1
|
Skóra B, Matuszewska P, Masicz M, Sikora K, Słomczewska M, Sołtysek P, Szychowski KA. Crosstalk between the aryl hydrocarbon receptor (AhR) and the peroxisome proliferator-activated receptor gamma (PPARγ) as a key factor in the metabolism of silver nanoparticles in neuroblastoma (SH-SY5Y) cells in vitro. Toxicol Appl Pharmacol 2023; 458:116339. [PMID: 36473513 DOI: 10.1016/j.taap.2022.116339] [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: 10/31/2022] [Revised: 11/20/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
The potential usefulness of silver nanoparticles (AgNPs) in anticancer therapy has been postulated for many years. However, little is known to date about the exact impact of such NPs on intracellular detoxication pathways. Therefore, the aim of this study was to determine the impact of AgNPs on the AhR-PPARγ-CYP1A1 pathway in neuroblastoma (SH-SY5Y) cells. The obtained results showed a decrease in the metabolic activity of the SH-SY5Y cells at the 50 and 100 μg/mL concentrations with an increase in caspase-3 activity. An increase in the intercellular ROS production was observed at the 1 and 10 μg/mL concentrations. The co-treatment of the AgNP-treated cells with the AhR and PPARγ inhibitors abolished the effect of the tested AgNPs in the SH-SY5Y cells. In turn, the CYP1A1 activity assay showed a decrease in this parameter in the AgNP-treated cells. Moreover, the gene expression analysis demonstrated that AgNPs were able to increase the AhR and CYP1A1 mRNA expression and decrease the PPARγ gene expression after the 6-h treatment. In turn, an increase in the AhR and PPARγ protein expression was observed after 24 h. Summarizing, the study shows for the first time that AgNPs with a 5-nm diameter size are able to exert a cytotoxic effect on SH-SH5Y cells in a ROS-dependent manner affect the AhR-PPARγ-CYP1A1 pathway inter alia by inhibiting the activity of CYP1A1. This is important due to given present research approaches using such NPs as enhancer agents in the modern PPARγ inhibitor-based anticancer therapy.
Collapse
Affiliation(s)
- Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland.
| | - Paulina Matuszewska
- Medical Biotechnology Student's Science Group "Helisa", Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Martyna Masicz
- Medical Biotechnology Student's Science Group "Helisa", Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Karolina Sikora
- Medical Biotechnology Student's Science Group "Helisa", Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Magnolia Słomczewska
- Medical Biotechnology Student's Science Group "Helisa", Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Paulina Sołtysek
- Medical Biotechnology Student's Science Group "Helisa", Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Konrad A Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| |
Collapse
|
2
|
Generation of resolving memory neutrophils through pharmacological training with 4-PBA or genetic deletion of TRAM. Cell Death Dis 2022; 13:345. [PMID: 35418110 PMCID: PMC9007399 DOI: 10.1038/s41419-022-04809-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023]
Abstract
Neutrophils are the dominant leukocytes in circulation and the first responders to infection and inflammatory cues. While the roles of neutrophils in driving inflammation have been widely recognized, the contribution of neutrophils in facilitating inflammation resolution is under-studied. Here, through single-cell RNA sequencing analysis, we identified a subpopulation of neutrophils exhibiting pro-resolving characteristics with greater Cd200r and Cd86 expression at the resting state. We further discovered that 4-PBA, a peroxisomal stress-reducing agent, can potently train neutrophils into the resolving state with enhanced expression of CD200R, CD86, as well as soluble pro-resolving mediators Resolvin D1 and SerpinB1. Resolving neutrophils trained by 4-PBA manifest enhanced phagocytosis and bacterial-killing functions. Mechanistically, the generation of resolving neutrophils is mediated by the PPARγ/LMO4/STAT3 signaling circuit modulated by TLR4 adaptor molecule TRAM. We further demonstrated that genetic deletion of TRAM renders the constitutive expansion of resolving neutrophils, with an enhanced signaling circuitry of PPARγ/LMO4/STAT3. These findings may have profound implications for the effective training of resolving neutrophils with therapeutic potential in the treatment of both acute infection as well as chronic inflammatory diseases.
