1
|
Lymperopoulos A, Borges JI, Stoicovy RA. Cyclic Adenosine Monophosphate in Cardiac and Sympathoadrenal GLP-1 Receptor Signaling: Focus on Anti-Inflammatory Effects. Pharmaceutics 2024; 16:693. [PMID: 38931817 PMCID: PMC11206770 DOI: 10.3390/pharmaceutics16060693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/18/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is a multifunctional incretin hormone with various physiological effects beyond its well-characterized effect of stimulating glucose-dependent insulin secretion in the pancreas. An emerging role for GLP-1 and its receptor, GLP-1R, in brain neuroprotection and in the suppression of inflammation, has been documented in recent years. GLP-1R is a G protein-coupled receptor (GPCR) that couples to Gs proteins that stimulate the production of the second messenger cyclic 3',5'-adenosine monophosphate (cAMP). cAMP, acting through its two main effectors, protein kinase A (PKA) and exchange protein directly activated by cAMP (Epac), exerts several anti-inflammatory (and some pro-inflammatory) effects in cells, depending on the cell type. The present review discusses the cAMP-dependent molecular signaling pathways elicited by the GLP-1R in cardiomyocytes, cardiac fibroblasts, central neurons, and even in adrenal chromaffin cells, with a particular focus on those that lead to anti-inflammatory effects by the GLP-1R. Fully elucidating the role cAMP plays in GLP-1R's anti-inflammatory properties can lead to new and more precise targets for drug development and/or provide the foundation for novel therapeutic combinations of the GLP-1R agonist medications currently on the market with other classes of drugs for additive anti-inflammatory effect.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA; (J.I.B.); (R.A.S.)
| | | | | |
Collapse
|
2
|
Wang J, Zhi Z, Ding J, Jia N, Hu Y, Cai J, Li H, Tang J, Tang W, Mao X. Suppression of PGE2/EP2 signaling alleviates Hirschsprung disease by upregulating p38 mitogen-activated protein kinase activity. J Mol Med (Berl) 2023; 101:1125-1139. [PMID: 37522903 DOI: 10.1007/s00109-023-02353-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/11/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023]
Abstract
Hirschsprung disease (HSCR) is a congenital disorder caused by the failure of enteric neural crest cells (ENCCs) to colonize the distal bowel, resulting in absence of enteric nervous system. While a range of molecules and signaling pathways have been found to contribute to HSCR development, the risk factors and pathogenesis of this disease in many patients remain unknown. We previously demonstrated that increased activity of the prostaglandin E2 (PGE2)/PGE2 receptor subtype EP2 pathway can be a risk factor for HSCR. In this study, an Ednrb-deficient mouse model of HSCR was generated and used to investigate if PGE2/EP2 pathway could be a potential therapeutic target for HSCR. We found that downregulation of PGE2/EP2 signaling by siRNA-mediated ablation of a PGE2 synthase or pharmacologic blockage of EP2 enhanced ENCC colonization in the distal bowel of Ednrb-/- mice and alleviated their HSCR-like symptoms. Furthermore, blockage of EP2 was shown to promote ENCC migration through upregulating p38 mitogen-activated protein kinase activity, which was downregulated in the colon of Ednrb-/- mice and in the distal aganglionic bowel of HSCR patients. These data provide evidence that maternal exposure during embryonic development to an environment with dysregulated activation of the PGE2/EP2 pathway may predispose genetically susceptible offspring to HSCR, and avoidance or early disruption of maternal events (e.g. inflammation) that possibly enhance PGE2/EP2 signaling during pregnancy would reduce the occurrence and severity of this disease. KEY MESSAGES : Knockdown of PTGES alleviates HSCR severity in Ednrb-/- mice. Blockage of EP2-mediated PGE2 signaling alleviates HSCR severity in Ednrb-/- mice. Blockage of EP2-mediated PGE2 signaling promotes ENCC migration via enhancing p38 activity.
