Natural product-based bioactive agents in combination attenuate neuroinflammation in a tri-culture model

Introduction: Neuroinflammation is an important pathological event contributing to the onset and progression of neurodegenerative diseases. The hyperactivation of microglia triggers the release of excessive proinflammatory mediators that lead to the leaky blood-brain barrier and impaired neuronal survival. Andrographolide (AN), baicalein (BA) and 6-shogaol (6-SG) possess anti-neuroinflammatory properties through diverse mechanisms of action. The present study aims to investigate the effects of the pair-combinations of these bioactive compounds in attenuating neuroinflammation.Methods: A tri-culture model with microglial N11 cells, microvascular endothelial MVEC(B3) cells, and neuroblastoma N2A cells was established in a transwell system. AN, BA and 6-SG used alone (25 µM) or in pair-wised combinations (12.5 + 12.5 µM) were subjected to the tri-culture system. Upon the stimulation of lipopolysaccharides (LPS) at 1 μg/mL, tumor necrosis factor-alpha (TNF-α) and interleukin 6 (IL-6) levels were determined by ELISA assays. Immunofluorescence staining was applied to investigate the nuclear translocation of nuclear factor kappa B p65 (NF-κB p65) on N11 cells, expressions of protein zonula occludens-1 (ZO-1) on MVEC cells and phosphorylated tau (p-tau) on N2A cells, respectively. The endothelial barrier permeability of MVEC cells was assessed by the Evans blue dye, and the resistance from the endothelial barrier was measured by transepithelial/endothelial electrical resistance (TEER) value. Neuronal survival of N2A cells was determined by Alamar blue and MTT assays.Results: Combinations of AN-SG and BA-SG synergistically lowered the TNF and IL-6 levels in LPS-induced N11 cells. Remarkably, the combined anti-neuroinflammatory effects of AN-SG and BA-SG remained significantly greater compared to their individual components at the same concentration level. The molecular mechanism of the attenuated neuroinflammation was likely to be mediated by downregulation of NF-κB p65 translocation (p < 0.0001 vs. LPS stimulation) in N11 cells. In the MVEC cells, both AN-SG and BA-SG restored TEER values, ZO-1 expression and reduced permeability. Furthermore, AN-SG and BA-SG significantly improved neuronal survival and reduced expressions of p-tau on N2A cells.Discussion: The AN-SG and BA-SG combinations showed greater anti-neuroinflammatory potential than those used alone in mono- and tri-cultured N11 cells, thereby further protecting endothelial tight junction and neuronal survival. Taken together, AN-SG and BA-SG may provide improved anti-neuroinflammatory and neuroprotective activities.

[1]  J. Hai,et al.  URB597 and Andrographolide Improve Brain Microvascular Endothelial Cell Permeability and Apoptosis by Reducing Oxidative Stress and Inflammation Associated with Activation of Nrf2 Signaling in Oxygen-Glucose Deprivation , 2022, Oxidative medicine and cellular longevity.

[2]  E. Hol,et al.  The neurovascular unit in leukodystrophies: towards solving the puzzle , 2022, Fluids and barriers of the CNS.

[3]  Lin F. Yang,et al.  Estimation of the global prevalence of dementia in 2019 and forecasted prevalence in 2050: an analysis for the Global Burden of Disease Study 2019 , 2022, The Lancet. Public health.

[4]  N. Cicero,et al.  Role of nutraceuticals on neurodegenerative diseases: neuroprotective and immunomodulant activity , 2021, Natural product research.

[5]  T. Kanda,et al.  New BBB Model Reveals That IL-6 Blockade Suppressed the BBB Disorder, Preventing Onset of NMOSD , 2021, Neurology: Neuroimmunology & Neuroinflammation.

[6]  S. Dohgu,et al.  Blood-Brain Barrier Dysfunction Amplifies the Development of Neuroinflammation: Understanding of Cellular Events in Brain Microvascular Endothelial Cells for Prevention and Treatment of BBB Dysfunction , 2021, Frontiers in Cellular Neuroscience.

[7]  Sidharth Mehan,et al.  Targeting Nrf2/HO-1 anti-oxidant signaling pathway in the progression of multiple sclerosis and influences on neurological dysfunctions , 2021 .

[8]  Y. Shang,et al.  Flavonoids from Scutellaria baicalensis Georgi Stems and Leaves Regulate the Brain Tau Hyperphosphorylation at M|ultiple Sites Induced by Composited Aβ in Rats. , 2021, CNS & neurological disorders drug targets.

[9]  Junlei Chang,et al.  Blood–Brain Barrier Breakdown: An Emerging Biomarker of Cognitive Impairment in Normal Aging and Dementia , 2021, Frontiers in Neuroscience.