Collapse
|
3
|
Neuroprotective Function of High Glycolytic Activity in Astrocytes: Common Roles in Stroke and Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms22126568. [PMID: 34207355 PMCID: PMC8234992 DOI: 10.3390/ijms22126568] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 12/21/2022] Open
Abstract
Astrocytes (also, astroglia) consume huge amounts of glucose and produce lactate regardless of sufficient oxygen availability, indicating a high capacity for aerobic glycolysis. Glycolysis in astrocytes is activated in accordance with neuronal excitation and leads to increases in the release of lactate from astrocytes. Although the fate of this lactate remains somewhat controversial, it is believed to fuel neurons as an energy substrate. Besides providing lactate, astrocytic glycolysis plays an important role in neuroprotection. Among the minor pathways of glucose metabolism, glucose flux to the pentose-phosphate pathway (PPP), a major shunt pathway of glycolysis, is attracting research interest. In fact, PPP activity in astrocytes is five to seven times higher than that in neurons. The astrocytic PPP plays a key role in protecting neurons against oxidative stress by providing neurons with a reduced form of glutathione, which is necessary to eliminate reactive oxygen species. Therefore, enhancing astrocytic glycolysis might promote neuronal protection during acute ischemic stroke. Contrariwise, the dysfunction of astrocytic glycolysis and the PPP have been implicated in the pathogenesis of various neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis, since mitochondrial dysfunction and oxidative stress trigger and accelerate disease progression.
Collapse
|
4
|
De Caro C, Di Cesare Mannelli L, Branca JJV, Micheli L, Citraro R, Russo E, De Sarro G, Ghelardini C, Calignano A, Russo R. Pain Modulation in WAG/Rij Epileptic Rats (A Genetic Model of Absence Epilepsy): Effects of Biological and Pharmacological Histone Deacetylase Inhibitors. Front Pharmacol 2020; 11:549191. [PMID: 33343343 PMCID: PMC7745735 DOI: 10.3389/fphar.2020.549191] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 10/29/2020] [Indexed: 01/18/2023] Open
Abstract
Epigenetic mechanisms are involved in epilepsy and chronic pain development. About that, we studied the effects of the natural histone deacetylase (HDAC) inhibitor sodium butyrate (BUT) in comparison with valproic acid (VPA) in a validated genetic model of generalized absence epilepsy and epileptogenesis. WAG/Rij rats were treated with BUT (30 mg/kg), VPA (300 mg/kg), and their combination (BUT + VPA) daily per os for 6 months. Rats were subjected at Randall–Selitto, von Frey, hot plate, and tail flick tests after 1, 3, and 6 months of treatment to evaluate hypersensitivity to noxious and non-noxiuous stimuli. Moreover, PPAR-γ (G3335 1 mg/kg), GABA-B (CGP35348 80 mg/kg), and opioid (naloxone 1 mg/kg) receptor antagonists were administrated to investigate the possible mechanisms involved in analgesic activity. The expression of NFkB, glutathione reductase, and protein oxidation (carbonylation) was also evaluated by Western blot analysis. WAG/Rij rats showed an altered pain threshold throughout the study (p < 0.001). BUT and BUT + VPA treatment reduced hypersensitivity (p < 0.01). VPA was significantly effective only after 1 month (p < 0.01). All the three receptors are involved in BUT + VPA effects (p < 0.001). BUT and BUT + VPA decreased the expression of NFkB and enhanced glutathione reductase (p < 0.01); protein oxidation (carbonylation) was reduced (p < 0.01). No effect was reported with VPA. In conclusion BUT, alone or in coadministration with VPA, is a valuable candidate for managing the epilepsy-related persistent pain.
