et al. The hormesis database: the occurrence of hormetic dose responses in the toxicological literature. , 2011, Regulatory toxicology and pharmacology : RTP.
[57] G. Cai,et al. Comparison of neoeriocitrin and naringin on proliferation and osteogenic differentiation in MC3T3-E1. , 2011, Phytomedicine : international journal of phytotherapy and phytopharmacology.
[58] A. Fenning,et al. High-carbohydrate, High-fat Diet–induced Metabolic Syndrome and Cardiovascular Remodeling in Rats , 2011, Journal of cardiovascular pharmacology.
[59] M. Mattson,et al. Hormesis provides a generalized quantitative estimate of biological plasticity , 2011, Journal of Cell Communication and Signaling.
[60] L. Pari,et al. Hepatoprotective role of naringin on nickel-induced toxicity in male Wistar rats. , 2011, European journal of pharmacology.
[61] M. Mattson,et al. Resveratrol commonly displays hormesis: Occurrence and biomedical significance , 2010, Human & experimental toxicology.
[62] S. Prabu,et al. Cadmium-induced hepatotoxicity in rats and the protective effect of naringenin. , 2010, Experimental and toxicologic pathology : official journal of the Gesellschaft fur Toxikologische Pathologie.
[63] L Bravo,et al. Polyphenols: chemistry, dietary sources, metabolism, and nutritional significance. , 2009, Nutrition reviews.
[64] Shi-gui Yan,et al. Effects of naringin on the proliferation and osteogenic differentiation of human bone mesenchymal stem cell. , 2009, European journal of pharmacology.
[65] Kumar Ganesan,et al. Effect of Helicteres isora bark extracts on heart antioxidant status and lipid peroxidation in streptozotocin diabetic rats , 2008 .
[66] H. Tsai,et al. Naringin-induced bone morphogenetic protein-2 expression via PI3K, Akt, c-Fos/c-Jun and AP-1 pathway in osteoblasts. , 2008, European journal of pharmacology.
[67] Edward J. Calabrese,et al. Hormesis: Why it is important to toxicology and toxicologists , 2008 .
[68] E. Calabrese. Hormesis and mixtures. , 2008, Toxicology and applied pharmacology.
[69] F. Bauss,et al. Preclinical evidence for nitrogen-containing bisphosphonate inhibition of farnesyl diphosphate (FPP) synthase in the kidney: implications for renal safety. , 2008, Toxicology in vitro : an international journal published in association with BIBRA.
[70] Spencer J. Williams,et al. Understanding the cardioprotective effects of flavonols: discovery of relaxant flavonols without antioxidant activity. , 2008, Journal of medicinal chemistry.
[71] P. Stanely Mainzen Prince,et al. Preventive effect of naringin on cardiac mitochondrial enzymes during isoproterenol‐induced myocardial infarction in rats: A transmission electron microscopic study , 2007, Journal of biochemical and molecular toxicology.
[72] P. Stanely Mainzen Prince,et al. Preventive effect of naringin on lipid peroxides and antioxidants in isoproterenol-induced cardiotoxicity in Wistar rats: biochemical and histopathological evidences. , 2006, Toxicology.
[73] E. Calabrese,et al. The occurrence of hormetic dose responses in the toxicological literature, the hormesis database: an overview. , 2005, Toxicology and applied pharmacology.
[74] K. Jeong,et al. The hypoglycemic effects of hesperidin and naringin are partly mediated by hepatic glucose-regulating enzymes in C57BL/KsJ-db/db mice. , 2004, The Journal of nutrition.
[75] E. Calabrese,et al. Inorganics and Hormesis , 2003, Critical reviews in toxicology.
[76] E J Calabrese,et al. Defining hormesis , 2002, Human & experimental toxicology.
[77] Young‐Bae Park,et al. Naringin Has an Antiatherogenic Effect With the Inhibition of Intercellular Adhesion Molecule-1 in Hypercholesterolemic Rabbits , 2001, Journal of cardiovascular pharmacology.
[78] E. Calabrese,et al. Radiation hormesis: the demise of a legitimate hypothesis , 2000, Human & experimental toxicology.
[79] E J Calabrese,et al. Radiation hormesis: its historical foundations as a biological hypothesis , 2000, Human & experimental toxicology.
[80] E J Calabrese,et al. Chemical hormesis: its historical foundations as a biological hypothesis , 2000, Human & experimental toxicology.
[81] E. Calabrese,et al. Tales of two similar hypotheses: the rise and fall of chemical and radiation hormesis , 2000, Human & experimental toxicology.
[82] E J Calabrese,et al. The marginalization of hormesis , 2000, Toxicologic pathology.
[83] C. Rice-Evans,et al. Interactions of the flavonoid naringenin in the gastrointestinal tract and the influence of glycosylation. , 1999, Biochemical and biophysical research communications.
[84] Y. Kumazawa,et al. Suppression of lipopolysaccharide-induced tumor necrosis factor-release and liver injury in mice by naringin. , 1999, European journal of pharmacology.
[85] E. Calabrese. Evidence that hormesis represents an "overcompensation" response to a disruption in homeostasis. , 1999, Ecotoxicology and environmental safety.
[86] Johanna T. Dwyer,et al. Flavonoids: Dietary occurrence and biochemical activity , 1998 .
[87] K. Croft. The Chemistry and Biological Effects of Flavonoids and Phenolic Acids a , 1998, Annals of the New York Academy of Sciences.
[88] N. Bogduk,et al. The Prevalence and Clinical Features of Internal Disc Disruption in Patients With Chronic Low Back Pain , 1995, Spine.
[89] S. Cui,et al. Naringenin prolongs lifespan and delays aging mediated by IIS and MAPK in Caenorhabditis elegans , 2021, Food & Function.
[90] M. Prasanth,et al. Analyzing the Synergistic Effects of Antioxidants in Combating Photoaging Using Model Nematode, Caenorhabditis elegans , 2019, Photochemistry and photobiology.
[91] Q. Mei,et al. [Effects of naringin on proliferation, differentiation and maturation of rat calvarial osteoblasts in vitro]. , 2013, Zhongguo Zhong yao za zhi = Zhongguo zhongyao zazhi = China journal of Chinese materia medica.
[92] J. Morley,et al. Hormesis and amyloid-β protein: physiology or pathology? , 2012, Journal of Alzheimer's disease : JAD.
[93] N. Kamalakkannan,et al. Antihyperglycaemic and antioxidant effect of rutin, a polyphenolic flavonoid, in streptozotocin-induced diabetic wistar rats. , 2006, Basic & clinical pharmacology & toxicology.
[94] Mamdouh M. Ali,et al. The influence of naringin on the oxidative state of rats with streptozotocin-induced acute hyperglycaemia. , 2004, Zeitschrift fur Naturforschung. C, Journal of biosciences.