1
|
Shafiek MS, Mekky RY, Nassar NN, El-Yamany MF, Rabie MA. Vortioxetine ameliorates experimental autoimmune encephalomyelitis model of multiple sclerosis in mice via activation of PI3K/Akt/CREB/BDNF cascade and modulation of serotonergic pathway signaling. Eur J Pharmacol 2024; 982:176929. [PMID: 39181226 DOI: 10.1016/j.ejphar.2024.176929] [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: 05/22/2024] [Revised: 08/01/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Multiple sclerosis (MS) is a chronic condition characterized by immune cell infiltration and cytokine overproduction that led to myelin sheath inflammatory assaults, thus causing axonal destruction. The former consequently provokes motor impairment and psychological disorders. Markedly, depression is one of the most prevalent lifelong comorbidities that negatively impacts the quality of life in MS patients. Vortioxetine (VTX), a multi-modal molecule prescribed to manage depression and anxiety disorder, additionally, it displays a promising neuroprotective properties against neurodegenerative diseases such as Alzheimer's and Parkinson's. To this end, the present study investigated the potential therapeutic efficacy of VTX against experimental autoimmune encephalomyelitis (EAE) model of MS in mice. Notably, treatment with VTX significantly ameliorated EAE-induced motor disability, as evident by enhanced performance in open field, rotarod and grip strength tests, alongside a reduction in immobility time during the forced swimming test, indicating a mitigation of the depressive-like behavior; outcomes that were corroborated with histological examinations and biochemical analyses. Mechanistically, VTX enhanced serotonin levels by inhibiting both serotonin transporter (SERT) and indoleamine 2,3-dioxygenase (IDO) enzyme, thereby promoting the activation of serotonin 1A (5-HT1A) receptor. The latter triggered the stimulation of phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) cascade that entailed activation/phosphorylation of cAMP response element-binding protein (CREB). This activation increased brain derived neurotrophic factor (BDNF) and myelin basic protein (MBP) contents that mitigated demyelination in the corpus callosum. Furthermore, VTX suppressed phospho serine 536 nuclear factor kappa B (pS536 NF-κB p65) activity and reduced tumor necrosis factor-alpha (TNF-α) production. The results underscore VTX's beneficial effects on disease severity in EAE model of MS in mice by amending both inflammatory and neurodegenerative components of MS progression.
Collapse
Affiliation(s)
- Marwa S Shafiek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Radwa Y Mekky
- Department of Pharmacology and Toxicology, October University for Modern Science and Arts (MSA), Giza, 12622, Egypt
| | - Noha N Nassar
- Department of Pharmacology and Toxicology, October University for Modern Science and Arts (MSA), Giza, 12622, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.
| |
Collapse
|
2
|
Emamnejad R, Pagnin M, Petratos S. The iron maiden: Oligodendroglial metabolic dysfunction in multiple sclerosis and mitochondrial signaling. Neurosci Biobehav Rev 2024; 164:105788. [PMID: 38950685 DOI: 10.1016/j.neubiorev.2024.105788] [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: 04/02/2024] [Revised: 06/06/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune disease, governed by oligodendrocyte (OL) dystrophy and central nervous system (CNS) demyelination manifesting variable neurological impairments. Mitochondrial mechanisms may drive myelin biogenesis maintaining the axo-glial unit according to dynamic requisite demands imposed by the axons they ensheath. The promotion of OL maturation and myelination by actively transporting thyroid hormone (TH) into the CNS and thereby facilitating key transcriptional and metabolic pathways that regulate myelin biogenesis is fundamental to sustain the profound energy demands at each axo-glial interface. Deficits in regulatory functions exerted through TH for these physiological roles to be orchestrated by mature OLs, can occur in genetic and acquired myelin disorders, whereby mitochondrial efficiency and eventual dysfunction can lead to profound oligodendrocytopathy, demyelination and neurodegenerative sequelae. TH-dependent transcriptional and metabolic pathways can be dysregulated during acute and chronic MS lesion activity depriving OLs from critical acetyl-CoA biochemical mechanisms governing myelin lipid biosynthesis and at the same time altering the generation of iron metabolism that may drive ferroptotic mechanisms, leading to advancing neurodegeneration.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| |
Collapse
|
3
|
Taheri R, Mokhtari Y, Yousefi AM, Bashash D. The PI3K/Akt signaling axis and type 2 diabetes mellitus (T2DM): From mechanistic insights into possible therapeutic targets. Cell Biol Int 2024; 48:1049-1068. [PMID: 38812089 DOI: 10.1002/cbin.12189] [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: 07/07/2023] [Revised: 02/03/2024] [Accepted: 05/12/2024] [Indexed: 05/31/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is an immensely debilitating chronic disease that progressively undermines the well-being of various bodily organs and, indeed, most patients succumb to the disease due to post-T2DM complications. Although there is evidence supporting the activation of the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway by insulin, which is essential in regulating glucose metabolism and insulin resistance, the significance of this pathway in T2DM has only been explored in a few studies. The current review aims to unravel the mechanisms by which different classes of PI3Ks control the metabolism of glucose; and also to discuss the original data obtained from international research laboratories on this topic. We also summarized the role of the PI3K/Akt signaling axis in target tissues spanning from the skeletal muscle to the adipose tissue and liver. Furthermore, inquiries regarding the impact of disrupting this axis on insulin function and the development of insulin resistance have been addressed. We also provide a general overview of the association of impaired PI3K/Akt signaling pathways in the pathogenesis of the most prevalent diabetes-related complications. The last section provides a special focus on the therapeutic potential of this axis by outlining the latest advances in active compounds that alleviate diabetes via modulation of the PI3K/Akt pathway. Finally, we comment on the future research aspects in which the field of T2DM therapies using PI3K modulators might be developed.
Collapse
Affiliation(s)
- Rana Taheri
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yazdan Mokhtari
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Mohammad Yousefi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Ge C, Liu D, Sun Y. The promotive effect of activation of the Akt/mTOR/p70S6K signaling pathway in oligodendrocytes on nerve myelin regeneration in rats with spinal cord injury. Br J Neurosurg 2024; 38:284-292. [PMID: 33345640 DOI: 10.1080/02688697.2020.1862056] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 01/22/2023]
Abstract
PURPOSE Akt/mTOR/p70S6K signaling pathway promotes motor function recovery after spinal cord injury (SCI) in both neurons and astrocytes. But the role and mechanism of this pathway in oligodendrocytes during nerve repair following SCI has not been researched. This study aimed to investigate the effect and mechanism of this signaling pathway in oligodendrocytes on nerve myelin regeneration and motor function recovery in rats with SCI. METHODS After inhibiting or activating this signaling pathway, Western blotting and double immunofluorescence labeling were used to determine the levels of the signaling molecules in this pathway and myelin formation-related proteins in the plane of the thoracic segment of the injured spinal cord. The level of motor function recovery was evaluated and the oligodendrocytes involved in nerve myelin regeneration were studied. Primary oligodendrocytes were isolated and cultured in vitro, then MBP, PLP, and MOG were measured with reverse transcription-quantitative polymerase chain reaction (RT-qPCR). RESULTS Akt/mTOR/p70S6K signaling pathway was activated after SCI compared with the sham-operated rats, prominently elevated levels of the pathway components were observed in the SC79-treated group. The activation of the signaling pathway significantly increased the expression levels of myelin formation-related proteins, including MBP, PLP, and MOG, and improved the Basso, Beattie, and Bresnahan (BBB) scores in the injured spinal cord. Conversely, rapamycin suppressed the expression of these signaling molecules and reduced the levels of myelin formation-related proteins. CONCLUSION Akt/mTOR/p70S6K signaling pathway activation can contribute to nerve myelin regeneration and has the potential to improve the regenerative environment and motor function, as well as the potential to promote repair of SCI.
Collapse
Affiliation(s)
- Chen Ge
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Orthopedics, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yongming Sun
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
5
|
Al-kuraishy HM, Jabir MS, Al-Gareeb AI, Saad HM, Batiha GES, Klionsky DJ. The beneficial role of autophagy in multiple sclerosis: Yes or No? Autophagy 2024; 20:259-274. [PMID: 37712858 PMCID: PMC10813579 DOI: 10.1080/15548627.2023.2259281] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/16/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic progressive demyelinating disease of the central nervous system (CNS) due to an increase of abnormal peripherally auto-reactive T lymphocytes which elicit autoimmunity. The main pathophysiology of MS is myelin sheath damage by immune cells and a defect in the generation of myelin by oligodendrocytes. Macroautophagy/autophagy is a critical degradation process that eliminates dysfunctional or superfluous cellular components. Autophagy has the property of a double-edged sword in MS in that it may have both beneficial and detrimental effects on MS neuropathology. Therefore, this review illustrates the protective and harmful effects of autophagy with regard to this disease. Autophagy prevents the progression of MS by reducing oxidative stress and inflammatory disorders. In contrast, over-activated autophagy is associated with the progression of MS neuropathology and in this case the use of autophagy inhibitors may alleviate the pathogenesis of MS. Furthermore, autophagy provokes the activation of different immune and supporting cells that play an intricate role in the pathogenesis of MS. Autophagy functions in the modulation of MS neuropathology by regulating cell proliferation related to demyelination and remyelination. Autophagy enhances remyelination by increasing the activity of oligodendrocytes, and astrocytes. However, autophagy induces demyelination by activating microglia and T cells. In conclusion, specific autophagic activators of oligodendrocytes, and astrocytes, and specific autophagic inhibitors of dendritic cells (DCs), microglia and T cells induce protective effects against the pathogenesis of MS.Abbreviations: ALS: amyotrophic lateral sclerosis; APCs: antigen-presenting cells; BBB: blood-brain barrier; CSF: cerebrospinal fluid; CNS: central nervous system; DCs: dendritic cells; EAE: experimental autoimmune encephalomyelitis; ER: endoplasmic reticulum; LAP: LC3-associated phagocytosis; MS: multiple sclerosis; NCA: non-canonical autophagy; OCBs: oligoclonal bands; PBMCs: peripheral blood mononuclear cells; PD: Parkinson disease; ROS: reactive oxygen species; UPR: unfolded protein response.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Iraq, Baghdad
| | - Majid S. Jabir
- Department of Applied Science, University of Technology, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Iraq, Baghdad
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, El Beheira, Egypt
| | | |
Collapse
|
6
|
Amore G, Calì E, Spanò M, Ceravolo G, Mangano GD, Scorrano G, Efthymiou S, Salpietro V, Houlden H, Di Rosa G. ATP6V1B2-related disorders featuring Lennox-Gastaut-syndrome: A case-based overview. Brain Dev 2023; 45:588-596. [PMID: 37633739 DOI: 10.1016/j.braindev.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 06/15/2023] [Accepted: 07/21/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND ATP6V1B2 (ATPase, H+ transporting, lysosomal VI subunit B, isoform 2) encodes for a subunit of a ubiquitous transmembrane lysosomal proton pump, implicated in the acidification of intracellular organelles and in several additional cellular functions. Variants in ATP6V1B2 have been related to a heterogeneous group of multisystemic disorders sometimes associated with variable neurological involvement. However, our knowledge of genotype-phenotype correlations and the neurological spectrum of ATP6V1B2-related disorders remain limited due to the few numbers of reported cases. CASE STUDY We hereby report the case of an 18-year-old male Sicilian patient affected by a global developmental delay, skeletal abnormalities, and epileptic encephalopathy featuring Lennox-Gastaut syndrome (LGS), in which exome sequencing led to the identification of a novel de novo variant in ATP6V1B2 (NM_001693.4: c.973G > C, p.Gly325Arg). CONCLUSIONS Our report provides new insights on the inclusion of developmental epileptic encephalopathies (DEEs) within the continuum group of ATP6V1B2-related disorders, expanding the phenotypic and molecular spectrum associated with these conditions.
Collapse
Affiliation(s)
- Greta Amore
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom; Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age "Gaetano Barresi", University of Messina, Via C. Valeria 1, 98125 Messina, Italy
| | - Elisa Calì
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Maria Spanò
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age "Gaetano Barresi", University of Messina, Via C. Valeria 1, 98125 Messina, Italy
| | - Giorgia Ceravolo
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom; Unit of Emergency Pediatrics, Department of Human Pathology and Evolutive Age "Gaetano Barresi", University of Messina, Via C. Valeria 1, 98125 Messina, Italy
| | - Giuseppe Donato Mangano
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Human Anatomy Section, Via del Vespro 129, 90127 Palermo, Italy
| | - Giovanna Scorrano
- Department of Pediatrics "G. D'Annunzio", University of Chieti-Pescara, Chieti, Italy
| | - Stephanie Efthymiou
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Vincenzo Salpietro
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom; Department of Pediatrics, University of L'Aquila, 67100 L'Aquila, Italy
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Gabriella Di Rosa
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age "Gaetano Barresi", University of Messina, Via C. Valeria 1, 98125 Messina, Italy.
| |
Collapse
|
7
|
Khan Z, Gupta GD, Mehan S. Cellular and Molecular Evidence of Multiple Sclerosis Diagnosis and Treatment Challenges. J Clin Med 2023; 12:4274. [PMID: 37445309 DOI: 10.3390/jcm12134274] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease that impacts the central nervous system and can result in disability. Although the prevalence of MS has increased in India, diagnosis and treatment continue to be difficult due to several factors. The present study examines the difficulties in detecting and treating multiple sclerosis in India. A lack of MS knowledge among healthcare professionals and the general public, which delays diagnosis and treatment, is one of the significant issues. Inadequate numbers of neurologists and professionals with knowledge of MS management also exacerbate the situation. In addition, MS medications are expensive and not covered by insurance, making them inaccessible to most patients. Due to the absence of established treatment protocols and standards for MS care, India's treatment techniques vary. In addition, India's population diversity poses unique challenges regarding genetic variations, cellular and molecular abnormalities, and the potential for differing treatment responses. MS is more difficult to accurately diagnose and monitor due to a lack of specialized medical supplies and diagnostic instruments. Improved awareness and education among healthcare professionals and the general public, as well as the development of standardized treatment regimens and increased investment in MS research and infrastructure, are required to address these issues. By addressing these issues, it is anticipated that MS diagnosis and treatment in India will improve, leading to better outcomes for those affected by this chronic condition.