Collapse
Affiliation(s)
- Carmen De Caro
- Department of Pharmacy, University of Naples Federico II, Naples, Italy.,Department of Science of Health, School of Medicine and Surgery, University of Catanzaro, Catanzaro, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health-Neurofarba-Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Jacopo Junio Valerio Branca
- Department of Experimental and Clinical Medicine, Anatomy and Histology Section, University of Florence, Florence, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health-Neurofarba-Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Rita Citraro
- Department of Science of Health, School of Medicine and Surgery, University of Catanzaro, Catanzaro, Italy
| | - Emilio Russo
- Department of Science of Health, School of Medicine and Surgery, University of Catanzaro, Catanzaro, Italy
| | - Giovambattista De Sarro
- Department of Science of Health, School of Medicine and Surgery, University of Catanzaro, Catanzaro, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health-Neurofarba-Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Antonio Calignano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Roberto Russo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
5
|
Ganguli G, Pattanaik KP, Jagadeb M, Sonawane A. Mycobacterium tuberculosis Rv3034c regulates mTORC1 and PPAR-γ dependant pexophagy mechanism to control redox levels in macrophages. Cell Microbiol 2020; 22:e13214. [PMID: 32388919 DOI: 10.1111/cmi.13214] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/01/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022]
Abstract
Mycobacterium tuberculosis survives inside the macrophages by employing several host immune evasion strategies. Here, we reported a novel mechanism in which M. tuberculosis acetyltransferase, encoded by Rv3034c, induces peroxisome homeostasis to regulate host oxidative stress levels to facilitate intracellular mycobacterial infection. Presence of M. tuberculosis Rv3034c induces the expression of peroxisome biogenesis and proliferation factors such as Pex3, Pex5, Pex19, Pex11b, Fis-1 and DLP-1; while depletion of Rv3034c decreased the expression of these molecules, thereby selective degradation of peroxisomes via pexophagy. Further studies revealed that M. tuberculosis Rv3034c inhibit induction of pexophagy mechanism by down-regulating the expression of pexophagy associated proteins (p-AMPKα, p-ULK-1, Atg5, Atg7, Beclin-1, LC3-II, TFEB and Keap-1) and adaptor molecules (NBR1 and p62). Inhibition was found to be dependent on the phosphorylation of mTORC1 and activation of peroxisome proliferator activated receptor-γ. In order to maintain intracellular homeostasis during oxidative stress, M. tuberculosis Rv3034c was found to induce degradation of dysfunctional and damaged peroxisomes through activation of Pex14 in infected macrophages. In conclusion, this is the first report which demonstrated that M. tuberculosis acetyltransferase regulate peroxisome homeostasis in response to intracellular redox levels to favour mycobacterial infection in macrophage.
Collapse
Affiliation(s)
- Geetanjali Ganguli
- School of Biotechnology, KIIT (Deemed to be University), Bhubaneswar, India
| | | | - Manaswini Jagadeb
- School of Biotechnology, KIIT (Deemed to be University), Bhubaneswar, India
| | - Avinash Sonawane
- School of Biotechnology, KIIT (Deemed to be University), Bhubaneswar, India.,Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, India
| |
Collapse
|
6
|
Uzor NE, McCullough LD, Tsvetkov AS. Peroxisomal Dysfunction in Neurological Diseases and Brain Aging. Front Cell Neurosci 2020; 14:44. [PMID: 32210766 PMCID: PMC7075811 DOI: 10.3389/fncel.2020.00044] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/18/2020] [Indexed: 12/17/2022] Open
Abstract
Peroxisomes exist in most cells, where they participate in lipid metabolism, as well as scavenging the reactive oxygen species (ROS) that are produced as by-products of their metabolic functions. In certain tissues such as the liver and kidneys, peroxisomes have more specific roles, such as bile acid synthesis in the liver and steroidogenesis in the adrenal glands. In the brain, peroxisomes are critically involved in creating and maintaining the lipid content of cell membranes and the myelin sheath, highlighting their importance in the central nervous system (CNS). This review summarizes the peroxisomal lifecycle, then examines the literature that establishes a link between peroxisomal dysfunction, cellular aging, and age-related disorders that affect the CNS. This review also discusses the gap of knowledge in research on peroxisomes in the CNS.