Collapse
Affiliation(s)
- Jiao Wang
- School of Life Science and Technology, Key Laboratory of Ministry of Education for Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Zhengke Zhi
- Department of Pediatric Surgery, Childrens Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210008, China
| | - Jie Ding
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Na Jia
- School of Life Science and Technology, Key Laboratory of Ministry of Education for Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Yuqing Hu
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Jiali Cai
- School of Life Science and Technology, Key Laboratory of Ministry of Education for Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Hongxing Li
- Department of Pediatric Surgery, Childrens Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210008, China
| | - Jie Tang
- Department of Pediatric Surgery, Childrens Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210008, China
| | - Weibing Tang
- Department of Pediatric Surgery, Childrens Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210008, China.
| | - Xiaohua Mao
- School of Life Science and Technology, Key Laboratory of Ministry of Education for Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu, 210096, China.
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
3
|
Belarbi K, Cuvelier E, Bonte MA, Desplanque M, Gressier B, Devos D, Chartier-Harlin MC. Glycosphingolipids and neuroinflammation in Parkinson's disease. Mol Neurodegener 2020; 15:59. [PMID: 33069254 PMCID: PMC7568394 DOI: 10.1186/s13024-020-00408-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 10/01/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease is a progressive neurodegenerative disease characterized by the loss of dopaminergic neurons of the nigrostriatal pathway and the formation of neuronal inclusions known as Lewy bodies. Chronic neuroinflammation, another hallmark of the disease, is thought to play an important role in the neurodegenerative process. Glycosphingolipids are a well-defined subclass of lipids that regulate crucial aspects of the brain function and recently emerged as potent regulators of the inflammatory process. Deregulation in glycosphingolipid metabolism has been reported in Parkinson's disease. However, the interrelationship between glycosphingolipids and neuroinflammation in Parkinson's disease is not well known. This review provides a thorough overview of the links between glycosphingolipid metabolism and immune-mediated mechanisms involved in neuroinflammation in Parkinson's disease. After a brief presentation of the metabolism and function of glycosphingolipids in the brain, it summarizes the evidences supporting that glycosphingolipids (i.e. glucosylceramides or specific gangliosides) are deregulated in Parkinson's disease. Then, the implications of these deregulations for neuroinflammation, based on data from human inherited lysosomal glycosphingolipid storage disorders and gene-engineered animal studies are outlined. Finally, the key molecular mechanisms by which glycosphingolipids could control neuroinflammation in Parkinson's disease are highlighted. These include inflammasome activation and secretion of pro-inflammatory cytokines, altered calcium homeostasis, changes in the blood-brain barrier permeability, recruitment of peripheral immune cells or production of autoantibodies.
Collapse
Affiliation(s)
- Karim Belarbi
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
- Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France
| | - Elodie Cuvelier
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
- Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France
| | - Marie-Amandine Bonte
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
| | - Mazarine Desplanque
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
- Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France
| | - Bernard Gressier
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
- Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France
| | - David Devos
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
- Département de Pharmacologie Médicale, I-SITE ULNE, LiCEND, Lille, France
| | | |
Collapse
|
4
|
Tavares LP, Negreiros-Lima GL, Lima KM, E Silva PMR, Pinho V, Teixeira MM, Sousa LP. Blame the signaling: Role of cAMP for the resolution of inflammation. Pharmacol Res 2020; 159:105030. [PMID: 32562817 DOI: 10.1016/j.phrs.2020.105030] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/06/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022]
Abstract
A complex intracellular signaling governs different cellular responses in inflammation. Extracellular stimuli are sensed, amplified, and transduced through a dynamic cellular network of messengers converting the first signal into a proper response: production of specific mediators, cell activation, survival, or death. Several overlapping pathways are coordinated to ensure specific and timely induction of inflammation to neutralize potential harms to the tissue. Ideally, the inflammatory response must be controlled and self-limited. Resolution of inflammation is an active process that culminates with termination of inflammation and restoration of tissue homeostasis. Comparably to the onset of inflammation, resolution responses are triggered by coordinated intracellular signaling pathways that transduce the message to the nucleus. However, the key messengers and pathways involved in signaling transduction for resolution are still poorly understood in comparison to the inflammatory network. cAMP has long been recognized as an inducer of anti-inflammatory responses and cAMP-dependent pathways have been extensively exploited pharmacologically to treat inflammatory diseases. Recently, cAMP has been pointed out as coordinator of key steps of resolution of inflammation. Here, we summarize the evidence for the role of cAMP at inducing important features of resolution of inflammation.
