1
|
Khaled N, Ibrahim N, Ali AE, Youssef FS, El-Ahmady SH. LC-qTOF-MS/MS phytochemical profiling of Tabebuia impetiginosa (Mart. Ex DC.) Standl. leaf and assessment of its neuroprotective potential in rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118292. [PMID: 38705428 DOI: 10.1016/j.jep.2024.118292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tabebuia impetiginosa (Bignoniaceae) was traditionally used for memory enhancement and central nervous system (CNS) stimulation. AIM OF THE STUDY This study aims to create a metabolic profile of the ethyl acetate fraction of T. impetiginosa (TEF) and investigate for the first time its neuroprotective potential on cyclophosphamide (CP)-induced chemobrain, validating its traditional use. MATERIALS AND METHODS Metabolite profiling of TEF was performed using Liquid Chromatography coupled with Quadrupole Time of Flight-Mass/Mass Spectrometry (LC-qTOF-MS/MS). For the in vivo study, CP (200 mg/kg, i.p.) was administered to induce cognitive impairment in rats; TEF (30 mg/kg, p.o.) was administered throughout the 14 days of the experiment to assess its role in mitigating CP-induced neuronal deficits. Behavioral tests including locomotor, Y-maze, and passive avoidance tests were conducted. Additionally, biochemical markers such as reduced glutathione (GSH), malondialdehyde (MDA), tumor necrosis factor-α (TNF-α), and caspase-3 immunoexpression were assessed in the hippocampus area. RESULTS Forty-four phytoconstituents were tentatively identified in TEF, mainly iridoids and organic acids. TEF showed significant memory enhancement as evidenced by the increase in step-through latency in the passive avoidance test by 1.5 folds and the increase in sequence alternation percentage (SAP) in the Y-maze test by 67.3%, as compared to CP-group. Moreover, it showed pronounced antioxidant and anti-inflammatory potentials evidenced by the significant elevation in reduced glutathione (GSH) levels by 80% and a pronounced decline in MDA and TNF-α levels by 24% and 45%, respectively relative to the CP group. TEF treatment restored normal hippocampal histological features and attenuated apoptotic caspase-3 expression by 70% compared to the CP group. CONCLUSIONS TEF can act as a promising natural scaffold in managing the chemobrain induced by CP in cancer patients.
Collapse
Affiliation(s)
- Nesma Khaled
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo, 11566, Egypt.
| | - Nehal Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo, 11566, Egypt.
| | - Alaa E Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| | - Fadia S Youssef
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo, 11566, Egypt.
| | - Sherweit H El-Ahmady
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo, 11566, Egypt.
| |
Collapse
|
2
|
Svandova E, Vesela B, Janeckova E, Chai Y, Matalova E. Exploring caspase functions in mouse models. Apoptosis 2024; 29:938-966. [PMID: 38824481 PMCID: PMC11263464 DOI: 10.1007/s10495-024-01976-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2024] [Indexed: 06/03/2024]
Abstract
Caspases are enzymes with protease activity. Despite being known for more than three decades, caspase investigation still yields surprising and fascinating information. Initially associated with cell death and inflammation, their functions have gradually been revealed to extend beyond, targeting pathways such as cell proliferation, migration, and differentiation. These processes are also associated with disease mechanisms, positioning caspases as potential targets for numerous pathologies including inflammatory, neurological, metabolic, or oncological conditions. While in vitro studies play a crucial role in elucidating molecular pathways, they lack the context of the body's complexity. Therefore, laboratory animals are an indispensable part of successfully understanding and applying caspase networks. This paper aims to summarize and discuss recent knowledge, understanding, and challenges in caspase knock-out mice.
Collapse
Affiliation(s)
- Eva Svandova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic.
| | - Barbora Vesela
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic
| | - Eva Janeckova
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, USA
| | - Eva Matalova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic
- Department of Physiology, University of Veterinary Sciences, Brno, Czech Republic
| |
Collapse
|
3
|
Lee D, Lee SY, Ra MJ, Jung SM, Yu JN, Kang KS, Kim KH. Cancer therapeutic potential of hovetrichoside C from Jatropha podagrica on apoptosis of MDA-MB-231 human breast cancer cells. Food Chem Toxicol 2024; 190:114794. [PMID: 38849046 DOI: 10.1016/j.fct.2024.114794] [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: 01/04/2024] [Revised: 05/18/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Phytochemical analysis of the methanolic extracts of Jatropha podagrica stalks and roots using liquid chromatography-mass spectrometry (LC-MS) led to the isolation of six compounds: corchoionoside C (1), isobiflorin (2), fraxin (3), hovetrichoside C (4), fraxetin (5), and corillagin (6). The isolated compounds (1-6) were tested for their cytotoxicity against MDA-MB-231 human breast cancer cells. Remarkably, compound 4 (hovetrichoside C) exhibited robust cytotoxicity against MDA-MB-231 cells, displaying an IC50 value of 50.26 ± 1.22 μM, along with an apoptotic cell death rate of 24.21 ± 2.08% at 100 μM. Treatment involving compound 4 amplified protein levels of cleaved caspase-8, -9, -3, -7, BH3-interacting domain death agonist (Bid), Bcl-2-associated X protein (Bax), and cleaved poly (ADP-ribose) polymerase (cleaved PARP), while concurrently reducing B-cell lymphoma 2 (Bcl-2) levels. In totality, these findings underscore that hovetrichoside C (4) possesses anti-breast cancer activity that revolves around apoptosis induction via both extrinsic and intrinsic signaling pathways.