Collapse
Affiliation(s)
- Ndidi-Ese Uzor
- Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, TX, United States
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Louise D. McCullough
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, United States
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX, United States
- UTHealth Consortium on Aging, University of Texas McGovern Medical School, Houston, TX, United States
| | - Andrey S. Tsvetkov
- Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, TX, United States
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, United States
- UTHealth Consortium on Aging, University of Texas McGovern Medical School, Houston, TX, United States
| |
Collapse
|
7
|
Quintão NLM, Santin JR, Stoeberl LC, Corrêa TP, Melato J, Costa R. Pharmacological Treatment of Chemotherapy-Induced Neuropathic Pain: PPARγ Agonists as a Promising Tool. Front Neurosci 2019; 13:907. [PMID: 31555078 PMCID: PMC6722212 DOI: 10.3389/fnins.2019.00907] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/14/2019] [Indexed: 12/24/2022] Open
Abstract
Chemotherapy-induced neuropathic pain (CINP) is one of the most severe side effects of anticancer agents, such as platinum- and taxanes-derived drugs (oxaliplatin, cisplatin, carboplatin and paclitaxel). CINP may even be a factor of interruption of treatment and consequently increasing the risk of death. Besides that, it is important to take into consideration that the incidence of cancer is increasing worldwide, including colorectal, gastric, lung, cervical, ovary and breast cancers, all treated with the aforementioned drugs, justifying the concern of the medical community about the patient’s quality of life. Several physiopathological mechanisms have already been described for CINP, such as changes in axonal transport, mitochondrial damage, increased ion channel activity and inflammation in the central nervous system (CNS). Another less frequent event that may occur after chemotherapy, particularly under oxaliplatin treatment, is the central neurotoxicity leading to disorders such as mental confusion, catatonia, hyporeflexia, etc. To date, no pharmacological therapy has shown satisfactory effect in these cases. In this scenario, duloxetine is the only drug currently in clinical use. Peroxisome proliferator-activated receptors (PPARs) belong to the class of nuclear receptors and are present in several tissues, mainly participating in lipid and glucose metabolism and inflammatory response. There are three PPAR isoforms: α, β/δ and γ. PPARγ, the protagonist of this review, is expressed in adipose tissue, large intestine, spleen and neutrophils. This subtype also plays important role in energy balance, lipid biosynthesis and adipogenesis. The effects of PPARγ agonists, known for their positive activity on type II diabetes mellitus, have been explored and present promising effects in the control of neuropathic pain, including CINP, and also cancer. This review focuses largely on the mechanisms involved in chemotherapy-induced neuropathy and the effects of the activation of PPARγ to treat CINP. It is the aim of this review to help understanding and developing novel CINP therapeutic strategies integrating PPARγ signalling.
Collapse
Affiliation(s)
| | | | | | | | - Jéssica Melato
- School of Heath Science, Universidade do Vale do Itajaí, Itajaí, Brazil
| | - Robson Costa
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| |
Collapse
|
8
|
Ganguli G, Mukherjee U, Sonawane A. Peroxisomes and Oxidative Stress: Their Implications in the Modulation of Cellular Immunity During Mycobacterial Infection. Front Microbiol 2019; 10:1121. [PMID: 31258517 PMCID: PMC6587667 DOI: 10.3389/fmicb.2019.01121] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 05/03/2019] [Indexed: 12/12/2022] Open
Abstract
Host redox dependent physiological responses play crucial roles in the determination of mycobacterial infection process. Mtb explores oxygen rich lung microenvironments to initiate infection process, however, later on the bacilli adapt to oxygen depleted conditions and become non-replicative and unresponsive toward anti-TB drugs to enter in the latency stage. Mtb is equipped with various sensory mechanisms and a battery of pro- and anti-oxidant enzymes to protect themselves from the host oxidative stress mechanisms. After host cell invasion, mycobacteria induces the expression of NADPH oxidase 2 (NOX2) to generate superoxide radicals (O 2 - ), which are then converted to more toxic hydrogen peroxide (H2O2) by superoxide dismutase (SOD) and subsequently reduced to water by catalase. However, the metabolic cascades and their key regulators associated with cellular redox homeostasis are poorly understood. Phagocytosed mycobacteria en route through different subcellular organelles, where the local environment generated during infection determines the outcome of disease. For a long time, mitochondria were considered as the key player in the redox regulation, however, accumulating evidences report vital role for peroxisomes in the maintenance of cellular redox equilibrium in eukaryotic cells. Deletion of peroxisome-associated peroxin genes impaired detoxification of reactive oxygen species and peroxisome turnover post-infection, thereby leading to altered synthesis of transcription factors, various cell-signaling cascades in favor of the bacilli. This review focuses on how mycobacteria would utilize host peroxisomes to alter redox balance and metabolic regulatory mechanisms to support infection process. Here, we discuss implications of peroxisome biogenesis in the modulation of host responses against mycobacterial infection.