Collapse
Affiliation(s)
- Luciana P Tavares
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil; Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA..
| | - Graziele L Negreiros-Lima
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil.
| | - Kátia M Lima
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil; Post-Graduation Program in Pharmaceutical Sciences, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil.
| | - Patrícia M R E Silva
- Inflammation Laboratory, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.
| | - Vanessa Pinho
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Department of Morphology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil.
| | - Mauro M Teixeira
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil.
| | - Lirlândia P Sousa
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil; Post-Graduation Program in Pharmaceutical Sciences, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil.
| |
Collapse
|
5
|
Turner SMF, Falk DJ, Byrne BJ, Fuller DD. Transcriptome assessment of the Pompe (Gaa-/-) mouse spinal cord indicates widespread neuropathology. Physiol Genomics 2016; 48:785-794. [PMID: 27614205 PMCID: PMC6223572 DOI: 10.1152/physiolgenomics.00075.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/06/2016] [Indexed: 11/22/2022] Open
Abstract
Pompe disease, caused by deficiency of acid alpha-glucosidase (GAA), leads to widespread glycogen accumulation and profound neuromuscular impairments. There has been controversy, however, regarding the role of central nervous system pathology in Pompe motor dysfunction. We hypothesized that absence of GAA protein causes progressive activation of neuropathological signaling, including pathways associated with cell death. To test this hypothesis, genomic data (Affymetrix Mouse Gene Array 2.0ST) from the midcervical spinal cord in 6 and 16 mo old Pompe (Gaa-/-) mice were evaluated (Broad Institute Molecular Signature Database), along with spinal cord histology. The midcervical cord was selected because it contains phrenic motoneurons, and phrenic-diaphragm dysfunction is prominent in Pompe disease. Several clinically important themes for the neurologic etiology of Pompe disease emerged from this unbiased genomic assessment. First, pathways associated with cell death were strongly upregulated as Gaa-/- mice aged, and motoneuron apoptosis was histologically verified. Second, proinflammatory signaling was dramatically upregulated in the Gaa-/- spinal cord. Third, many signal transduction pathways in the Gaa-/- cervical cord were altered in a manner suggestive of impaired synaptic function. Notably, glutamatergic signaling pathways were downregulated, as were "synaptic plasticity pathways" including genes related to neuroplasticity. Fourth, many genes and pathways related to cellular metabolism are dysregulated. Collectively, the data unequivocally confirm that systemic absence of GAA induces a complex neuropathological cascade in the spinal cord. Most importantly, the results indicate that Pompe is a neurodegenerative condition, and this underscores the need for early therapeutic intervention capable of targeting the central nervous system.