Collapse
Affiliation(s)
- Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, IK Gujral Punjab Technical University, Jalandhar 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, IK Gujral Punjab Technical University, Jalandhar 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, IK Gujral Punjab Technical University, Jalandhar 144603, India
| |
Collapse
|
8
|
Devanand M, V N S, Madhu K. Signaling mechanisms involved in the regulation of remyelination in multiple sclerosis: a mini review. J Mol Med (Berl) 2023:10.1007/s00109-023-02312-9. [PMID: 37084092 DOI: 10.1007/s00109-023-02312-9] [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: 05/14/2022] [Revised: 02/22/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
Multiple sclerosis is an autoimmune neurodegenerative disease of the CNS that causes progressive disabilities, owing to CNS axon degeneration as a late result of demyelination. In the search for the prevention of axonal loss, mitigating inflammatory attacks in the CNS and myelin restoration are two possible approaches. As a result, therapies that target diverse signaling pathways involved in neuroprotection and remyelination have the potential to overcome the challenges in the development of multiple sclerosis treatments. LINGO1 (Leucine rich repeat and Immunoglobulin domain containing, Nogo receptor- interaction protein), AKT/PIP3/mTOR, Notch, Wnt, RXR (Retinoid X receptor gamma), and Nrf2 (nuclear factor erythroid 2-related factor 2) signaling pathways are highlighted in this section. This article reviews the present knowledge regarding numerous signaling pathways and their functions in regulating remyelination in multiple sclerosis pathogenesis. These pathways are potential biomarkers and therapeutic targets in MS.
Collapse
Affiliation(s)
- Midhuna Devanand
- Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Saiprabha V N
- Department of Pharmaceutical Chemistry and Analysis, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| | - Krishnadas Madhu
- Department of Pharmacology, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| |
Collapse
|
9
|
Kooshki L, Zarneshan SN, Fakhri S, Moradi SZ, Echeverria J. The pivotal role of JAK/STAT and IRS/PI3K signaling pathways in neurodegenerative diseases: Mechanistic approaches to polyphenols and alkaloids. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154686. [PMID: 36804755 DOI: 10.1016/j.phymed.2023.154686] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/10/2023] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Neurodegenerative diseases (NDDs) are characterized by progressive neuronal dysfunctionality which results in disability and human life-threatening events. In recent decades, NDDs are on the rise. Besides, conventional drugs have not shown potential effectiveness to attenuate the complications of NDDs. So, exploring novel therapeutic agents is an urgent need to combat such disorders. Accordingly, growing evidence indicates that polyphenols and alkaloids are promising natural candidates, possessing several beneficial pharmacological effects against diseases. Considering the complex pathophysiological mechanisms behind NDDs, Janus kinase (JAK), insulin receptor substrate (IRS), phosphoinositide 3-kinase (PI3K), and signal transducer and activator of transcription (STAT) seem to play critical roles during neurodegeneration/neuroregeneration. In this line, modulation of the JAK/STAT and IRS/PI3K signaling pathways and their interconnected mediators by polyphenols/alkaloids could play pivotal roles in combating NDDs, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), stroke, aging, multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), depression and other neurological disorders. PURPOSE Thus, the present study aimed to investigate the neuroprotective roles of polyphenols/alkaloids as multi-target natural products against NDDs which are critically passing through the modulation of the JAK/STAT and IRS/PI3K signaling pathways. STUDY DESIGN AND METHODS A systematic and comprehensive review was performed to highlight the modulatory roles of polyphenols and alkaloids on the JAK/STAT and IRS/PI3K signaling pathways in NDDs, according to the PRISMA guideline, using scholarly electronic databases, including Scopus, PubMed, ScienceDirect, and associated reference lists. RESULTS In the present study 141 articles were included from a total of 1267 results. The results showed that phenolic compounds such as curcumin, epigallocatechin-3-gallate, and quercetin, and alkaloids such as berberine could be introduced as new strategies in combating NDDs through JAK/STAT and IRS/PI3K signaling pathways. This is the first systematic review that reveals the correlation between the JAK/STAT and IRS/PI3K axis which is targeted by phytochemicals in NDDs. Hence, this review highlighted promising insights into the neuroprotective potential of polyphenols and alkaloids through the JAK/STAT and IRS/PI3K signaling pathway and interconnected mediators toward neuroprotection. CONCLUSION Amongst natural products, phenolic compounds and alkaloids are multi-targeting agents with the most antioxidants and anti-inflammatory effects possessing the potential of combating NDDs with high efficacy and lower toxicity. However, additional reports are needed to prove the efficacy and possible side effects of natural products.
Collapse
Affiliation(s)
- Leila Kooshki
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Javier Echeverria
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|
10
|
Redenšek Trampuž S, Vogrinc D, Goričar K, Dolžan V. Shared miRNA landscapes of COVID-19 and neurodegeneration confirm neuroinflammation as an important overlapping feature. Front Mol Neurosci 2023; 16:1123955. [PMID: 37008787 PMCID: PMC10064073 DOI: 10.3389/fnmol.2023.1123955] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/20/2023] [Indexed: 03/19/2023] Open
Abstract
Introduction Development and worsening of most common neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis, have been associated with COVID-19 However, the mechanisms associated with neurological symptoms in COVID-19 patients and neurodegenerative sequelae are not clear. The interplay between gene expression and metabolite production in CNS is driven by miRNAs. These small non-coding molecules are dysregulated in most common neurodegenerative diseases and COVID-19. Methods We have performed a thorough literature screening and database mining to search for shared miRNA landscapes of SARS-CoV-2 infection and neurodegeneration. Differentially expressed miRNAs in COVID-19 patients were searched using PubMed, while differentially expressed miRNAs in patients with five most common neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and multiple sclerosis) were searched using the Human microRNA Disease Database. Target genes of the overlapping miRNAs, identified with the miRTarBase, were used for the pathway enrichment analysis performed with Kyoto Encyclopedia of Genes and Genomes and Reactome. Results In total, 98 common miRNAs were found. Additionally, two of them (hsa-miR-34a and hsa-miR-132) were highlighted as promising biomarkers of neurodegeneration, as they are dysregulated in all five most common neurodegenerative diseases and COVID-19. Additionally, hsa-miR-155 was upregulated in four COVID-19 studies and found to be dysregulated in neurodegeneration processes as well. Screening for miRNA targets identified 746 unique genes with strong evidence for interaction. Target enrichment analysis highlighted most significant KEGG and Reactome pathways being involved in signaling, cancer, transcription and infection. However, the more specific identified pathways confirmed neuroinflammation as being the most important shared feature. Discussion Our pathway based approach has identified overlapping miRNAs in COVID-19 and neurodegenerative diseases that may have a valuable potential for neurodegeneration prediction in COVID-19 patients. Additionally, identified miRNAs can be further explored as potential drug targets or agents to modify signaling in shared pathways. Graphical AbstractShared miRNA molecules among the five investigated neurodegenerative diseases and COVID-19 were identified. The two overlapping miRNAs, hsa-miR-34a and has-miR-132, present potential biomarkers of neurodegenerative sequelae after COVID-19. Furthermore, 98 common miRNAs between all five neurodegenerative diseases together and COVID-19 were identified. A KEGG and Reactome pathway enrichment analyses was performed on the list of shared miRNA target genes and finally top 20 pathways were evaluated for their potential for identification of new drug targets. A common feature of identified overlapping miRNAs and pathways is neuroinflammation. AD, Alzheimer's disease; ALS, amyotrophic lateral sclerosis; COVID-19, coronavirus disease 2019; HD, Huntington's disease; KEGG, Kyoto Encyclopedia of Genes and Genomes; MS, multiple sclerosis; PD, Parkinson's disease.
Collapse
Affiliation(s)
| | | | | | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
11
|
Dulaglutide Improves Gliosis and Suppresses Apoptosis/Autophagy Through the PI3K/Akt/mTOR Signaling Pathway in Vascular Dementia Rats. Neurochem Res 2022; 48:1561-1579. [PMID: 36571662 DOI: 10.1007/s11064-022-03853-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/11/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Dulaglutide is a new type of hypoglycemic agent that agonizes glucagon-like peptide-1 receptor (GLP-1RA). It can be concluded from previous studies that a GLP-1RA can reduce apoptosis and regulate autophagy in the nervous system, while related research on dulaglutide in vascular dementia (VD) has not been reported. In our study, the VD rat model was established by bilateral carotid artery occlusion, and the results of the Morris water maze test (MWM) and open-field test showed that the application of dulaglutide could effectively reduce the cognitive decline of VD rats without changing the behavior in the open-field test, which was used to assess an anxiety-like phenotype. We applied HE staining and immunofluorescence labeling to show that dulaglutide treatment significantly alleviated neuronal damage in the hippocampal region of VD rats, and reduced microglial and astrocyte proliferation. Western blot results showed that dulaglutide reduced VD-induced neuronal apoptosis (BCL2/BAX, c-caspase3) and autophagy (P62, LC3B, Beclin-1), and upregulated phosphorylation of PI3K/Akt/mTOR signaling pathway. KEGG pathway analysis of RNA-Sequence results showed that the differentially expressed genes in the dulaglutide treatment group were significantly enriched in the mTOR signaling pathway, and the repressor of mTOR, Deptor, was down-regulated. In conclusion, this study suggested that dulaglutide may alleviate learning and memory impairment and neuron damage in VD rats by attenuating apoptosis, regulating autophagy, and activating the PI3K/Akt/mTOR signaling pathway in neurons, which may make it a promising candidate for the simultaneous treatment of VD and diabetes.