Collapse
Affiliation(s)
- Geetanjali Ganguli
- School of Biotechnology, KIIT (deemed to be University), Bhubaneswar, India
| | - Utsav Mukherjee
- School of Biotechnology, KIIT (deemed to be University), Bhubaneswar, India
| | - Avinash Sonawane
- School of Biotechnology, KIIT (deemed to be University), Bhubaneswar, India
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| |
Collapse
|
9
|
Cornelis MC. Genetic determinants of beverage consumption: Implications for nutrition and health. ADVANCES IN FOOD AND NUTRITION RESEARCH 2019; 89:1-52. [PMID: 31351524 PMCID: PMC7047661 DOI: 10.1016/bs.afnr.2019.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Beverages make important contributions to nutritional intake and their role in health has received much attention. This review focuses on the genetic determinants of common beverage consumption and how research in this field is contributing insight to what and how much we consume and why this genetic knowledge matters from a research and public health perspective. The earliest efforts in gene-beverage behavior mapping involved genetic linkage and candidate gene analysis but these approaches have been largely replaced by genome-wide association studies (GWAS). GWAS have identified biologically plausible loci underlying alcohol and coffee drinking behavior. No GWAS has identified variants specifically associated with consumption of tea, juice, soda, wine, beer, milk or any other common beverage. Thus far, GWAS highlight an important behavior-reward component (as opposed to taste) to beverage consumption which may serve as a potential barrier to dietary interventions. Loci identified have been used in Mendelian randomization and gene×beverage interaction analysis of disease but results have been mixed. This research is necessary as it informs the clinical relevance of SNP-beverage associations and thus genotype-based personalized nutrition, which is gaining interest in the commercial and public health sectors.
Collapse
Affiliation(s)
- Marilyn C Cornelis
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.
| |
Collapse
|
10
|
Giampietro L, Gallorini M, De Filippis B, Amoroso R, Cataldi A, di Giacomo V. PPAR-γ agonist GL516 reduces oxidative stress and apoptosis occurrence in a rat astrocyte cell line. Neurochem Int 2019; 126:239-245. [PMID: 30946848 DOI: 10.1016/j.neuint.2019.03.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 03/19/2019] [Accepted: 03/27/2019] [Indexed: 01/11/2023]
Abstract
AIMS The worldwide increase in aging population is prevalently associated with the increase of neurodegenerative diseases. Peroxisome Proliferator-Activated Receptors (PPARs) are ligand-modulated transcriptional factors which belong to the nuclear hormone receptor superfamily which regulates peroxisome proliferation. The PPAR-γ is the most extensively studied among the three isoforms and the neuroprotective effects of PPAR-γ agonists have been recently demonstrated in a variety of preclinical models of neurological disorders. The aim of the study is to biologically evaluate the neuroprotective effects of new PPAR-γ selective agonists in an in vitro model. MAIN METHODS CTX-TNA2 rat astrocytes were treated with G3335, a PPAR-γ antagonist, to simulate the conditions of a neurological disorder. Newly synthetized PPAR-γ selective agonists were added to the cell culture. Cytotoxicity was assessed by MTT assay, catalase activity was investigated by a colorimetric assay, Reactive Oxygen Species (ROS) production and apoptosis occurrence were measured by flow cytometry. Western blotting were performed to measure the levels of protein involved in the apoptotic pathway. KEY FINDINGS Four PPAR-γ agonists were selected. Among them, the GL516, a fibrate derivative, showed low cytotoxicity and proved effective in restoring the catalase activity, reducing ROS production and decreasing the apoptosis occurrence triggered by the G3335 administration. The effects of this molecule appear to be comparable to the reference compound rosiglitazone, a potent and selective PPAR-γ agonist, mainly at prolonged exposure times (96 h). SIGNIFICANCE Based on recent evidence, hypofunctionality of the PPAR-γ in glial cells could be present in neurodegenerative diseases and could participate in pathological mechanisms through peroxisomal damage. The fibrate derivative PPAR-γ agonist GL516 emerged as the most promising molecule of the series and could have a role in preventing the pathophysiology of neurodegenerative disorders.
Collapse
Affiliation(s)
- Letizia Giampietro
- Department of Pharmacy, University G. d'Annunzio, Chieti-Pescara, via dei Vestini 31, Chieti Scalo, Italy
| | - Marialucia Gallorini
- Department of Pharmacy, University G. d'Annunzio, Chieti-Pescara, via dei Vestini 31, Chieti Scalo, Italy.