Collapse
Affiliation(s)
- S M F Turner
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, Florida
| | - D J Falk
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, Florida
- Department of Pediatrics, Divisions of Cellular and Molecular Therapy and Pediatric Cardiology, College of Medicine, University of Florida, Gainesville, Florida; and
- Powell Gene Therapy Center, University of Florida, Gainesville, Florida
| | - B J Byrne
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, Florida
- Department of Pediatrics, Divisions of Cellular and Molecular Therapy and Pediatric Cardiology, College of Medicine, University of Florida, Gainesville, Florida; and
- Powell Gene Therapy Center, University of Florida, Gainesville, Florida
| | - D D Fuller
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, Florida;
- McKnight Brain Institute, University of Florida, Gainesville, Florida
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, Florida
| |
Collapse
|
6
|
Turner SMF, Hoyt AK, ElMallah MK, Falk DJ, Byrne BJ, Fuller DD. Neuropathology in respiratory-related motoneurons in young Pompe (Gaa(-/-)) mice. Respir Physiol Neurobiol 2016; 227:48-55. [PMID: 26921786 PMCID: PMC4880056 DOI: 10.1016/j.resp.2016.02.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/17/2016] [Accepted: 02/21/2016] [Indexed: 12/25/2022]
Abstract
Respiratory and/or lingual dysfunction are among the first motor symptoms in Pompe disease, a disorder resulting from absence or dysfunction of the lysosomal enzyme acid α-glucosidase (GAA). Here, we histologically evaluated the medulla, cervical and thoracic spinal cords in 6 weeks old asymptomatic Pompe (Gaa(-/-)) mice to determine if neuropathology in respiratory motor regions has an early onset. Periodic acid-Schiff (PAS) staining indicated glycogen accumulation was exclusively occurring in Gaa(-/-) hypoglossal, mid-cervical and upper thoracic motoneurons. Markers of DNA damage (Tunel) and ongoing apoptosis (Cleaved Caspase 3) did not co-localize with PAS staining, but were prominent in a medullary region which included the nucleus tractus solitarius, and also in the thoracic spinal dorsal horn. We conclude that respiratory-related motoneurons are particularly susceptible to GAA deficiency and that neuronal glycogen accumulation and neurodegeneration may occur independently in early stage disease. The data support early therapeutic intervention in Pompe disease.
Collapse
Affiliation(s)
- Sara M F Turner
- Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States
| | - Aaron K Hoyt
- Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States
| | - Mai K ElMallah
- Department of Pediatrics, Division of Pulmonary Medicine, University of Florida, Gainesville, FL 32610, United States
| | - Darin J Falk
- Department of Pediatrics, Child Health Research Institute, University of Florida, Gainesville, FL 32610, United States; Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, United States; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States
| | - Barry J Byrne
- Department of Pediatrics, Child Health Research Institute, University of Florida, Gainesville, FL 32610, United States; Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, United States; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States
| | - David D Fuller
- Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States.
| |
Collapse
|
7
|
Kawashita E, Tsuji D, Kanno Y, Tsuchida K, Itoh K. Enhancement by Uridine Diphosphate of Macrophage Inflammatory Protein-1 Alpha Production in Microglia Derived from Sandhoff Disease Model Mice. JIMD Rep 2015; 28:85-93. [PMID: 26545879 DOI: 10.1007/8904_2015_496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/26/2015] [Accepted: 09/08/2015] [Indexed: 11/27/2022] Open
Abstract
Sandhoff disease (SD) is a lysosomal β-hexosaminidase (Hex) deficiency involving excessive accumulation of undegraded substrates, including GM2 ganglioside, and progressive neurodegeneration. Macrophage inflammatory protein-1α (MIP-1α) is a crucial factor for microglia-mediated neuroinflammation in the onset or progression of SD. However, the transmitter-mediated production of MIP-1α in SD is still poorly understood.Extracellular nucleotides, including uridine diphosphate (UDP), leaked by either injured or damaged neuronal cells activate microglia to trigger chemotaxis, phagocytosis, macropinocytosis, and cytokine production.In this study, we demonstrated that UDP enhanced the production of MIP-1α by microglia derived from SD mice (SD-Mg), but not that from wild-type mice (WT-Mg). The UDP-induced MIP-1α production was mediated by the activation of P2Y6 receptor, ERK, and JNK. We also found the amount of dimeric P2Y6 receptor protein to have increased in SD-Mg in comparison to WT-Mg. In addition, we demonstrated that the disruption of lipid rafts enhanced the effect of UDP on MIP-1α production and the disordered maintenance of the lipid rafts in SD-Mg. Thus, the accumulation of undegraded substrates might cause the enhanced effect of UDP in SD-Mg through the increased expression of the dimeric P2Y6 receptors and the disordered maintenance of the lipid rafts. These findings provide new insights into the pathogenic mechanism and therapeutic strategies for SD.