[10]  J. Schober,et al.  NNC 26-9100 increases Aβ1-42 phagocytosis, inhibits nitric oxide production and decreases calcium in BV2 microglia cells , 2021, PloS one.

[11]  Karamjeet Kaur,et al.  Protective effect of andrographolide against STZ induced Alzheimer’s disease in experimental rats: possible neuromodulation and Aβ(1–42) analysis , 2021, Inflammopharmacology.

[12]  Fang Ma,et al.  Andrographolide ameliorates neuroinflammation in APP/PS1 transgenic mice. , 2021, International immunopharmacology.

[13]  Qidi Ai,et al.  The Anti-Neuroinflammatory Effect of Fuzi and Ganjiang Extraction on LPS-Induced BV2 Microglia and Its Intervention Function on Depression-Like Behavior of Cancer-Related Fatigue Model Mice , 2021, Frontiers in Pharmacology.

[14]  E. D. Khilazheva,et al.  Blood–Brain Barrier and Neurovascular Unit In Vitro Models for Studying Mitochondria-Driven Molecular Mechanisms of Neurodegeneration , 2021, International journal of molecular sciences.

[15]  S. Vidyadaran,et al.  Nitric oxide modulation in neuroinflammation and the role of mesenchymal stem cells , 2021, Experimental biology and medicine.

[16]  Jiarui Wu,et al.  A review on α-mangostin as a potential multi-target-directed ligand for Alzheimer's disease. , 2021, European journal of pharmacology.

[17]  V. Uversky,et al.  Baicalein inhibits heparin-induced Tau aggregation by initializing non-toxic Tau oligomer formation , 2021, Cell communication and signaling : CCS.

[18]  S. Seto,et al.  A novel tri‐culture model for neuroinflammation , 2020, Journal of neurochemistry.

[19]  R. Maccioni,et al.  The Emerging Role of Nutraceuticals and Phytochemicals in the Prevention and Treatment of Alzheimer's Disease. , 2020, Journal of Alzheimer's disease : JAD.

[20]  Zhen Zhao,et al.  G Protein-Coupled Receptors in the Mammalian Blood-Brain Barrier , 2020, Frontiers in Cellular Neuroscience.

[21]  G. Zengin,et al.  Nutraceuticals in Neurological Disorders , 2020, International journal of molecular sciences.

[22]  John H. Zhang,et al.  TREM2 activation attenuates neuroinflammation and neuronal apoptosis via PI3K/Akt pathway after intracerebral hemorrhage in mice , 2020, Journal of Neuroinflammation.

[23]  Anwen Shao,et al.  Neurovascular Unit Dysfunction and Neurodegenerative Disorders , 2020, Frontiers in Neuroscience.

[24]  W. Banks,et al.  In vitro modeling of blood–brain barrier and interface functions in neuroimmune communication , 2020, Fluids and Barriers of the CNS.

[25]  K. Sirajudeen,et al.  Natural Products and Their Bioactive Compounds: Neuroprotective Potentials against Neurodegenerative Diseases , 2020, Evidence-based complementary and alternative medicine : eCAM.

[26]  Nicholas E. Propson,et al.  Type I interferon response drives neuroinflammation and synapse loss in Alzheimer disease. , 2020, The Journal of clinical investigation.

[27]  S. Rai,et al.  NF-κB-Mediated Neuroinflammation in Parkinson’s Disease and Potential Therapeutic Effect of Polyphenols , 2019, Neurotoxicity Research.

[28]  N. Greig,et al.  Neuroinflammation as a Factor of Neurodegenerative Disease: Thalidomide Analogs as Treatments , 2019, Front. Cell Dev. Biol..

[29]  Y. Tachibana,et al.  Dual microglia effects on blood brain barrier permeability induced by systemic inflammation , 2019, Nature Communications.

[30]  R. Maccioni,et al.  Neuroinflammation as a Common Feature of Neurodegenerative Disorders , 2019, Front. Pharmacol..

[31]  Chunfu Wu,et al.  Baicalein administered in the subacute phase ameliorates ischemia-reperfusion-induced brain injury by reducing neuroinflammation and neuronal damage. , 2019, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

[32]  Giulia Mantini,et al.  To Combine or Not Combine: Drug Interactions and Tools for Their Analysis. Reflections from the EORTC-PAMM Course on Preclinical and Early-phase Clinical Pharmacology , 2019, AntiCancer Research.

[33]  S. Bhowmick,et al.  Impairment of pericyte-endothelium crosstalk leads to blood-brain barrier dysfunction following traumatic brain injury , 2019, Experimental Neurology.