Collapse
|
12
|
Burkard T, Herrero San Juan M, Dreis C, Kiprina A, Namgaladze D, Siebenbrodt K, Luger S, Foerch C, Pfeilschifter JM, Weigert A, Radeke HH. Differential expression of CD8 defines phenotypically distinct cytotoxic T cells in cancer and multiple sclerosis. Clin Transl Med 2022; 12:e1068. [PMID: 36504430 PMCID: PMC9742381 DOI: 10.1002/ctm2.1068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cytotoxic T lymphocytes take on a leading role in many immune-related diseases. They function as key effector immune cells fighting cancer cells, but they are also considerably involved in autoimmune diseases. Common to both situations, CD8+ T cells need to adapt their metabolism and effector function to the harsh and nutrient-deprived conditions of the disease-associated microenvironment. METHODS We used an in vitro starvation as well as rapamycin treatment protocol mimicking nutrient deprivation to generate CD8Low versus CD8High T cells and performed FACS-Sorting followed by transcriptomic profiling of the cytotoxic T cell subsets. Prominent markers identified in the CD8Low versus the CD8High T cells were then used to investigate the presence of these cell subsets in immune-related human diseases. Employing cancer tissue microarrays and PhenOptics multispectral imaging as well as flow cytometry, we studied these CD8+ T cell subsets in cancer and relapsing-remitting multiple sclerosis patients. RESULTS Starvation induced a decreased expression of CD8, yielding a CD8Low T cell subpopulation with an altered transcriptomic signature and reduced effector function. CD8Low T cell showed enhanced ST2L and IL6ST (CD130) expression compared to CD8High T cells which expressed elevated KLRD1 (CD94) and granzyme B levels within the tumour microenvironment (TME). Spatial analysis revealed the presence of CD8High T cells in close proximity to tumour cells, while the CD8Low T cells resided at the tumour boundaries. Importantly, the number of tumour-infiltrating CD8Low T lymphocytes correlated with a poor prognosis as well as with enhanced cancer progression in human mammary carcinoma. We also found a reduced frequency of CD8Low T lymphocytes in a cohort of relapse (disease active) multiple sclerosis patients compared to healthy subjects during immune cell starvation in vitro. CONCLUSIONS In summary, our data show that functionally distinct cytotoxic T lymphocytes can be identified based on their expression of CD8. Indicating a more general role in CD8 T cell immunity, these cells may play opposing roles in the TME, and also in the pathophysiology of autoimmune diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Tobias Burkard
- Pharmazentrum Frankfurt/ZAFESInstitute of Pharmacology and ToxicologyHospital of the Goethe UniversityFrankfurt am MainGermany
| | - Martina Herrero San Juan
- Pharmazentrum Frankfurt/ZAFESInstitute of Pharmacology and ToxicologyHospital of the Goethe UniversityFrankfurt am MainGermany
| | - Caroline Dreis
- Pharmazentrum Frankfurt/ZAFESInstitute of Pharmacology and ToxicologyHospital of the Goethe UniversityFrankfurt am MainGermany
| | - Anastasiia Kiprina
- Faculty of MedicineInstitute of Biochemistry IGoethe‐University Frankfurt/MainFrankfurt am MainGermany
| | - Dmitry Namgaladze
- Faculty of MedicineInstitute of Biochemistry IGoethe‐University Frankfurt/MainFrankfurt am MainGermany
| | - Kai Siebenbrodt
- Department of NeurologyGoethe University Hospital FrankfurtFrankfurt am MainGermany
- Epilepsy Center Frankfurt Rhine‐MainDepartment of NeurologyUniversity Hospital Frankfurt, Frankfurt, Germany
| | - Sebastian Luger
- Department of NeurologyGoethe University Hospital FrankfurtFrankfurt am MainGermany
| | - Christian Foerch
- Department of NeurologyGoethe University Hospital FrankfurtFrankfurt am MainGermany
| | - Josef M. Pfeilschifter
- Pharmazentrum Frankfurt/ZAFESInstitute of Pharmacology and ToxicologyHospital of the Goethe UniversityFrankfurt am MainGermany
| | - Andreas Weigert
- Faculty of MedicineInstitute of Biochemistry IGoethe‐University Frankfurt/MainFrankfurt am MainGermany
- Frankfurt Cancer InstituteGoethe‐University FrankfurtFrankfurtGermany
- Cardio‐Pulmonary Institute (CPI)FrankfurtGermany
| | - Heinfried H. Radeke
- Pharmazentrum Frankfurt/ZAFESInstitute of Pharmacology and ToxicologyHospital of the Goethe UniversityFrankfurt am MainGermany
| |
Collapse
|
13
|
Sun W, Wen M, Liu M, Wang Q, Liu Q, Li L, Siebert HC, Loers G, Zhang R, Zhang N. Effect of β-hydroxybutyrate on behavioral alterations, molecular and morphological changes in CNS of multiple sclerosis mouse model. Front Aging Neurosci 2022; 14:1075161. [PMID: 36533180 PMCID: PMC9752847 DOI: 10.3389/fnagi.2022.1075161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/14/2022] [Indexed: 09/29/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and degenerative disease of central nervous system (CNS). Aging is the most significant risk factor for the progression of MS. Dietary modulation (such as ketogenic diet) and caloric restriction, can increase ketone bodies, especially β-hydroxybutyrate (BHB). Increased BHB has been reported to prevent or improve age-related disease. The present studies were performed to understand the therapeutic effect and potential mechanisms of exogenous BHB in cuprizone (CPZ)-induced demyelinating model. In this study, a continuous 35 days CPZ mouse model with or without BHB was established. The changes of behavior function, pathological hallmarks of CPZ, and intracellular signal pathways in mice were detected by Open feld test, Morris water maze, RT-PCR, immuno-histochemistry, and western blot. The results showed that BHB treatment improved behavioral performance, prevented myelin loss, decreased the activation of astrocyte as well as microglia, and up-regulated the neurotrophin brain-derived neurotrophic factor in both the corpus callosum and hippocampus. Meanwhile, BHB treatment increased the number of MCT1+ cells and APC+ oligodendrocytes. Furthermore, the treatment decreased the expression of HDAC3, PARP1, AIF and TRPA1 which is related to oligodendrocyte (OL) apoptosis in the corpus callosum, accompanied by increased expression of TrkB. This leads to an increased density of doublecortin (DCX)+ neuronal precursor cells and mature NeuN+ neuronal cells in the hippocampus. As a result, BHB treatment effectively promotes the generation of PDGF-Ra+ (oligodendrocyte precursor cells, OPCs), Sox2+ cells and GFAP+ (astrocytes), and decreased the production of GFAP+ TRAP1+ cells, and Oligo2+ TRAP1+ cells in the corpus callosum of mouse brain. Thus, our results demonstrate that BHB treatment efficiently supports OPC differentiation and decreases the OLs apoptosis in CPZ-intoxicated mice, partly by down-regulating the expression of TRPA1 and PARP, which is associated with the inhibition of the p38-MAPK/JNK/JUN pathway and the activation of ERK1/2, PI3K/AKT/mTOR signaling, supporting BHB treatment adjunctive nutritional therapy for the treatment of chronic demyelinating diseases, such as multiple sclerosis (MS).
Collapse
Affiliation(s)
- Wei Sun
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Min Wen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Min Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Quiqin Liu
- Shandong Donkey Industry, Technology Collaborative Innovation Center, Liaocheng University, Liaocheng, China
| | - Lanjie Li
- Shandong Donkey Industry, Technology Collaborative Innovation Center, Liaocheng University, Liaocheng, China
| | - Hans-Christian Siebert
- Schauenburgerstr, RI-B-NT - Research Institute of Bioinformatics and Nanotechnology, Kiel University, Kiel, Germany
| | - Gabriele Loers
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg, Germany
| | - Ruiyan Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
- Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
14
|
Hu J, Baydyuk M, Huang JK. Impact of amino acids on microglial activation and CNS remyelination. Curr Opin Pharmacol 2022; 66:102287. [PMID: 36067684 DOI: 10.1016/j.coph.2022.102287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/27/2022]
Abstract
Amino acids and their derivatives function as building blocks as well as signaling molecules to modulate various cellular processes in living organisms. In mice, amino acids accumulate in demyelinated lesions and return to basal levels during remyelination. Studies have found that amino acids and their metabolites modulate immune activity in the central nervous system (CNS) and influence oligodendrocyte differentiation and remyelination efficiency. In this review, we discuss current studies on amino acid metabolism in the context of CNS remyelination. By understanding the mechanisms of amino acid signaling and metabolism in demyelinated lesions, we may deepen our understanding of compartmentalized CNS inflammation in demyelinating disease like multiple sclerosis (MS) and provide evidence to develop novel pharmacological therapies targeting amino acid metabolism to prevent disease worsening.
Collapse
Affiliation(s)
- Jingwen Hu
- Department of Biology, Georgetown University, 37th and O St., NW, Washington, DC, 20057, USA
| | - Maryna Baydyuk
- Department of Biology, Georgetown University, 37th and O St., NW, Washington, DC, 20057, USA; Center for Cell Reprogramming, Georgetown University Medical Center, 37th and O St., NW, Washington, DC, 20057, USA
| | - Jeffrey K Huang
- Department of Biology, Georgetown University, 37th and O St., NW, Washington, DC, 20057, USA; Center for Cell Reprogramming, Georgetown University Medical Center, 37th and O St., NW, Washington, DC, 20057, USA.
| |
Collapse
|
15
|
Emamnejad R, Dass M, Mahlis M, Bozkurt S, Ye S, Pagnin M, Theotokis P, Grigoriadis N, Petratos S. Thyroid hormone-dependent oligodendroglial cell lineage genomic and non-genomic signaling through integrin receptors. Front Pharmacol 2022; 13:934971. [PMID: 36133808 PMCID: PMC9483185 DOI: 10.3389/fphar.2022.934971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous autoimmune disease whereby the pathological sequelae evolve from oligodendrocytes (OLs) within the central nervous system and are targeted by the immune system, which causes widespread white matter pathology and results in neuronal dysfunction and neurological impairment. The progression of this disease is facilitated by a failure in remyelination following chronic demyelination. One mediator of remyelination is thyroid hormone (TH), whose reliance on monocarboxylate transporter 8 (MCT8) was recently defined. MCT8 facilitates the entry of THs into oligodendrocyte progenitor cell (OPC) and pre-myelinating oligodendrocytes (pre-OLs). Patients with MS may exhibit downregulated MCT8 near inflammatory lesions, which emphasizes an inhibition of TH signaling and subsequent downstream targeted pathways such as phosphoinositide 3-kinase (PI3K)-Akt. However, the role of the closely related mammalian target of rapamycin (mTOR) in pre-OLs during neuroinflammation may also be central to the remyelination process and is governed by various growth promoting signals. Recent research indicates that this may be reliant on TH-dependent signaling through β1-integrins. This review identifies genomic and non-genomic signaling that is regulated through mTOR in TH-responsive pre-OLs and mature OLs in mouse models of MS. This review critiques data that implicates non-genomic Akt and mTOR signaling in response to TH-dependent integrin receptor activation in pre-OLs. We have also examined whether this can drive remyelination in the context of neuroinflammation and associated sequelae. Importantly, we outline how novel therapeutic small molecules are being designed to target integrin receptors on oligodendroglial lineage cells and whether these are viable therapeutic options for future use in clinical trials for MS.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Michael Mahlis
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Salome Bozkurt
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Paschalis Theotokis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
- *Correspondence: Steven Petratos,
| |
Collapse
|
16
|
Integrated Approaches to Identify miRNA Biomarkers Associated with Cognitive Dysfunction in Multiple Sclerosis Using Text Mining, Gene Expression, Pathways, and GWAS. Diagnostics (Basel) 2022; 12:diagnostics12081914. [PMID: 36010264 PMCID: PMC9406323 DOI: 10.3390/diagnostics12081914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS), a chronic autoimmune disorder, affects the central nervous system of many young adults. More than half of MS patients develop cognition problems. Although several genomic and transcriptomic studies are currently reported in MS cognitive impairment, a comprehensive repository dealing with all the experimental data is still underdeveloped. In this study, we combined text mining, gene regulation, pathway analysis, and genome-wide association studies (GWAS) to identify miRNA biomarkers to explore the cognitive dysfunction in MS, and to understand the genomic etiology of the disease. We first identified the dysregulated miRNAs associated with MS and cognitive dysfunction using PubTator (text mining), HMDD (experimental associations), miR2Disease, and PhenomiR database (differentially expressed miRNAs). Our results suggest that miRNAs such as hsa-mir-148b-3p, hsa-mir-7b-5p, and hsa-mir-7a-5p are commonly associated with MS and cognitive dysfunction. Next, we retrieved GWAS signals from GWAS Catalog, and analyzed the enrichment analysis of association signals in genes/miRNAs and their association networks. Then, we identified susceptible genetic loci, rs17119 (chromosome 6; p = 1 × 10−10), rs1843938 (chromosome 7; p = 1 × 10−10), and rs11637611 (chromosome 15; p = 1.00 × 10−15), associated with significant genetic risk. Lastly, we conducted a pathway analysis for the susceptible genetic variants and identified novel risk pathways. The ECM receptor signaling pathway (p = 3.98 × 10−8) and PI3K/Akt signaling pathway (p = 5.98 × 10−5) were found to be associated with differentially expressed miRNA biomarkers.
Collapse
|
17
|
Akbari M, Eshghyar F, Gholipour M, Eslami S, Hussen BM, Taheri M, Omrani MD, Ghafouri-Fard S. Expression analysis of mTOR-associated lncRNAs in multiple sclerosis. Metab Brain Dis 2022; 37:2061-2066. [PMID: 35622264 DOI: 10.1007/s11011-022-01010-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/16/2022] [Indexed: 10/18/2022]
Abstract
mTOR has been shown to be involved in the regulation of immune responses and differentiation of immune cells. This protein is a candidate molecule for unraveling the molecular mechanisms of autoimmune disorders such as multiple sclerosis (MS). We designed the current study to assess expression of MTOR, and four associated long non-coding RNAs (lncRNAs), namely SNHG1, SNHG3, SHNG5 and DANCR in the peripheral blood of patients with MS compared with healthy controls. Analysis of real-time PCR results has shown down-regulation of SNHG5 and DANCR in MS patients compared with controls. Sex of study participants had no significant effect on expression of either genes and the interaction of sex and disease on expression levels of all studied genes were insignificant. There was a significant negative correlation between expression levels of MTOR gene and disease duration. No other significant correlations were detected between genes expressions and clinical/demographic data. SNHG5 and DANCR transcript levels had AUC values of 0.88 and 0.68 in separation of patients with MS from healthy controls, respectively. Taken together, our study suggests participation of two mTOR-related lncRNAs, i.e. SNHG5 and DANCR in the pathophysiology of MS.
Collapse
Affiliation(s)
- Mohammadarian Akbari
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Eshghyar
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholipour
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Solat Eslami
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region, Iraq
| | - Mohammad Taheri
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Institute of Human Genetics, Jena University Hospital, Jena, Germany.
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Basile MS, Bramanti P, Mazzon E. The Role of Cytotoxic T-Lymphocyte Antigen 4 in the Pathogenesis of Multiple Sclerosis. Genes (Basel) 2022; 13:genes13081319. [PMID: 35893056 PMCID: PMC9394409 DOI: 10.3390/genes13081319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 02/05/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune neurodegenerative disorder of the central nervous system that presents heterogeneous clinical manifestations and course. It has been shown that different immune checkpoints, including Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4), can be involved in the pathogenesis of MS. CTLA-4 is a critical regulator of T-cell homeostasis and self-tolerance and represents a key inhibitor of autoimmunity. In this scopingreview, we resume the current preclinical and clinical studies investigating the role of CTLA-4 in MS with different approaches. While some of these studies assessed the expression levels of CTLA-4 on T cells by comparing MS patients with healthy controls, others focused on the evaluation of the effects of common MS therapies on CTLA-4 modulation or on the study of the CTLA-4 blockade or deficiency in experimental autoimmune encephalomyelitis models. Moreover, other studies in this field aimed to discover if the CTLA-4 gene might be involved in the predisposition to MS, whereas others evaluated the effects of treatment with CTLA4-Ig in MS. Although these results are of great interest, they are often conflicting. Therefore, further studies are needed to reveal the exact mechanisms underlying the action of a crucial immune checkpoint such as CTLA-4 in MS to identify novel immunotherapeutic strategies for MS patients.