Collapse
Affiliation(s)
- Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam, 13120, Republic of Korea
| | - Seo Yoon Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Moon-Jin Ra
- Hongcheon Institute of Medicinal Herb, Hongcheon-gun, Gangwon-do, 25142, Republic of Korea
| | - Sang-Mi Jung
- Hongcheon Institute of Medicinal Herb, Hongcheon-gun, Gangwon-do, 25142, Republic of Korea
| | - Jeong-Nam Yu
- Nakdonggang National Institute of Biological Resources, Sangju, Gyeongsangbuk-do, 37242, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam, 13120, Republic of Korea.
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
4
|
Lee SW, Oh YM, Victor MB, Yang Y, Chen S, Strunilin I, Dahiya S, Dolle RE, Pak SC, Silverman GA, Perlmutter DH, Yoo AS. Longitudinal modeling of human neuronal aging reveals the contribution of the RCAN1-TFEB pathway to Huntington's disease neurodegeneration. NATURE AGING 2024; 4:95-109. [PMID: 38066314 PMCID: PMC11456361 DOI: 10.1038/s43587-023-00538-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 11/03/2023] [Indexed: 12/19/2023]
Abstract
Aging is a common risk factor in neurodegenerative disorders. Investigating neuronal aging in an isogenic background stands to facilitate analysis of the interplay between neuronal aging and neurodegeneration. Here we perform direct neuronal reprogramming of longitudinally collected human fibroblasts to reveal genetic pathways altered at different ages. Comparative transcriptome analysis of longitudinally aged striatal medium spiny neurons (MSNs) in Huntington's disease identified pathways involving RCAN1, a negative regulator of calcineurin. Notably, RCAN1 protein increased with age in reprogrammed MSNs as well as in human postmortem striatum and RCAN1 knockdown rescued patient-derived MSNs of Huntington's disease from degeneration. RCAN1 knockdown enhanced chromatin accessibility of genes involved in longevity and autophagy, mediated through enhanced calcineurin activity, leading to TFEB's nuclear localization by dephosphorylation. Furthermore, G2-115, an analog of glibenclamide with autophagy-enhancing activities, reduced the RCAN1-calcineurin interaction, phenocopying the effect of RCAN1 knockdown. Our results demonstrate that targeting RCAN1 genetically or pharmacologically can increase neuronal resilience in Huntington's disease.
Collapse
Affiliation(s)
- Seong Won Lee
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Sciences, Mercer University School of Medicine, Columbus, GA, USA
| | - Young Mi Oh
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Sciences, Mercer University School of Medicine, Columbus, GA, USA
| | - Matheus B Victor
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yan Yang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shawei Chen
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ilya Strunilin
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Roland E Dolle
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephen C Pak
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Gary A Silverman
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Perlmutter
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
5
|
Girija D, Deepa K, Chubicka T, Shidhi PR, Hussan S, Raghavamenon AC, Babu TD. Structural and functional validation of a cloned parasporin from Bacillus thuringiensis isolate KAU 41 native to Western Ghats of India. Proteins 2023; 91:1487-1495. [PMID: 37401522 DOI: 10.1002/prot.26544] [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: 05/09/2022] [Revised: 04/20/2023] [Accepted: 06/14/2023] [Indexed: 07/05/2023]
Abstract
Parasporins of Bacillus thuringiensis (Bt) exhibit specific toxicity to cancer cells. PCR based mining has identified apoptosis inducing parasporin in KAU41 Bt isolate from the Western Ghats of India. The study aimed to clone and overexpress the parasporin of native KAU41 Bt isolate for determining structural and functional characteristics of the protein. Parasporin gene was cloned in pGEM-T, sequenced, sub-cloned in pET30+ and overexpressed in Escherichia coli. The expressed protein was characterized by SDS-PAGE and in silico methods. Cytotoxicity of cleaved peptide was assessed by MTT assay. SDS-PAGE displayed a 31 kDa protein (rp-KAU41) overexpressed. Upon proteinase K digestion, the protein was cleaved into 29 kDa peptide which was found to be cytotoxic to HeLa cells. The protein has a deduced sequence of 267 amino acids with β-strands folding pattern of crystal protein. Even though rp-KAU41 shared a 99.15% identity to chain-A of non-toxic crystal protein, it only showed a less similarity to the existing parasporins like PS4 (38%) and PS5 (24%) in UPGMA analysis, emphasizing the novelty of rp-KAU41. The protein is predicted to have more similarity to the pore forming toxins of Aerolysin superfamily and an additional loop in rp-KAU41 may be contributing towards its cytotoxicity. The molecular docking with caspase 3 resulted in higher Z dock and Z rank score substantiating its role in the activation of intrinsic pathway of apoptosis. The recombinant parasporin protein, rp-KAU41 is presumed to belong to the Aerolysin superfamily. An interaction with caspase 3 substantiates its role in activating the intrinsic pathway of apoptosis in cancer cells.