Collapse
Affiliation(s)
- Eri Kawashita
- Department of Pathological Biochemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, Japan
| | - Daisuke Tsuji
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, 1-78 Sho-machi, Tokushima, 770-8505, Japan
| | - Yosuke Kanno
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto, Japan
| | - Kaho Tsuchida
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto, Japan
| | - Kohji Itoh
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, 1-78 Sho-machi, Tokushima, 770-8505, Japan.
| |
Collapse
|
8
|
BHBA suppresses LPS-induced inflammation in BV-2 cells by inhibiting NF-κB activation. Mediators Inflamm 2014; 2014:983401. [PMID: 24803746 PMCID: PMC3997897 DOI: 10.1155/2014/983401] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Revised: 12/23/2013] [Accepted: 01/06/2014] [Indexed: 12/15/2022] Open
Abstract
β-Hydroxybutyric acid (BHBA) has neuroprotective effects, but the underlying molecular mechanisms are unclear. Microglial activation plays an important role in neurodegenerative diseases by producing several proinflammatory enzymes and proinflammatory cytokines. The current study investigates the potential mechanisms whereby BHBA affects the expression of potentially proinflammatory proteins by cultured murine microglial BV-2 cells stimulated with lipopolysaccharide (LPS). The results showed that BHBA significantly reduced LPS-induced protein and mRNA expression levels of iNOS, COX-2, TNF-α, IL-1β, and IL-6. Blocking of GPR109A by PTX resulted in a loss of this anti-inflammatory effect in BV-2 cells. Western blot analysis showed that BHBA reduced LPS-induced degradation of IκB-α and translocation of NF-κB, while no effect was observed on MAPKs phosphorylation. All results imply that BHBA significantly reduces levels of proinflammatory enzymes and proinflammatory cytokines by inhibition of the NF-κB signaling pathway but not MAPKs pathways, and GPR109A is essential to this function. Overall, these data suggest that BHBA has a potential as neuroprotective drug candidate in neurodegenerative diseases.
Collapse
|
9
|
Archer LD, Langford-Smith KJ, Bigger BW, Fildes JE. Mucopolysaccharide diseases: a complex interplay between neuroinflammation, microglial activation and adaptive immunity. J Inherit Metab Dis 2014; 37:1-12. [PMID: 23653226 DOI: 10.1007/s10545-013-9613-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 04/16/2013] [Indexed: 12/23/2022]
Abstract
Mucopolysaccharide (MPS) diseases are lysosomal storage disorders (LSDs) caused by deficiencies in enzymes required for glycosaminoglycan (GAG) catabolism. Mucopolysaccharidosis I (MPS I), MPS IIIA, MPS IIIB and MPS VII are deficient in the enzymes α-L-Iduronidase, Heparan-N-Sulphatase, N-Acetylglucosaminidase and Beta-Glucuronidase, respectively. Enzyme deficiency leads to the progressive multi-systemic build-up of heparan sulphate (HS) and dermatan sulphate (DS) within cellular lysosomes, followed by cell, tissue and organ damage and in particular neurodegeneration. Clinical manifestations of MPS are well established; however as lysosomes represent vital components of immune cells, it follows that lysosomal accumulation of GAGs could affect diverse immune functions and therefore influence disease pathogenesis. Theoretically, MPS neurodegeneration and GAGs could be substantiating a threat of danger and damage to alert the immune system for cellular clearance, which due to the progressive nature of MPS storage would propagate disease pathogenesis. Innate immunity appears to have a key role in MPS; however the extent of adaptive immune involvement remains to be elucidated. The current literature suggests a complex interplay between neuroinflammation, microglial activation and adaptive immunity in MPS disease.
Collapse
Affiliation(s)
- Louise D Archer
- The Transplant Centre, UHSM, University of Manchester, Manchester, England, UK
| | | | | | | |
Collapse
|