Collapse
|
19
|
Petralia MC, Mangano K, Quattropani MC, Lenzo V, Nicoletti F, Fagone P. Computational Analysis of Pathogenetic Pathways in Alzheimer’s Disease and Prediction of Potential Therapeutic Drugs. Brain Sci 2022; 12:brainsci12070827. [PMID: 35884634 PMCID: PMC9313152 DOI: 10.3390/brainsci12070827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Background. Alzheimer’s disease (AD) is a chronic and progressive neurodegenerative disease which affects more than 50 million patients and represents 60–80% of all cases of dementia. Mutations in the APP gene, mostly affecting the γ-secretase site of cleavage and presenilin mutations, have been identified in inherited forms of AD. Methods. In the present study, we performed a meta-analysis of the transcriptional signatures that characterize two familial AD mutations (APPV7171F and PSEN1M146V) in order to characterize the common altered biomolecular pathways affected by these mutations. Next, an anti-signature perturbation analysis was performed using the AD meta-signature and the drug meta-signatures obtained from the L1000 database, using cosine similarity as distance metrics. Results. Overall, the meta-analysis identified 1479 differentially expressed genes (DEGs), 684 downregulated genes, and 795 upregulated genes. Additionally, we found 14 drugs with a significant anti-similarity to the AD signature, with the top five drugs being naftifine, moricizine, ketoconazole, perindopril, and fexofenadine. Conclusions. This study aimed to integrate the transcriptional profiles associated with common familial AD mutations in neurons in order to characterize the pathogenetic mechanisms involved in AD and to find more effective drugs for AD.
Collapse
Affiliation(s)
- Maria Cristina Petralia
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (K.M.); (P.F.)
| | | | - Vittorio Lenzo
- Department of Social and Educational Sciences of the Mediterranean Area, University for Foreigners “Dante Alighieri” of Reggio Calabria, 89125 Reggio Calabria, Italy;
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (K.M.); (P.F.)
- Correspondence:
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (K.M.); (P.F.)
| |
Collapse
|
20
|
Neuroprotective Effect of Chrysophanol as a PI3K/AKT/mTOR Signaling Inhibitor in an Experimental Model of Autologous Blood-induced Intracerebral Hemorrhage. Curr Med Sci 2022; 42:249-266. [PMID: 35079960 DOI: 10.1007/s11596-022-2496-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/29/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Intracerebral hemorrhage (ICH) refers to predominant, sporadic, and non-traumatic bleeding in the brain parenchyma. The PI3K/AKT/mTOR signaling pathway is an important signal transduction pathway regulated by enzyme-linked receptors and has many biological functions in mammals. It plays a key role in neuronal metabolism, gene expression regulation, and tissue homeostasis in the healthy and diseased brain. METHODS In the present study, the role of the PI3K/AKT/mTOR pathway inhibitor chrysophanol (CPH) (10 mg/kg and 20 mg/kg, orally) in the improvement of ICH-associated neurological defects in rats was investigated. Autologous blood (20 µL/5 min/unilateral/intracerebroventricular) mimics ICH-like defects involving cellular and molecular dysfunction and neurotransmitter imbalance. The current study also included various behavioral assessments to examine cognition, memory, and motor and neuromuscular coordination. The protein expression levels of PI3K, AKT, and mTOR as well as myelin basic protein and apoptotic markers, such as Bax, Bcl-2, and caspase-3, were examined using ELISA kits. Furthermore, the levels of various neuroinflammatory cytokines and oxidative stress markers were assessed. Additionally, the neurological severity score, brain water content, gross brain pathology, and hematoma size were used to indicate neurological function and brain edema. RESULTS CPH was found to be neuroprotective by restoring neurobehavioral alterations and significantly reducing the elevated PI3K, AKT, and mTOR protein levels, and modulating the apoptotic markers such as Bax, Bcl-2, and caspase-3 in rat brain homogenate. CPH substantially reduced the inflammatory cytokines like interleukin (IL)-1β, IL-6, and tumor necrosis factor-α. CPH administration restored the neurotransmitters GABA, glutamate, acetylcholine, dopamine, and various oxidative stress markers. CONCLUSION Our results show that CPH may be a promising therapeutic approach for overcoming neuronal damage caused by the overexpression of the PI3K/AKT/mTOR signaling pathway in ICH-induced neurological dysfunctions in rats.
Collapse
|
21
|
Brocchi A, Rebelos E, Dardano A, Mantuano M, Daniele G. Effects of Intermittent Fasting on Brain Metabolism. Nutrients 2022; 14:nu14061275. [PMID: 35334932 PMCID: PMC8954770 DOI: 10.3390/nu14061275] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 01/27/2023] Open
Abstract
We are facing an obesity epidemic, and obesity itself and its close companion, type 2 diabetes, are independent risk factors for neurodegeneration. While most medical treatments fail to induce a clinically meaningful improvement in neurodegenerative disorders, lifestyle interventions have emerged in the spotlight. A recently rediscovered approach is intermittent fasting (IF), which, compared to the classic caloric restriction regimens, limits only the time of eating, rather than the number of calories allowed per day. There is already a large amount of evidence from preclinical and clinical studies showing the beneficial effects of IF. In this review, we specifically focus on the effects of IF on brain metabolism. Key molecular players modified during IF and involved in its beneficial central effects (ketone bodies, BDNF, GABA, GH/IGF-1, FGF2, sirtuin-3, mTOR, and gut microbiota) are identified and discussed. Studies suggest that IF induces several molecular and cellular adaptations in neurons, which, overall, enhance cellular stress resistance, synaptic plasticity, and neurogenesis. Still, the absence of guidelines regarding the application of IF to patients hampers its broad utilization in clinical practice, and further studies are needed to improve our knowledge on the different IF protocols and long-term effects of IF on brain metabolism before it can be widely prescribed.
Collapse
Affiliation(s)
- Alex Brocchi
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy; (A.B.); (A.D.); (M.M.)
| | - Eleni Rebelos
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy;
| | - Angela Dardano
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy; (A.B.); (A.D.); (M.M.)
| | - Michele Mantuano
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy; (A.B.); (A.D.); (M.M.)
| | - Giuseppe Daniele
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy; (A.B.); (A.D.); (M.M.)
- Correspondence: ; Tel.: +39-3404618257
| |
Collapse
|
22
|
Movahedpour A, Vakili O, Khalifeh M, Mousavi P, Mahmoodzadeh A, Taheri-Anganeh M, Razmeh S, Shabaninejad Z, Yousefi F, Behrouj H, Ghasemi H, Khatami SH. Mammalian target of rapamycin (mTOR) signaling pathway and traumatic brain injury: A novel insight into targeted therapy. Cell Biochem Funct 2022; 40:232-247. [PMID: 35258097 DOI: 10.1002/cbf.3692] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 11/11/2022]
Abstract
Traumatic brain injury (TBI) is one of the most concerning health issues in which the normal brain function may be disrupted as a result of a blow, bump, or jolt to the head. Loss of consciousness, amnesia, focal neurological defects, alteration in mental state, and destructive diseases of the nervous system such as cognitive impairment, Parkinson's, and Alzheimer's disease. Parkinson's disease is a chronic progressive neurodegenerative disorder, characterized by the early loss of striatal dopaminergic neurons. TBI is a major risk factor for Parkinson's disease. Existing therapeutic approaches have not been often effective, indicating the necessity of discovering more efficient therapeutic targets. The mammalian target of rapamycin (mTOR) signaling pathway responds to different environmental cues to modulate a large number of cellular processes such as cell proliferation, survival, protein synthesis, autophagy, and cell metabolism. Moreover, mTOR has been reported to affect the regeneration of the injured nerves throughout the central nervous system (CNS). In this context, recent evaluations have revealed that mTOR inhibitors could be potential targets to defeat a group of neurological disorders, and thus, a number of clinical trials are investigating their efficacy in treating dementia, autism, epilepsy, stroke, and brain injury, as irritating neurological defects. The current review describes the interplay between mTOR signaling and major CNS-related disorders (esp. neurodegenerative diseases), as well as the mTOR signaling-TBI relationship. It also aims to discuss the promising therapeutic capacities of mTOR inhibitors during the TBI.
Collapse
Affiliation(s)
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoomeh Khalifeh
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pegah Mousavi
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Amir Mahmoodzadeh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Saeed Razmeh
- Department of Internal Medicine, School of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Zahra Shabaninejad
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Yousefi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamid Behrouj
- Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | | | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Basile MS, Cavalli E, McCubrey J, Hernández-Bello J, Muñoz-Valle JF, Fagone P, Nicoletti F. The PI3K/Akt/mTOR pathway: A potential pharmacological target in COVID-19. Drug Discov Today 2022; 27:848-856. [PMID: 34763066 PMCID: PMC8574122 DOI: 10.1016/j.drudis.2021.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/24/2021] [Accepted: 11/01/2021] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has emerged as a serious threat to global health. The disregulation of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) cell signaling pathway observed in patients with COVID-19 has attracted attention for the possible use of specific inhibitors of this pathway for the treatment of the disease. Here, we review emerging data on the involvement of the PI3K/Akt/mTOR pathway in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the clinical studies investigating its tailored inhibition in COVID-19. Current in silico, in vitro, and in vivo data convergently support a role for the PI3K/Akt/mTOR pathway in COVID-19 and suggest the use of specific inhibitors of this pathway that, by a combined mechanism entailing downregulation of excessive inflammatory reactions, cell protection, and antiviral effects, could ameliorate the course of COVID-19.
Collapse
Affiliation(s)
- Maria Sofia Basile
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy
| | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - James McCubrey
- Department of Microbiology and Immunology, Brody Medical Sciences Building, East Carolina University, Greenville, NC 27834, USA
| | - Jorge Hernández-Bello
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud Universidad de Guadalajara, 44340 Guadalajara, Mexico
| | - José Francisco Muñoz-Valle
- University Center for Health Science, Department of Molecular Biology and Genomics, University of Guadalajara, Jalisco 49000, Mexico
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy,Corresponding author
| |
Collapse
|
24
|
Jadaun KS, Mehan S, Sharma A, Siddiqui EM, Kumar S, Alsuhaymi N. Neuroprotective Effect of Chrysophanol as a PI3K/AKT/mTOR Signaling Inhibitor in an Experimental Model of Autologous Blood-induced Intracerebral Hemorrhage. Curr Med Sci 2022:10.1007/s11596-022-2522-7. [PMID: 35099677 DOI: 10.1007/s11596-022-2522-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/29/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Intracerebral hemorrhage (ICH) refers to predominant, sporadic, and non-traumatic bleeding in the brain parenchyma. The PI3K/AKT/mTOR signaling pathway is an important signal transduction pathway regulated by enzyme-linked receptors and has many biological functions in mammals. It plays a key role in neuronal metabolism, gene expression regulation, and tissue homeostasis in the healthy and diseased brain. METHODS In the present study, the role of the PI3K/AKT/mTOR pathway inhibitor chrysophanol (CPH) (10 mg/kg and 20 mg/kg, orally) in the improvement of ICH-associated neurological defects in rats was investigated. Autologous blood (20 µL/5 min/unilateral/intracerebroventricular) mimics ICH-like defects involving cellular and molecular dysfunction and neurotransmitter imbalance. The current study also included various behavioral assessments to examine cognition, memory, and motor and neuromuscular coordination. The protein expression levels of PI3K, AKT, and mTOR as well as myelin basic protein and apoptotic markers, such as Bax, Bcl-2, and caspase-3, were examined using ELISA kits. Furthermore, the levels of various neuroinflammatory cytokines and oxidative stress markers were assessed. Additionally, the neurological severity score, brain water content, gross brain pathology, and hematoma size were used to indicate neurological function and brain edema. RESULTS CPH was found to be neuroprotective by restoring neurobehavioral alterations and significantly reducing the elevated PI3K, AKT, and mTOR protein levels, and modulating the apoptotic markers such as Bax, Bcl-2, and caspase-3 in rat brain homogenate. CPH substantially reduced the inflammatory cytokines like interleukin (IL)-1β, IL-6, and tumor necrosis factor-α. CPH administration restored the neurotransmitters GABA, glutamate, acetylcholine, dopamine, and various oxidative stress markers. CONCLUSION Our results show that CPH may be a promising therapeutic approach for overcoming neuronal damage caused by the overexpression of the PI3K/AKT/mTOR signaling pathway in ICH-induced neurological dysfunctions in rats.