[34]  J. Morales-Garcia,et al.  Novel Approaches for the Treatment of Alzheimer’s and Parkinson’s Disease , 2019, International journal of molecular sciences.

[35]  W. Cubała,et al.  Blood-brain barrier permeability and physical exercise , 2019, Journal of Neuroinflammation.

[36]  Haiying Li,et al.  Andrographolide ameliorates intracerebral hemorrhage induced secondary brain injury by inhibiting neuroinflammation induction , 2018, Neuropharmacology.

[37]  B. Puri,et al.  Leaky brain in neurological and psychiatric disorders: Drivers and consequences , 2018, The Australian and New Zealand journal of psychiatry.

[38]  Hong Lu,et al.  Andrographolide Protects PC12 Cells Against β-Amyloid-Induced Autophagy-Associated Cell Death Through Activation of the Nrf2-Mediated p62 Signaling Pathway , 2018, International journal of molecular sciences.

[39]  R. Maccioni,et al.  Alzheimer´s Disease in the Perspective of Neuroimmunology , 2018, The open neurology journal.

[40]  R. Tanzi,et al.  A 3D Human Tri-Culture System Modeling Neurodegeneration and Neuroinflammation in Alzheimer’s Disease , 2018, Nature Neuroscience.

[41]  K. Plate,et al.  Functional morphology of the blood-brain barrier in health and disease , 2018, Acta Neuropathologica.

[42]  Luc Buée,et al.  Tau and neuroinflammation: What impact for Alzheimer's Disease and Tauopathies? , 2018, Biomedical journal.

[43]  W. Xiao,et al.  In silico-based screen synergistic drug combinations from herb medicines: a case using Cistanche tubulosa , 2017, Scientific Reports.

[44]  B. Zlokovic,et al.  Alzheimer’s disease: A matter of blood–brain barrier dysfunction? , 2017, The Journal of experimental medicine.

[45]  S. Lorkowski,et al.  Optimized incubation regime for nitric oxide measurements in murine macrophages using the Griess assay. , 2017, Journal of immunological methods.

[46]  G. Du,et al.  Baicalein exerts anti‐neuroinflammatory effects to protect against rotenone‐induced brain injury in rats , 2017, International immunopharmacology.

[47]  Zhongming Chen,et al.  Reducing Neuroinflammation in Psychiatric Disorders: Novel Target of Phosphodiesterase 4 (PDE4) and Developing of the PDE4 Inhibitors , 2017 .

[48]  S. Yao,et al.  NLRP3 Inflammasome in Neurological Diseases, from Functions to Therapies , 2017, Front. Cell. Neurosci..

[49]  Roger D. Kamm,et al.  A 3D neurovascular microfluidic model consisting of neurons, astrocytes and cerebral endothelial cells as a blood-brain barrier. , 2017, Lab on a chip.

[50]  Bo Wei,et al.  The flavonoid baicalein rescues synaptic plasticity and memory deficits in a mouse model of Alzheimer’s disease , 2016, Behavioural Brain Research.

[51]  R. Thangavel,et al.  Neuroinflammation Induces Neurodegeneration. , 2016, Journal of neurology, neurosurgery and spine.

[52]  Shao-Cong Sun,et al.  NF-κB in inflammation and renal diseases , 2015, Cell & Bioscience.

[53]  Jennifer Luebke,et al.  Depletion of microglia and inhibition of exosome synthesis halt tau propagation , 2015, Nature Neuroscience.

[54]  Xin Wei,et al.  Increasing the Permeability of the Blood–brain Barrier in Three Different Models in vivo , 2015, CNS neuroscience & therapeutics.

[55]  Mickael Guedj,et al.  Analysis of drug combinations: current methodological landscape , 2015, Pharmacology research & perspectives.

[56]  B. Frey,et al.  Disruption in the Blood-Brain Barrier: The Missing Link between Brain and Body Inflammation in Bipolar Disorder? , 2015, Neural plasticity.

[57]  N. Inestrosa,et al.  Andrographolide reduces cognitive impairment in young and mature AβPPswe/PS-1 mice , 2014, Molecular Neurodegeneration.

[58]  Toshio Tanaka,et al.  IL-6 in inflammation, immunity, and disease. , 2014, Cold Spring Harbor perspectives in biology.

[59]  P. Mecocci,et al.  Nutraceuticals in cognitive impairment and Alzheimer’s disease , 2014, Front. Pharmacol..

[60]  R. Maccioni,et al.  Neuroinflammation in the pathogenesis of Alzheimer’s disease. A rational framework for the search of novel therapeutic approaches , 2014, Front. Cell. Neurosci..