| | - Barbara De Filippis
- Department of Pharmacy, University G. d'Annunzio, Chieti-Pescara, via dei Vestini 31, Chieti Scalo, Italy
| | - Rosa Amoroso
- Department of Pharmacy, University G. d'Annunzio, Chieti-Pescara, via dei Vestini 31, Chieti Scalo, Italy
| | - Amelia Cataldi
- Department of Pharmacy, University G. d'Annunzio, Chieti-Pescara, via dei Vestini 31, Chieti Scalo, Italy
| | - Viviana di Giacomo
- Department of Pharmacy, University G. d'Annunzio, Chieti-Pescara, via dei Vestini 31, Chieti Scalo, Italy
| |
Collapse
|
11
|
Reilly MT, Noronha A, Goldman D, Koob GF. Genetic studies of alcohol dependence in the context of the addiction cycle. Neuropharmacology 2017; 122:3-21. [PMID: 28118990 DOI: 10.1016/j.neuropharm.2017.01.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/13/2017] [Accepted: 01/19/2017] [Indexed: 12/16/2022]
Abstract
Family, twin and adoption studies demonstrate clearly that alcohol dependence and alcohol use disorders are phenotypically complex and heritable. The heritability of alcohol use disorders is estimated at approximately 50-60% of the total phenotypic variability. Vulnerability to alcohol use disorders can be due to multiple genetic or environmental factors or their interaction which gives rise to extensive and daunting heterogeneity. This heterogeneity makes it a significant challenge in mapping and identifying the specific genes that influence alcohol use disorders. Genetic linkage and (candidate gene) association studies have been used now for decades to map and characterize genomic loci and genes that underlie the genetic vulnerability to alcohol use disorders. These approaches have been moderately successful in identifying several genes that contribute to the complexity of alcohol use disorders. Recently, genome-wide association studies have become one of the major tools for identifying genes for alcohol use disorders by examining correlations between millions of common single-nucleotide polymorphisms with diagnosis status. Genome-wide association studies are just beginning to uncover novel biology; however, the functional significance of results remains a matter of extensive debate and uncertainty. In this review, we present a select group of genome-wide association studies of alcohol dependence, as one example of a way to generate functional hypotheses, within the addiction cycle framework. This analysis may provide novel directions for validating the functional significance of alcohol dependence candidate genes. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Matthew T Reilly
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Division of Neuroscience and Behavior, 5635 Fishers Lane, Bethesda, MD 20852, USA.
| | - Antonio Noronha
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Division of Neuroscience and Behavior, 5635 Fishers Lane, Bethesda, MD 20852, USA
| | - David Goldman
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Chief, Laboratory of Neurogenetics, 5635 Fishers Lane, Bethesda, MD 20852, USA
| | - George F Koob
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Director NIAAA, 5635 Fishers Lane, Bethesda, MD 20852, USA
| |
Collapse
|
12
|
Iglesias J, Morales L, Barreto GE. Metabolic and Inflammatory Adaptation of Reactive Astrocytes: Role of PPARs. Mol Neurobiol 2016; 54:2518-2538. [PMID: 26984740 DOI: 10.1007/s12035-016-9833-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/04/2016] [Indexed: 01/10/2023]
Abstract
Astrocyte-mediated inflammation is associated with degenerative pathologies such as Alzheimer's and Parkinson's diseases and multiple sclerosis. The acute inflammation and morphological and metabolic changes that astrocytes develop after the insult are known as reactive astroglia or astrogliosis that is an important response to protect and repair the lesion. Astrocytes optimize their metabolism to produce lactate, glutamate, and ketone bodies in order to provide energy to the neurons that are deprived of nutrients upon insult. Firstly, we review the basis of inflammation and morphological changes of the different cell population implicated in reactive gliosis. Next, we discuss the more active metabolic pathways in healthy astrocytes and explain the metabolic response of astrocytes to the insult in different pathologies and which metabolic alterations generate complications in these diseases. We emphasize the role of peroxisome proliferator-activated receptors isotypes in the inflammatory and metabolic adaptation of astrogliosis developed in ischemia or neurodegenerative diseases. Based on results reported in astrocytes and other cells, we resume and hypothesize the effect of peroxisome proliferator-activated receptor (PPAR) activation with ligands on different metabolic pathways in order to supply energy to the neurons. The activation of selective PPAR isotype activity may serve as an input to better understand the role played by these receptors on the metabolic and inflammatory compensation of astrogliosis and might represent an opportunity to develop new therapeutic strategies against traumatic brain injuries and neurodegenerative diseases.
Collapse
Affiliation(s)
- José Iglesias
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia.
| | - Ludis Morales
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
- Universidad Científica del Sur, Lima, Peru
| |
Collapse
|