Collapse
Affiliation(s)
- Kuldeep Singh Jadaun
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Aarti Sharma
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Ehraz Mehmood Siddiqui
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Sumit Kumar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Naif Alsuhaymi
- Department of Emergency Medical Services, Faculty of Health Sciences - AlQunfudah, Umm Al-Qura University, Mekkah, Saudi Arabia
| |
Collapse
|
25
|
Charabati M, Grasmuck C, Ghannam S, Bourbonnière L, Fournier AP, Lécuyer MA, Tastet O, Kebir H, Rébillard RM, Hoornaert C, Gowing E, Larouche S, Fortin O, Pittet C, Filali-Mouhim A, Lahav B, Moumdjian R, Bouthillier A, Girard M, Duquette P, Cayrol R, Peelen E, Quintana FJ, Antel JP, Flügel A, Larochelle C, Arbour N, Zandee S, Prat A. DICAM promotes T H17 lymphocyte trafficking across the blood-brain barrier during autoimmune neuroinflammation. Sci Transl Med 2022; 14:eabj0473. [PMID: 34985970 DOI: 10.1126/scitranslmed.abj0473] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Marc Charabati
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Camille Grasmuck
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Soufiane Ghannam
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Lyne Bourbonnière
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Antoine P Fournier
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Marc-André Lécuyer
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada.,Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen D-37073, Germany
| | - Olivier Tastet
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Hania Kebir
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Rose-Marie Rébillard
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Chloé Hoornaert
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Elizabeth Gowing
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Sandra Larouche
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Olivier Fortin
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Camille Pittet
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Ali Filali-Mouhim
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Boaz Lahav
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec H2L 4M1, Canada
| | - Robert Moumdjian
- Division of Neurosurgery, Université de Montréal and CHUM, Montreal, Quebec H2L 4M1, Canada
| | - Alain Bouthillier
- Division of Neurosurgery, Université de Montréal and CHUM, Montreal, Quebec H2L 4M1, Canada
| | - Marc Girard
- Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec H2L 4M1, Canada
| | - Pierre Duquette
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec H2L 4M1, Canada
| | - Romain Cayrol
- Department of Pathology, Université de Montréal and CHUM, Montreal, Quebec H2L 4M1, Canada
| | - Evelyn Peelen
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Francisco J Quintana
- Ann Romney Carter for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen D-37073, Germany
| | - Catherine Larochelle
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada.,Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec H2L 4M1, Canada
| | - Nathalie Arbour
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Stephanie Zandee
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Alexandre Prat
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada.,Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec H2L 4M1, Canada
| |
Collapse
|
26
|
Raja K, Prabahar A, Arputhanatham SS. A Simple Computational Approach to Identify Potential Drugs for Multiple Sclerosis and Cognitive Disorders from Expert Curated Resources. Methods Mol Biol 2022; 2496:111-121. [PMID: 35713861 DOI: 10.1007/978-1-0716-2305-3_6] [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] [Indexed: 06/15/2023]
Abstract
Multiple sclerosis, a disease of central nervous system leads to potential disability. In the USA, one million cases are diagnosed with multiple sclerosis in 2019. Multiple sclerosis is identified as one of the diseases causing global burden. Cognitive disorder is highly prevalent among 43-70% of multiple sclerosis patients. However, treating cognitive disorder in multiple sclerosis patients is mostly ignored and this leads to several complications. We utilized various expert curated resources to identify potential drugs for multiple sclerosis and cognitive disorder, with specific focus on identifying drugs that are capable of treating both the conditions. We used simple text mining techniques to compile two databases, disease-drug association database and gene-drug interaction database from various existing standard resources. Our study suggests four drugs, Baclofen, Levodopa, Minocycline, and Vitamin B12, for treating both multiple sclerosis and cognitive disorder. In addition, our approach suggests six drugs for multiple sclerosis and 10 drugs for cognitive disorder. We obtained pharmacologist opinion on the drugs suggested for each condition and provided literature evidence for our claim. Here, we present our computational approach as a protocol such that it can be applied to other comorbid diseases that did not gain much attention so far.
Collapse
Affiliation(s)
- Kalpana Raja
- Regenerative Biology, The Morgridge Institute for Research, Madison, WI, USA.
| | - Archana Prabahar
- R&D Division, Eriks-Precision Components India Pvt Ltd, Mohali, Punjab, India
| | | |
Collapse
|
27
|
Identification of miRNAs and Their Potential effects on Multiple Sclerosis Related Pathways Using In Silico Analysis. Mult Scler Relat Disord 2022; 59:103642. [DOI: 10.1016/j.msard.2022.103642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/09/2022] [Accepted: 01/29/2022] [Indexed: 11/22/2022]
|
28
|
Wu W, Chen L, Chen C, Yu L, Zheng J. miRNA-425-5p enhances diffuse large B cell lymphoma growth by targeting PTEN. Transl Cancer Res 2021; 10:4905-4913. [PMID: 35116342 PMCID: PMC8799000 DOI: 10.21037/tcr-21-2394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/24/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND At present, cancer is one of the greatest threats to mankind, and is associated with the highest rates of morbidity and comorbidity. Recently, the advancements in molecular biology have led to an in-depth understanding of the underlying pathophysiology, which may further impact the lead time in the context of early discovery and effective therapy of cancer. Therefore, the present study proposes a better understanding of the role of micro(miR)-425-5p in diffuse large B-cell lymphoma (DLBC). METHODS qRT-PCR was carried out to detect the relevant proteins, miRNA and mRNA RNA gene expression in DLBC cells. The effect of miR-425-5p on DLBC growth was examined by CCK-8 and colony formation assays. The binding relationship between genes was verified by dual-luciferase reporter gene assay. RESULTS We demonstrated how the over-expression of miR-425-5p can lead to increased progression of DLBC by increasing the cellular proliferation rate and colony-forming ability. Additionally, we also found that the expression of miR-425-5p could be significantly inhibited on the basis of phosphatase and tensin homolog (PTEN) signaling pathways. CONCLUSIONS The present study concludes that miR-425-5p is responsible for the oncogenic progression and relapse of DLBC tumorigenesis via PTEN/PI3K signaling, which can thus be effectively used to achieve better therapeutic outcomes.
Collapse
Affiliation(s)
- Weihao Wu
- Department of Hematology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Longtian Chen
- Department of Hematology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Congjie Chen
- Department of Hematology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Lian Yu
- Department of Hematology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Junqiong Zheng
- Department of Oncology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| |
Collapse
|
29
|
Rispoli MG, Valentinuzzi S, De Luca G, Del Boccio P, Federici L, Di Ioia M, Digiovanni A, Grasso EA, Pozzilli V, Villani A, Chiarelli AM, Onofrj M, Wise RG, Pieragostino D, Tomassini V. Contribution of Metabolomics to Multiple Sclerosis Diagnosis, Prognosis and Treatment. Int J Mol Sci 2021; 22:11112. [PMID: 34681773 PMCID: PMC8541167 DOI: 10.3390/ijms222011112] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Metabolomics-based technologies map in vivo biochemical changes that may be used as early indicators of pathological abnormalities prior to the development of clinical symptoms in neurological conditions. Metabolomics may also reveal biochemical pathways implicated in tissue dysfunction and damage and thus assist in the development of novel targeted therapeutics for neuroinflammation and neurodegeneration. Metabolomics holds promise as a non-invasive, high-throughput and cost-effective tool for early diagnosis, follow-up and monitoring of treatment response in multiple sclerosis (MS), in combination with clinical and imaging measures. In this review, we offer evidence in support of the potential of metabolomics as a biomarker and drug discovery tool in MS. We also use pathway analysis of metabolites that are described as potential biomarkers in the literature of MS biofluids to identify the most promising molecules and upstream regulators, and show novel, still unexplored metabolic pathways, whose investigation may open novel avenues of research.
Collapse
Affiliation(s)
- Marianna Gabriella Rispoli
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Silvia Valentinuzzi
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giovanna De Luca
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Piero Del Boccio
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Luca Federici
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Maria Di Ioia
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Anna Digiovanni
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Eleonora Agata Grasso
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Valeria Pozzilli
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Alessandro Villani
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
| | - Antonio Maria Chiarelli
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
| | - Marco Onofrj
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Richard G. Wise
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
| | - Damiana Pieragostino
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Paediatrics, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Valentina Tomassini
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| |
Collapse
|
30
|
Ravi AK, Muthukrishnan SK. Combination of Probiotics and Natural Compounds to Treat Multiple Sclerosis via Warburg Effect. Adv Pharm Bull 2021; 12:515-523. [PMID: 35935051 PMCID: PMC9348531 DOI: 10.34172/apb.2022.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/13/2021] [Accepted: 09/27/2021] [Indexed: 11/26/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS). It is an auto-immune disorder. Its usual symptoms are unique to each person. In MS lesions vast fractions of pyruvate molecules are instantly transformed into lactate. This reprogramming mechanism of glycolysis is known as the Warburg effect. MS has no efficient treatment yet. Hence, there is a requirement for profitable immunomodulatory agents in MS. Probiotics perform as an immunomodulator because they regulate the host’s immune responses. Its efficacy gets enhanced for an extended period when it combines with prebiotics. In this review, we focus on the metabolic alterations behind the MS lesions via the Warburg effect, and also suggesting, the combined efficacy of prebiotics and probiotics for the effective treatment of MS without side effects. The Warburg effect mechanism intensifies the infiltration of activated T-cells and B-cells into the CNS. It provokes the inflammation process on the myelin sheath. The infiltration of immune cells can be inhibited by the combination therapy of probiotics and prebiotics. By this review, we can recommend that the idea of this combinational therapy can do miracles in the treatment of MS in the future.
Collapse
|
31
|
Badam TVS, de Weerd HA, Martínez-Enguita D, Olsson T, Alfredsson L, Kockum I, Jagodic M, Lubovac-Pilav Z, Gustafsson M. A validated generally applicable approach using the systematic assessment of disease modules by GWAS reveals a multi-omic module strongly associated with risk factors in multiple sclerosis. BMC Genomics 2021; 22:631. [PMID: 34461822 PMCID: PMC8404328 DOI: 10.1186/s12864-021-07935-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 08/17/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There exist few, if any, practical guidelines for predictive and falsifiable multi-omic data integration that systematically integrate existing knowledge. Disease modules are popular concepts for interpreting genome-wide studies in medicine but have so far not been systematically evaluated and may lead to corroborating multi-omic modules. RESULT We assessed eight module identification methods in 57 previously published expression and methylation studies of 19 diseases using GWAS enrichment analysis. Next, we applied the same strategy for multi-omic integration of 20 datasets of multiple sclerosis (MS), and further validated the resulting module using both GWAS and risk-factor-associated genes from several independent cohorts. Our benchmark of modules showed that in immune-associated diseases modules inferred from clique-based methods were the most enriched for GWAS genes. The multi-omic case study using MS data revealed the robust identification of a module of 220 genes. Strikingly, most genes of the module were differentially methylated upon the action of one or several environmental risk factors in MS (n = 217, P = 10- 47) and were also independently validated for association with five different risk factors of MS, which further stressed the high genetic and epigenetic relevance of the module for MS. CONCLUSIONS We believe our analysis provides a workflow for selecting modules and our benchmark study may help further improvement of disease module methods. Moreover, we also stress that our methodology is generally applicable for combining and assessing the performance of multi-omic approaches for complex diseases.
Collapse
Affiliation(s)
- Tejaswi V S Badam
- School of Bioscience, Systems Biology Research Center, University of Skövde, Skövde, Sweden
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping university, Linköping, Sweden
| | - Hendrik A de Weerd
- School of Bioscience, Systems Biology Research Center, University of Skövde, Skövde, Sweden
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping university, Linköping, Sweden
| | - David Martínez-Enguita
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping university, Linköping, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Lars Alfredsson
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Zelmina Lubovac-Pilav
- School of Bioscience, Systems Biology Research Center, University of Skövde, Skövde, Sweden
| | - Mika Gustafsson
- Bioinformatics, Department of Physics, Chemistry and Biology, Linköping university, Linköping, Sweden.
| |
Collapse
|
32
|
Tian Y, Ren F, Xu L, Zhang X. Distinct effects of different doses of kaempferol on D‑GalN/LPS‑induced ALF depend on the autophagy pathway. Mol Med Rep 2021; 24:682. [PMID: 34318900 PMCID: PMC8335584 DOI: 10.3892/mmr.2021.12321] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 06/24/2021] [Indexed: 01/16/2023] Open
Abstract
Kaempferol, a flavonoid compound, has various biological functions, such as anti-inflammatory and antitumor activities. Acute liver failure (ALF) is a lethal clinical syndrome that occurs due to severe damage of the liver function. In the present study, the mechanisms underlying the therapeutic effects of kaempferol in ALF were evaluated. An ALF mouse model was established using D-galactosamine (D-GalN; 700 mg/kg)/lipopolysaccharide (LPS; 10 µg/kg). A total of 2 h before the administration of D-GalN/LPS, mice were pretreated with different doses of kaempferol (2.5, 5, 10, 20 and 40 mg/kg), and 6 h after injection of D-GalN/LPS, mice were euthanized. The survival rate, liver function and levels of inflammatory cytokines were assessed. The results demonstrated that kaempferol pretreatment protected hepatocytes from ALF induced by D-GalN/LPS via regulation of the autophagy pathway, both in vivo and in vitro. Pretreatment with a high dose of kaempferol significantly decreased the survival rates and increased severe liver damage; however, pretreatment with a low dose of kaempferol had the opposite effect. Furthermore, pretreatment with a high dose of kaempferol enhanced the levels of proinflammatory cytokines [TNF-α, IL-6, IL-12p40, IL-1β, C-X-C motif chemokine ligand (CXCL)-2, CXCL-10] and markers of the MAPK signaling pathway [phosphorylated (p)-JNK, p-ERK, p-p38], whereas pretreatment with a low dose of kaempferol had the opposite effect. Pretreatment with a high dose of kaempferol decreased autophagy, whereas pretreatment with a low dose of kaempferol increased autophagy in vivo and in vitro. It was also shown that pretreatment with 3-methyadenine or autophagy related 7 small interfering RNA, to inhibit autophagy, partially abrogated the hepatoprotective effects of pretreatment with 5 mg/kg kaempferol in the ALF mouse model. These results demonstrate that the effects of different doses of kaempferol on D-GalN/LPS-induced ALF varies based on the dose, and that kaempferol exerted its effects via regulation of the autophagy pathway.