[61]  M. Oh,et al.  6-Shogaol, an active compound of ginger, protects dopaminergic neurons in Parkinson's disease models via anti-neuroinflammation , 2013, Acta Pharmacologica Sinica.

[62]  M. Wolfe The Role of Tau in Neurodegenerative Diseases and Its Potential as a Therapeutic Target , 2012, Scientifica.

[63]  Dong Hyun Kim,et al.  6-Shogaol, a ginger product, modulates neuroinflammation: A new approach to neuroprotection , 2012, Neuropharmacology.

[64]  Yanwei Xing,et al.  Baicalin reduces the permeability of the blood-brain barrier during hypoxia in vitro by increasing the expression of tight junction proteins in brain microvascular endothelial cells. , 2012, Journal of ethnopharmacology.

[65]  T. Davis,et al.  Blood-brain barrier integrity and glial support: mechanisms that can be targeted for novel therapeutic approaches in stroke. , 2012, Current pharmaceutical design.

[66]  B. Lin,et al.  Polyphenols and neuroprotection against ischemia and neurodegeneration. , 2011, Mini reviews in medicinal chemistry.

[67]  M. Oh,et al.  Gami-Chunghyuldan ameliorates memory impairment and neurodegeneration induced by intrahippocampal Aβ1–42 oligomer injection , 2011, Neurobiology of Learning and Memory.

[68]  N. Lee,et al.  Limonene suppresses lipopolysaccharide-induced production of nitric oxide, prostaglandin E2, and pro-inflammatory cytokines in RAW 264.7 macrophages. , 2010, Journal of oleo science.

[69]  R. Kuljiš,et al.  The Role of Neuroimmunomodulation in Alzheimer's Disease , 2009, Annals of the New York Academy of Sciences.

[70]  M. Fuente Role of Neuroimmunomodulation in Aging , 2008 .

[71]  R. Kuljiš,et al.  The damage signals hypothesis of Alzheimer's disease pathogenesis. , 2008, Journal of Alzheimer's disease : JAD.

[72]  D. Tsikas Analysis of nitrite and nitrate in biological fluids by assays based on the Griess reaction: appraisal of the Griess reaction in the L-arginine/nitric oxide area of research. , 2007, Journal of chromatography. B, Analytical technologies in the biomedical and life sciences.

[73]  E. Benarroch Neurovascular unit dysfunction: A vascular component of Alzheimer disease? , 2007, Neurology.

[74]  L. Marsh,et al.  Non-dopaminergic treatment of cognitive impairment and dementia in Parkinson's disease: A review , 2006, Journal of the Neurological Sciences.

[75]  M. Block,et al.  Microglia and inflammation-mediated neurodegeneration: Multiple triggers with a common mechanism , 2005, Progress in Neurobiology.

[76]  Berislav V. Zlokovic,et al.  Neurovascular mechanisms of Alzheimer's neurodegeneration , 2005, Trends in Neurosciences.

[77]  G. Johnson,et al.  Tau phosphorylation in neuronal cell function and dysfunction , 2004, Journal of Cell Science.

[78]  A. S. Hamzah,et al.  Evaluation of antioxidant and nitric oxide inhibitory activities of selected Malaysian medicinal plants. , 2004, Journal of ethnopharmacology.

[79]  Xueji Zhang,et al.  Measurement of Nitric Oxide Production in Biological Systems by Using Griess Reaction Assay , 2003 .

[80]  Tony Wyss-Coray,et al.  Inflammation in Neurodegenerative Disease—A Double-Edged Sword , 2002, Neuron.

[81]  W. Mayhan Cellular mechanisms by which tumor necrosis factor-α produces disruption of the blood–brain barrier , 2002, Brain Research.

[82]  K. Audus,et al.  Nitric oxide and blood-brain barrier integrity. , 2001, Antioxidants & redox signaling.

[83]  J L West,et al.  Nitric oxide-generating polymers reduce platelet adhesion and smooth muscle cell proliferation. , 2000, Biomaterials.

[84]  Patrick R. Hof,et al.  Tau protein isoforms, phosphorylation and role in neurodegenerative disorders 1 1 These authors contributed equally to this work. , 2000, Brain Research Reviews.

[85]  T. Nikaido,et al.  [The study of Chinese herbal medicinal prescription with enzyme inhibitory activity. V. The study of hange-shashin-to, kanzo-shashin-to, shokyo-shashin-to with adenosine 3',5'-cyclic monophosphate phosphodiesterase]. , 1991, Yakugaku zasshi : Journal of the Pharmaceutical Society of Japan.

[86]  S. Moncada,et al.  Nitric oxide: physiology, pathophysiology, and pharmacology. , 1991, Pharmacological reviews.