Collapse
Affiliation(s)
- Yuan Tian
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Feng Ren
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Ling Xu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Xiangying Zhang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
33
|
Petralia MC, Ciurleo R, Bramanti A, Bramanti P, Saraceno A, Mangano K, Quattropani MC, Nicoletti F, Fagone P. Transcriptomic Data Analysis Reveals a Down-Expression of Galectin-8 in Schizophrenia Hippocampus. Brain Sci 2021; 11:brainsci11080973. [PMID: 34439592 PMCID: PMC8392448 DOI: 10.3390/brainsci11080973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 12/31/2022] Open
Abstract
Schizophrenia (SCZ) is a severe psychiatric disorder with several clinical manifestations that include cognitive dysfunction, decline in motivation, and psychosis. Current standards of care treatment with antipsychotic agents are often ineffective in controlling the disease, as only one-third of SCZ patients respond to medications. The mechanisms underlying the pathogenesis of SCZ remain elusive. It is believed that inflammatory processes may play a role as contributing factors to the etiology of SCZ. Galectins are a family of β-galactoside-binding lectins that contribute to the regulation of immune and inflammatory responses, and previous reports have shown their role in the maintenance of central nervous system (CNS) homeostasis and neuroinflammation. In the current study, we evaluated the expression levels of the galectin gene family in post-mortem samples of the hippocampus, associative striatum, and dorsolateral prefrontal cortex from SCZ patients. We found a significant downregulation of LGALS8 (Galectin-8) in the hippocampus of SCZ patients as compared to otherwise healthy donors. Interestingly, the reduction of LGALS8 was disease-specific, as no modulation was observed in the hippocampus from bipolar nor major depressive disorder (MDD) patients. Prediction analysis identified TBL1XR1, BRF2, and TAF7 as potential transcription factors controlling LGALS8 expression. In addition, MIR3681HG and MIR4296 were negatively correlated with LGALS8 expression, suggesting a role for epigenetics in the regulation of LGALS8 levels. On the other hand, no differences in the methylation levels of LGALS8 were observed between SCZ and matched control hippocampus. Finally, ontology analysis of the genes negatively correlated with LGALS8 expression identified an enrichment of the NGF-stimulated transcription pathway and of the oligodendrocyte differentiation pathway. Our study identified LGALS8 as a disease-specific gene, characterizing SCZ patients, that may in the future be exploited as a potential therapeutic target.
Collapse
Affiliation(s)
- Maria Cristina Petralia
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.C.P.); (M.C.Q.)
| | - Rosella Ciurleo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (R.C.); (P.B.)
| | - Alessia Bramanti
- Department of Medicine, University of Salerno, 84084 Salerno, Italy;
| | - Placido Bramanti
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (R.C.); (P.B.)
| | - Andrea Saraceno
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (K.M.); (F.N.)
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (K.M.); (F.N.)
| | - Maria Catena Quattropani
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.C.P.); (M.C.Q.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (K.M.); (F.N.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (K.M.); (F.N.)
- Correspondence:
| |
Collapse
|
34
|
Xu R, Zhang Y, Li A, Ma Y, Cai W, Song L, Xie Y, Zhou S, Cao W, Tang X. LY‑294002 enhances the chemosensitivity of liver cancer to oxaliplatin by blocking the PI3K/AKT/HIF‑1α pathway. Mol Med Rep 2021; 24:508. [PMID: 33982772 PMCID: PMC8134878 DOI: 10.3892/mmr.2021.12147] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Liver cancer remains one of the leading causes of cancer deaths worldwide. The therapeutic effect of oxaliplatin on liver cancer is often limited by acquired resistance of the cancer cells. Abnormal activation of the PI3K/AKT pathway plays an important role in the acquired resistance of oxaliplatin. The present study investigated the effects of the PI3K inhibitor LY-294002 and AKT inhibitor MK2206 on the chemosensitivity of oxaliplatin-resistant liver cancer cells and the molecular mechanism involved. An oxaliplatin-resistant liver cancer cell line HepG2R was developed. MTT assay, clone formation experiments, flow cytometry and Annexin V-FITC/PI staining were used to determine the proliferation, cycle and apoptosis of HepG2R cells when oxaliplatin was combined with LY-294002 or MK2206 treatment. The effects of LY-294002 and MK-2206 on the abnormal activation of PI3K/AKT pathway and hypoxia inducible factor (HIF)-1α protein level in HepG2R cells were detected using western blotting. The results indicated that the PI3K/AKT pathway is stably activated in HepG2R cells. Compared with the AKT inhibitor MK2206, the PI3K inhibitor LY-294002 more effectively downregulated the phosphorylation levels of p85, p110α, p110β, p110γ and AKT in the PI3K/AKT pathway in HepG2R cells, and more effectively inhibited the proliferation of the cells. LY-294002 enhanced the chemotherapy sensitivity of HepG2R cells to oxaliplatin by inducing G0/G1 phase arrest and increasing the proportion of apoptotic cells. In addition, LY-294002 reduced the level of HIF-1α, which is highly expressed in HepG2R cells. It was concluded that LY-294002 enhanced the chemosensitivity of liver cancer cells to oxaliplatin by inhibiting the PI3K/AKT signaling pathway, which may be related to the inhibition of HIF-1α expression. These findings may have clinical significance for the treatment of oxaliplatin-resistant liver cancer.
Collapse
Affiliation(s)
- Ruyue Xu
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Yinci Zhang
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Amin Li
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Yongfang Ma
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Wenpeng Cai
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Li Song
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Yinghai Xie
- Institute of Environmentally Friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, Anhui 241000, P.R. China
| | - Shuping Zhou
- Institute of Environmentally Friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, Anhui 241000, P.R. China
| | - Weiya Cao
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Xiaolong Tang
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| |
Collapse
|
35
|
Mosarrezaii Aghdam A, Rezaei S, Zarza Nalivan F, Babaie F, Amiri Nikpour MR, Torkamandi S. Downregulation of miR-125a-5p and miR-218-5p in Peripheral Blood Mononuclear Cells of Patients with Relapsing-Remitting Multiple Sclerosis. Immunol Invest 2021; 51:1149-1161. [PMID: 33866949 DOI: 10.1080/08820139.2021.1909616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Multiple sclerosis (MS) is a chronic neuroinflammatory disease of the brain and spinal cord. Evidences have demonstrated that microRNAs (miRNAs) are involved in the pathological process of MS that may confer a valuable diagnostic biomarker for disease diagnosis, prognosis, and treatment. Hence, we assessed the expression pattern of miR-125a-5p and miR-218-5p in the peripheral blood mononuclear cells (PBMCs) of subjects with relapsing-remitting multiple sclerosis (RRMS). We recruited 50 RRMS patients and 50 age- and sex-matched healthy control subjects. PBMCs were isolated from the peripheral blood samples, RNA content was extracted, cDNA was synthesized, and finally expression level of miRNAs was determined using quantitative real-time PCR. Our data indicate significant downregulation of both miR-125a-5p and miR-218-5p in RRMS patients compared to healthy controls (P< .0001). The levels of both miRNAs were significantly downregulated in an age-dependent manner compared with consistent healthy control groups (30-40 years old P< .0001). Expression level of miR-218-5p was significantly changed in only female patients (Female group P< .0001; Male group P= .12). Receiver operating characteristic (ROC) curve data indicated that the expression levels of both miRNAs were able to discriminate RRMS patients from healthy subjects (P< .05). Moreover, bioinformatic enrichment analysis revealed that the target genes of these miRNAs had cardinal roles in the regulation of key biological pathways involved in the clinical course and pathogenesis of MS. Collectively, our results suggested that miR-125a-5p and miR-218-5p play a role in RRMS pathogenesis and have an age- and sex-dependent expression pattern in these patients.
Collapse
Affiliation(s)
- Arash Mosarrezaii Aghdam
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.,Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran
| | - Somaye Rezaei
- Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran
| | - Fariba Zarza Nalivan
- Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran
| | - Farhad Babaie
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.,Department of Medical Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Reza Amiri Nikpour
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Shahram Torkamandi
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.,Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.,Department of Medical Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
36
|
Damstra-Oddy JL, Warren EC, Perry CJ, Desfougères Y, Fitzpatrick JMK, Schaf J, Costelloe L, Hind W, Downer EJ, Saiardi A, Williams RSB. Phytocannabinoid-dependent mTORC1 regulation is dependent upon inositol polyphosphate multikinase activity. Br J Pharmacol 2021; 178:1149-1163. [PMID: 33347604 PMCID: PMC9328663 DOI: 10.1111/bph.15351] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Cannabidiol (CBD) has been shown to differentially regulate the mechanistic target of rapamycin complex 1 (mTORC1) in preclinical models of disease, where it reduces activity in models of epilepsies and cancer and increases it in models of multiple sclerosis (MS) and psychosis. Here, we investigate the effects of phytocannabinoids on mTORC1 and define a molecular mechanism. EXPERIMENTAL APPROACH A novel mechanism for phytocannabinoids was identified using the tractable model system, Dictyostelium discoideum. Using mouse embryonic fibroblasts, we further validate this new mechanism of action. We demonstrate clinical relevance using cells derived from healthy individuals and from people with MS (pwMS). KEY RESULTS Both CBD and the more abundant cannabigerol (CBG) enhance mTORC1 activity in D. discoideum. We identify a mechanism for this effect involving inositol polyphosphate multikinase (IPMK), where elevated IPMK expression reverses the response to phytocannabinoids, decreasing mTORC1 activity upon treatment, providing new insight on phytocannabinoids' actions. We further validated this mechanism using mouse embryonic fibroblasts. Clinical relevance of this effect was shown in primary human peripheral blood mononuclear cells, where CBD and CBG treatment increased mTORC1 activity in cells derived from healthy individuals and decreased mTORC1 activity in cells derived from pwMS. CONCLUSION AND IMPLICATIONS Our findings suggest that both CBD and the abundant CBG differentially regulate mTORC1 signalling through a mechanism dependent on the activity of the upstream IPMK signalling pathway, with potential relevance to the treatment of mTOR-related disorders, including MS.
Collapse
Affiliation(s)
- Joseph L Damstra-Oddy
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Eleanor C Warren
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Christopher J Perry
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Yann Desfougères
- Laboratory for Molecular Cell Biology, University College London, London, UK
| | - John-Mark K Fitzpatrick
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Judith Schaf
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Lisa Costelloe
- Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | | | - Eric J Downer
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Adolfo Saiardi
- Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Robin S B Williams
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| |
Collapse
|
37
|
Dash R, Ali MC, Jahan I, Munni YA, Mitra S, Hannan MA, Timalsina B, Oktaviani DF, Choi HJ, Moon IS. Emerging potential of cannabidiol in reversing proteinopathies. Ageing Res Rev 2021; 65:101209. [PMID: 33181336 DOI: 10.1016/j.arr.2020.101209] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/22/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022]
Abstract
The aberrant accumulation of disease-specific protein aggregates accompanying cognitive decline is a pathological hallmark of age-associated neurological disorders, also termed as proteinopathies, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and multiple sclerosis. Along with oxidative stress and neuroinflammation, disruption in protein homeostasis (proteostasis), a network that constitutes protein surveillance system, plays a pivotal role in the pathobiology of these dementia disorders. Cannabidiol (CBD), a non-psychotropic phytocannabinoid of Cannabis sativa, is known for its pleiotropic neuropharmacological effects on the central nervous system, including the ability to abate oxidative stress, neuroinflammation, and protein misfolding. Over the past years, compelling evidence has documented disease-modifying role of CBD in various preclinical and clinical models of neurological disorders, suggesting the potential therapeutic implications of CBD in these disorders. Because of its putative role in the proteostasis network in particular, CBD could be a potent modulator for reversing not only age-associated neurodegeneration but also other protein misfolding disorders. However, the current understanding is insufficient to underpin this proposition. In this review, we discuss the potentiality of CBD as a pharmacological modulator of the proteostasis network, highlighting its neuroprotective and aggregates clearing roles in the neurodegenerative disorders. We anticipate that the current effort will advance our knowledge on the implication of CBD in proteostasis network, opening up a new therapeutic window for aging proteinopathies.
Collapse
|
38
|
Caltabiano R, De Pasquale R, Piombino E, Campo G, Nicoletti F, Cavalli E, Mangano K, Fagone P. Macrophage Migration Inhibitory Factor (MIF) and Its Homologue d-Dopachrome Tautomerase (DDT) Inversely Correlate with Inflammation in Discoid Lupus Erythematosus. Molecules 2021; 26:molecules26010184. [PMID: 33401503 PMCID: PMC7795694 DOI: 10.3390/molecules26010184] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/25/2020] [Accepted: 12/29/2020] [Indexed: 01/12/2023] Open
Abstract
Discoid Lupus Erythematosus (DLE) is a chronic cutaneous disease of unknown etiology and of immunoinflammatory origin that is characterized by inflammatory plaques and may lead to disfiguring scarring and skin atrophy. Current treatments are limited, with a large proportion of patients either poorly or not responsive, which makes DLE an unmet medical need. Macrophage migration inhibitory factor (MIF) is the prototype of a pleiotropic family of cytokine that also includes the recently discovered homologue D-dopachrome tautomerase (DDT) or MIF2. MIF and DDT/MIF-2 exert several biological properties, primarily, but not exclusively of a proinflammatory nature. MIF and DDT have been suggested to play a key role in the pathogenesis of several autoimmune diseases, such as multiple sclerosis and type 1 diabetes, as well as in the development and progression of certain forms of cancers. In the present study, we have performed an immunohistochemistry analysis for the evaluation of MIF in DLE lesions and normal skin. We found high levels of MIF in the basal layer of the epidermis as well as in the cutaneous appendage (eccrine glands and sebocytes) of normal skin. In DLE lesions, we observed a significant negative correlation between the expression of MIF and the severity of inflammation. In addition, we performed an analysis of MIF and DDT expression levels in the skin of DLE patients in a publicly available microarray dataset. Interestingly, while these in silico data only evidenced a trend toward reduced levels of MIF, they demonstrated a significant pattern of expression and correlation of DDT with inflammatory infiltrates in DLE skins. Overall, our data support a protective role for endogenous MIF and possibly DDT in the regulation of homeostasis and inflammation in the skin and open up novel avenues for the treatment of DLE.
Collapse
Affiliation(s)
- Rosario Caltabiano
- Department of Medical, Surgical and Advanced Technologies “G.F. Ingrassia”, University of Catania, Via Santa Sofia, 87, 95123 Catania, Italy; (R.C.); (E.P.)
| | - Rocco De Pasquale
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy;
| | - Eliana Piombino
- Department of Medical, Surgical and Advanced Technologies “G.F. Ingrassia”, University of Catania, Via Santa Sofia, 87, 95123 Catania, Italy; (R.C.); (E.P.)
| | - Giorgia Campo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.C.); (E.C.); (K.M.); (P.F.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.C.); (E.C.); (K.M.); (P.F.)
- Correspondence:
| | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.C.); (E.C.); (K.M.); (P.F.)
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.C.); (E.C.); (K.M.); (P.F.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.C.); (E.C.); (K.M.); (P.F.)
| |
Collapse
|
39
|
Akbarian F, Tabatabaiefar MA, Shaygannejad V, Shahpouri MM, Badihian N, Sajjadi R, Dabiri A, Jalilian N, Noori-Daloii MR. Upregulation of MTOR, RPS6KB1, and EIF4EBP1 in the whole blood samples of Iranian patients with multiple sclerosis compared to healthy controls. Metab Brain Dis 2020; 35:1309-1316. [PMID: 32809098 DOI: 10.1007/s11011-020-00590-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 06/23/2020] [Indexed: 12/22/2022]
Abstract
Various genetic and epigenetic mechanisms have been suggested to play roles as the underlying pathophysiology of Multiple Sclerosis (MS). Changes in different parts of the mTOR signaling pathway are among the potential suggested mechanisms based on the specific roles of this pathway in CNS. MTOR, RPS6KB1, and EIFEBP1 genes are among important genes in the mTOR pathway, responsible for the proper function of acting proteins in this signaling pathway. This study aimed to investigate the relative expression levels of these genes in the blood samples of relapsing-remitting MS (RRMS) patients compared to healthy controls. In this case-control study blood samples were collected from 30 newly diagnosed RRMS patients and 30 age and sex-matched healthy controls. mRNA level of MTOR, RPS6KB1, and EIFEBP1 genes were assessed using Real-Time PCR. The expression of MTOR, RPS6KB1, and EIF4EBP1 genes was up regulated in MS patients compared to healthy controls (p < 0.001 for all mentioned genes). Considering gender differences, expression of the mentioned genes was increased among female patients (all P < 0.001). However, no statistically significant changes were observed among male patients. Based on the receiver operating characteristic, MTOR gene had the highest diagnostic value followed by EIF4EBP1 and RPS6KB1 genes in differentiating RRMS patients from controls. In conclusion, we found the simultaneous upregulation of MTOR, RPS6KB1, and EIF4EBP1 genes among RRMS patients. MTOR showed to have the highest diagnostic value compared to other 2 genes in differentiating RRMS patients. Further studies evaluating the importance of these findings from pharmacological and prognostic perspectives are necessary.
Collapse
Affiliation(s)
- Fahimeh Akbarian
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Poursina St., Tehran, 14155-6447, Iran
| | - Mohammad Amin Tabatabaiefar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Noncommunicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vahid Shaygannejad
- Isfahan Neuroscience Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Negin Badihian
- Isfahan Neuroscience Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roshanak Sajjadi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arezou Dabiri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Nazanin Jalilian
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Reza Noori-Daloii
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Poursina St., Tehran, 14155-6447, Iran.
| |
Collapse
|
40
|
Tancheva L, Petralia MC, Miteva S, Dragomanova S, Solak A, Kalfin R, Lazarova M, Yarkov D, Ciurleo R, Cavalli E, Bramanti A, Nicoletti F. Emerging Neurological and Psychobiological Aspects of COVID-19 Infection. Brain Sci 2020; 10:E852. [PMID: 33198412 PMCID: PMC7696269 DOI: 10.3390/brainsci10110852] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
The SARS-CoV-2 virus, first reported in December 2019 in China, is the causative agent of the current COVID-19 pandemic that, at the time of writing (1 November 2020) has infected almost 43 million people and caused the death of more than 1 million people. The spectrum of clinical manifestations observed during COVID-19 infection varies from asymptomatic to critical life-threatening clinical conditions. Emerging evidence shows that COVID-19 affects far more organs than just the respiratory system, including the heart, kidneys, blood vessels, liver, as well as the central nervous system (CNS) and the peripheral nervous system (PNS). It is also becoming clear that the neurological and psychological disturbances that occur during the acute phase of the infection may persist well beyond the recovery. The aim of this review is to propel further this emerging and relevant field of research related to the pathophysiology of neurological manifestation of COVID-19 infection (Neuro-COVID). We will summarize the PNS and CNS symptoms experienced by people with COVID-19 both during infection and in the recovery phase. Diagnostic and pharmacological findings in this field of study are strongly warranted to address the neurological and psychological symptoms of COVID-19.
Collapse
Affiliation(s)
- Lyubka Tancheva
- Department of Behavior Neurobiology, Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (L.T.); (S.M.); (S.D.); (R.K.); (M.L.)
| | - Maria Cristina Petralia
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (M.C.P.); (R.C.); (A.B.)
| | - Simona Miteva
- Department of Behavior Neurobiology, Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (L.T.); (S.M.); (S.D.); (R.K.); (M.L.)
| | - Stela Dragomanova
- Department of Behavior Neurobiology, Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (L.T.); (S.M.); (S.D.); (R.K.); (M.L.)
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University, 9002 Varna, Bulgaria
| | - Ayten Solak
- Institute of Cryobiology and food technologies, Agricultural Academy, 1407 Sofia, Bulgaria;
| | - Reni Kalfin
- Department of Behavior Neurobiology, Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (L.T.); (S.M.); (S.D.); (R.K.); (M.L.)
| | - Maria Lazarova
- Department of Behavior Neurobiology, Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (L.T.); (S.M.); (S.D.); (R.K.); (M.L.)
| | - Dobri Yarkov
- Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Rosella Ciurleo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (M.C.P.); (R.C.); (A.B.)
| | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy;
| | - Alessia Bramanti
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (M.C.P.); (R.C.); (A.B.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy;
| |
Collapse
|
41
|
Yamamoto S, Sakemoto C, Iwasa K, Maruyama K, Shimizu K, Yoshikawa K. Ursolic acid treatment suppresses cuprizone-induced demyelination and motor dysfunction via upregulation of IGF-1. J Pharmacol Sci 2020; 144:119-122. [PMID: 32921392 DOI: 10.1016/j.jphs.2020.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/04/2020] [Accepted: 08/14/2020] [Indexed: 11/30/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system, characterized by apoptotic death of mature oligodendrocytes, neuroinflammation, and motor dysfunction. A pentacyclic triterpenoid compound, ursolic acid (UA), has various pharmacological activities, such as anti-inflammatory, anti-oxidative, and anti-apoptotic effects. In the present study, we investigated the effects of UA on cuprizone-induced demyelination, which is a model of MS. Oral administration of UA effectively suppressed cuprizone-induced demyelination and motor dysfunction via the enhancement of IGF-1 levels in the demyelinating lesions. Our results suggest that UA might be therapeutically useful for demyelination in MS.
Collapse
Affiliation(s)
- Shinji Yamamoto
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; School of Medical Technology, Faculty of Health and Medical Care, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama 350-1241, Japan
| | - Chiaki Sakemoto
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; School of Medical Technology, Faculty of Health and Medical Care, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama 350-1241, Japan
| | - Kensuke Iwasa
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan
| | - Kei Maruyama
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan
| | - Kuniyoshi Shimizu
- Laboratory of Systematic Forest and Forest Products Sciences, Division of Sustainable Bioresources Science, Department of Agro-Environmental Sciences, Faculty of Agriculture, Graduate School of Kyushu University, West Zone, Building 5, 744, Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Keisuke Yoshikawa
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan.
| |
Collapse
|
42
|
Lombardo SD, Bramanti A, Ciurleo R, Basile MS, Pennisi M, Bella R, Mangano K, Bramanti P, Nicoletti F, Fagone P. Profiling of inhibitory immune checkpoints in glioblastoma: Potential pathogenetic players. Oncol Lett 2020; 20:332. [PMID: 33123243 PMCID: PMC7583708 DOI: 10.3892/ol.2020.12195] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/06/2020] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) represents the most frequent glial tumor, with almost 3 new cases per 100,000 people per year. Despite treatment, the prognosis for GBM patients remains extremely poor, with a median survival of 14.6 months, and a 5-year survival less than 5%. It is generally believed that GBM creates a highly immunosuppressive microenvironment, sustained by the expression of immune-regulatory factors, including inhibitory immune checkpoints, on both infiltrating cells and tumor cells. However, the trials assessing the efficacy of current immune checkpoint inhibitors in GBM are still disappointing. In the present study, the expression levels of several inhibitory immune checkpoints in GBM (CD276, VTCN1, CD47, PVR, TNFRSF14, CD200, LGALS9, NECTIN2 and CD48) were characterized in order to evaluate their potential as prognostic and eventually, therapeutic targets. Among the investigated immune checkpoints, TNFRSF14 and NECTIN2 were identified as the most promising targets in GBM. In particular, a higher TNFRSF14 expression was associated with worse overall survival and disease-free survival, and with a lower Th1 response.
Collapse
Affiliation(s)
- Salvo Danilo Lombardo
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, A-1090 Vienna, Austria
| | | | - Rosella Ciurleo
- IRCCS Centro Neurolesi Bonino Pulejo, I-98124 Messina, Italy
| | | | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Rita Bella
- Department of Medical Sciences, Surgery and Advanced Technologies, University of Catania, I-95123 Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania, Italy
| |
Collapse
|
43
|
Sanfilippo C, Musumeci G, Kazakova M, Mazzone V, Castrogiovanni P, Imbesi R, Di Rosa M. GNG13 Is a Potential Marker of the State of Health of Alzheimer's Disease Patients' Cerebellum. J Mol Neurosci 2020; 71:1046-1060. [PMID: 33057964 DOI: 10.1007/s12031-020-01726-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
Brain regions such as the cerebellum (CB) have been neglected for a long time in the study of Alzheimer's disease (AD) pathogenesis. In reference to a new emerging hypothesis according to which there is an altered cerebellar synaptic processing in AD, we verified the possible role played by new biomarkers in the CB of AD patients compared with not-demented healthy control subjects (NDHS). Using a bioinformatics approach, we have collected several microarray datasets and obtained 626 cerebella sample biopsies belonging to subjects who did not die from causes related to neurological diseases and 199 cerebella belonging to AD. The analysis of logical relations between the transcriptome dataset highlighted guanine nucleotide-binding protein (G protein) gamma 13 (GNG13) as a potential new biomarker for Purkinje cells (PCs). We have correlated GNG13 expression levels with already widely existing bibliography of PC marker genes, such as Purkinje cell protein 2 (PCP2), Purkinje cell protein 4 (PCP4), and cerebellin 3 (CBLN3). We showed that expression levels of GNG13 and PCP2, PCP4, and CBLN3 were significantly correlated with each other in NDHS and in AD and significantly reduced in AD patients compared with NDHS subjects. In addition, we highlighted a negative correlation between the expression levels of PC biomarkers and age. From the outcome of our investigation, it is possible to conclude that the identification of GNG13 as a potentially biomarker in PCs represents also a state of health of CB, in association with the expression of PCP2, PCP4, and CBLN3.
Collapse
Affiliation(s)
- Cristina Sanfilippo
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124, Messina, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Maria Kazakova
- Department of Medical Biology, Medical Faculty, Medical University, Plovdiv, Bulgaria
| | - Venera Mazzone
- Department G.F. Ingrassia, Anatomy, School of Medicine, University of Catania, Catania, Italy
| | - Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Rosa Imbesi
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
44
|
Advani D, Gupta R, Tripathi R, Sharma S, Ambasta RK, Kumar P. Protective role of anticancer drugs in neurodegenerative disorders: A drug repurposing approach. Neurochem Int 2020; 140:104841. [PMID: 32853752 DOI: 10.1016/j.neuint.2020.104841] [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: 07/01/2020] [Revised: 07/24/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
The disease heterogeneity and little therapeutic progress in neurodegenerative diseases justify the need for novel and effective drug discovery approaches. Drug repurposing is an emerging approach that reinvigorates the classical drug discovery method by divulging new therapeutic uses of existing drugs. The common biological background and inverse tuning between cancer and neurodegeneration give weight to the conceptualization of repurposing of anticancer drugs as novel therapeutics. Many studies are available in the literature, which highlights the success story of anticancer drugs as repurposed therapeutics. Among them, kinase inhibitors, developed for various oncology indications evinced notable neuroprotective effects in neurodegenerative diseases. In this review, we shed light on the salient role of multiple protein kinases in neurodegenerative disorders. We also proposed a feasible explanation of the action of kinase inhibitors in neurodegenerative disorders with more attention towards neurodegenerative disorders. The problem of neurotoxicity associated with some anticancer drugs is also highlighted. Our review encourages further research to better encode the hidden potential of anticancer drugs with the aim of developing prospective repurposed drugs with no toxicity for neurodegenerative disorders.
Collapse
Affiliation(s)
- Dia Advani
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rohan Gupta
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rahul Tripathi
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Sudhanshu Sharma
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|
45
|
Jiang C, Yang H, Chen X, Qiu S, Wu C, Zhang B, Jin L. Macleaya cordata extracts exert antiviral effects in newborn mice with rotavirus-induced diarrhea via inhibiting the JAK2/STAT3 signaling pathway. Exp Ther Med 2020; 20:1137-1144. [PMID: 32742353 PMCID: PMC7388234 DOI: 10.3892/etm.2020.8766] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 01/16/2020] [Indexed: 12/22/2022] Open
Abstract
Accumulating evidence demonstrates that Macleaya cordata extract exerts antiviral and anti-inflammatory effects in various diseases. The present study aimed to investigate the potential effects of M. cordata on rotavirus SA11-induced diarrhea in mice. Diarrhea severity, levels of inflammatory cytokines, histological changes in the small intestine and the underlying mechanisms were evaluated in rotavirus-stimulated mice treated with 1, 2 and 4 mg/kg/day M. cordata or 4 mg/kg/day ribavirin (positive control). M. cordata treatment effectively ameliorated rotavirus-induced diarrhea in a dose-dependent manner by decreasing viral RNA levels. In addition, M. cordata reduced the release of pro-inflammatory cytokines including migration inhibitory factor, interleukin (IL)-8, IL-β, interferon-γ and tumor necrosis factor-α, and elevated the secretion of the anti-inflammatory cytokine IL-10 following rotavirus infection. M. cordata inhibited intestinal epithelial cell apoptosis and improved intestinal inflammation after rotavirus infection. The study also revealed that M. cordata exerted antiviral and anti-inflammatory effects on rotavirus-induced diarrhea by suppressing the Janus kinase 2 (JAK2)/STAT3 pathway, as reflected by decreased protein expression of phosphorylated (p)-JAK2 and p-STAT3. Overall, M. cordata effectively inhibited the inflammation caused by rotavirus, which was closely associated with the suppression of JAK2/STAT3 phosphorylation. These data suggested that M. cordata may be applied as a treatment for rotavirus-induced diarrhea.
Collapse
Affiliation(s)
- Chunmao Jiang
- School of Veterinary Medicine, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, P.R. China
| | - Haifeng Yang
- School of Animal Pharmaceutical Sciences, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, P.R. China
| | - Xiaolan Chen
- School of Animal Pharmaceutical Sciences, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, P.R. China
| | - Shulei Qiu
- School of Veterinary Medicine, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, P.R. China
| | - Caihong Wu
- School of Veterinary Medicine, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, P.R. China
| | - Bin Zhang
- School of Pet Science and Technology, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, P.R. China
| | - Liqin Jin
- School of Animal Pharmaceutical Sciences, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, P.R. China
| |
Collapse
|
46
|
Lazarević M, Battaglia G, Jevtić B, Đedović N, Bruno V, Cavalli E, Miljković Đ, Nicoletti F, Momčilović M, Fagone P. Upregulation of Tolerogenic Pathways by the Hydrogen Sulfide Donor GYY4137 and Impaired Expression of H 2S-Producing Enzymes in Multiple Sclerosis. Antioxidants (Basel) 2020; 9:E608. [PMID: 32664399 PMCID: PMC7402185 DOI: 10.3390/antiox9070608] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/22/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
The aim of this study was to examine the in vitro effects of the slow-releasing H2S donor GYY4137 on the immune cells involved in the pathogenesis of the central nervous system (CNS) autoimmune disease, multiple sclerosis (MS). GYY4137 specifically potentiated TGF-β expression and production in dendritic cells and significantly reduced IFN-γ and IL-17 production in the lymph node and spinal cord T cells obtained from mice immunized with CNS antigens. Both the proportion of FoxP3+ regulatory CD4+ T cells in the lymph node cells, and the percentage of IL-17+ CD4+ T cells in the spinal cord cells were reduced upon culturing with GYY4137. Interestingly, the peripheral blood mononuclear cells obtained from the MS patients had a lower expression of the H2S-producing enzyme, 3-mercaptopyruvate-sulfurtransferase (MPST), in comparison to those obtained from healthy donors. A significant inverse correlation between the expression of MPST and several pro-inflammatory factors was also observed. Further studies on the relevance of the observed results for the pathogenesis and therapy of MS are warranted.
Collapse
Affiliation(s)
- Milica Lazarević
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Giuseppe Battaglia
- Department of Physiology and Pharmacology, Sapienza University, Piazzale A. Moro, 5, 00185 Rome, Italy
- IRCCS Neuromed, Località Camerelle, 86077 Pozzilli, Italy
| | - Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Neda Đedović
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Valeria Bruno
- Department of Physiology and Pharmacology, Sapienza University, Piazzale A. Moro, 5, 00185 Rome, Italy
- IRCCS Neuromed, Località Camerelle, 86077 Pozzilli, Italy
| | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Miljana Momčilović
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| |
Collapse
|
47
|
Senousy MA, Shaker OG, Sayed NH, Fathy N, Kortam MA. LncRNA GAS5 and miR-137 Polymorphisms and Expression are Associated with Multiple Sclerosis Risk: Mechanistic Insights and Potential Clinical Impact. ACS Chem Neurosci 2020; 11:1651-1660. [PMID: 32348112 DOI: 10.1021/acschemneuro.0c00150] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of multiple sclerosis (MS) is influenced by the interaction of genetic and epigenetic mechanisms. The long noncoding RNA GAS5 acts as a competing endogenous RNA for microRNA-137 and is involved in demyelination. We investigated the association of GAS5 and miR-137 expression and their polymorphisms with MS susceptibility. One hundred and eight MS patients and 104 healthy controls were included. Expression analysis and genotyping of GAS5-rs2067079 and miR-137-rs1625579 single nucleotide polymorphisms were performed by qPCR. Serum GAS5 was upregulated, while serum miR-137 was downregulated in MS compared with the controls. Serum miR-137 was an excellent discriminator of MS patients from the controls (AUC = 0.97) and a negative independent predictor of MS in multivariate logistic analysis. Serum GAS5 expression was positively correlated with the expanded disability status scale scores in the relapsing-remitting MS patients. The rs2067079TT minor homozygote genotype was associated with an increased MS risk, while the rs1625579G minor allele was protective. rs1625579 showed an age-specific effect, while the rs2067079 affected the MS risk in gender- and age-specific manners. In MS patients, rs2067079TT was associated with a higher serum GAS5 than other genotypes, while serum miR-137 did not differ between rs1625579 genotypes. Our results suggest serum GAS5 and miR-137 as MS biomarkers, with miR-137 as a negative predictor of MS risk and GAS5 as a marker of MS severity. We propose rs2067079 and rs1625579 as novel genetic markers of MS susceptibility, and at least, rs2067079 possibly impacts the crosstalk between GAS5 and miR-137.
Collapse
Affiliation(s)
- Mahmoud A. Senousy
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Olfat G. Shaker
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Noha H. Sayed
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Nevine Fathy
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Mona A. Kortam
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
48
|
Kang J, Feng D, Yang F, Tian X, Han W, Jia H. Comparison of rapamycin and methylprednisolone for treating inflammatory muscle disease in a murine model of experimental autoimmune myositis. Exp Ther Med 2020; 20:219-226. [PMID: 32536994 PMCID: PMC7291653 DOI: 10.3892/etm.2020.8716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 12/09/2019] [Indexed: 01/03/2023] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a group of autoimmune inflammatory muscle diseases. Rapamycin has been shown to ameliorate inflammation and improve muscle function in a mouse model of experimental autoimmune myositis (EAM). In the present study, the therapeutic effect of rapamycin was compared with methylprednisolone (MP) on EAM. Mice were injected with myosin for 10 days to induce EAM and were subsequently treated with rapamycin (1.5 mg/kg), MP (40 mg/kg) or placebo (DMSO) for 14 days. The rapamycin-treated group exhibited significantly decreased severe inflammation and improved muscle strength compared with the MP-treated group. The plasma transforming growth factor-β (TGF-β) concentration in the rapamycin-treated group was significantly higher compared with the placebo group. However, both treatment groups exhibited significantly lower plasma interleukin-10 levels compared with the placebo group. Moreover, splenic regulatory T cell frequency in both the rapamycin- and MP-treated animals was significantly lower than that in the animals of the placebo group. Rapamycin showed better immune suppressive effects than MP in this model of EAM, and these effects were likely to be mediated by the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Juan Kang
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Dongyun Feng
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Feng Yang
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Xiaojia Tian
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Wenjuan Han
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China
| | - Hongge Jia
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xincheng, Xi'an, Shaanxi 710032, P.R. China.,Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518034, P.R. China
| |
Collapse
|
49
|
Petralia MC, Ciurleo R, Saraceno A, Pennisi M, Basile MS, Fagone P, Bramanti P, Nicoletti F, Cavalli E. Meta-Analysis of Transcriptomic Data of Dorsolateral Prefrontal Cortex and of Peripheral Blood Mononuclear Cells Identifies Altered Pathways in Schizophrenia. Genes (Basel) 2020; 11:genes11040390. [PMID: 32260267 PMCID: PMC7230488 DOI: 10.3390/genes11040390] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/13/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia (SCZ) is a psychiatric disorder characterized by both positive and negative symptoms, including cognitive dysfunction, decline in motivation, delusion and hallucinations. Antipsychotic agents are currently the standard of care treatment for SCZ. However, only about one-third of SCZ patients respond to antipsychotic medications. In the current study, we have performed a meta-analysis of publicly available whole-genome expression datasets on Brodmann area 46 of the brain dorsolateral prefrontal cortex in order to prioritize potential pathways underlying SCZ pathology. Moreover, we have evaluated whether the differentially expressed genes in SCZ belong to specific subsets of cell types. Finally, a cross-tissue comparison at both the gene and functional level was performed by analyzing the transcriptomic pattern of peripheral blood mononuclear cells of SCZ patients. Our study identified a robust disease-specific set of dysfunctional biological pathways characterizing SCZ patients that could in the future be exploited as potential therapeutic targets.
Collapse
Affiliation(s)
| | - Rosella Ciurleo
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (P.B.)
| | - Andrea Saraceno
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
- Correspondence: ; Tel.: +39-095-4781284
| | - Placido Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (P.B.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.S.); (M.P.); (M.S.B.); (F.N.); (E.C.)
| |
Collapse
|
50
|
Transcriptomic Analysis Reveals Abnormal Expression of Prion Disease Gene Pathway in Brains from Patients with Autism Spectrum Disorders. Brain Sci 2020; 10:brainsci10040200. [PMID: 32235346 PMCID: PMC7226514 DOI: 10.3390/brainsci10040200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/22/2022] Open
Abstract
The role of infections in the pathogenesis of autism spectrum disorder (ASD) is still controversial. In this study, we aimed to evaluate markers of infections and immune activation in ASD by performing a meta-analysis of publicly available whole-genome transcriptomic datasets of brain samples from autistic patients and otherwise normal people. Among the differentially expressed genes, no significant enrichment was observed for infectious diseases previously associated with ASD, including herpes simplex virus-1 (HSV-1), cytomegalovirus and Epstein–Barr virus in brain samples, nor was it found in peripheral blood from ASD patients. Interestingly, a significant number of genes belonging to the “prion diseases” pathway were found to be modulated in our ASD brain meta-analysis. Overall, our data do not support an association between infection and ASD. However, the data do provide support for the involvement of pathways related to other neurodegenerative diseases and give input to uncover novel pathogenetic mechanisms underlying ASD.
Collapse
|