Collapse
Affiliation(s)
- Devaki Girija
- Department of Agricultural Microbiology, College of Agriculture, Kerala Agricultural University, Thrissur, Kerala, India
| | - Kizhakkeettil Deepa
- Department of Agricultural Microbiology, College of Agriculture, Kerala Agricultural University, Thrissur, Kerala, India
| | - Thomas Chubicka
- Department of Biochemistry, Amala Cancer Research Centre, Thrissur, Kerala, India
| | - P R Shidhi
- Department of Computational Biology and Bioinformatics, North Campus-Karyavattam, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Safna Hussan
- Department of Biochemistry, Amala Cancer Research Centre, Thrissur, Kerala, India
| | | | | |
Collapse
|
6
|
Resveratrol, Endocrine Disrupting Chemicals, Neurodegenerative Diseases and Depression: Genes, Transcription Factors, microRNAs, and Sponges Involved. Neurochem Res 2023; 48:604-624. [PMID: 36245065 DOI: 10.1007/s11064-022-03787-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/05/2022] [Accepted: 10/06/2022] [Indexed: 02/04/2023]
Abstract
We aimed to examine the molecular basis of the positive effect of resveratrol against amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), cognitive impairment (CI), and depression induced by a mixture of bisphenol A (BPA), BPS, and BPF. The CTD, GeneMania, Metascape, SwissADME, Cytoscape, MIENTURNET, miRNAsong, and Autodock Vina were the fundamental tools for analysis. Resveratrol exerts its protective effects on selected diseases induced by a mixture of BPA, BPS, and BPF through the following genes: PTGS2 and GSR for ALS; INS, IL6, BDNF, and SOD1 for PD; BDNF, CASP3, TNF, INS, IGF1, IL1B for CI; and BDNF, PTGS2, and IL6 for depression. Detoxification was noted as the most important for ALS, dopamine metabolism for PD, apoptosis for CI, and the selenium micronutrient network for depression. hsa-miR-377-3p, hsa-miR-1-3p, hsa-miR-128-3p, and hsa-miR-204-5p were highlighted. We created and tested in silico sponges that inhibited these miRNAs. NFE2L2, BACH1, PPARG, and NR4A3 were listed as the key transcription factors implicated in resveratrol's protective effect against harmful studied chemicals. Furthermore, resveratrol's physicochemical properties and pharmacokinetics are consistent with its therapeutic benefits in ALS, PD, CI, and depression, owing to its high gastrointestinal absorption, drug-likeness, non-P-glycoprotein substrate, and capacity to penetrate the blood-brain barrier.
Collapse
|
7
|
The Effective Components, Core Targets, and Key Pathways of Ginseng against Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:9935942. [PMID: 36726526 PMCID: PMC9886485 DOI: 10.1155/2023/9935942] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 01/24/2023]
Abstract
Background Panax ginseng C. A. Mey (ginseng) is a traditional Chinese medicinal herb used for the treatment of nervous system disorders, such as Alzheimer's disease (AD). However, the pharmacological mechanisms of ginseng involved in AD have not been systematically investigated. Here, a network pharmacology approach was adopted to explore the effective components, core targets, and key pathways of ginseng against AD. Methods TCMSP database was used to screen the active ingredients of ginseng. Prediction of the targets of ginseng and AD-related genes was performed using online public databases. "Compound-Target," "Compound-Target-Disease," "Protein-Protein Interaction (PPI)," "Compound-Target-Pathway," and "Compound-Target-GO-Pathway" networks were constructed with Cytoscape 3.7.2 software. Gene Ontology (GO) function annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were performed by using the DAVID database. Results A total of 22 bioactive compounds were identified from ginseng, and 481 targets of ginseng and 763 AD-related targets were obtained from public databases. The PPI network screened out 19 hub genes of ginseng against AD. According to GO function enrichment, ginseng influenced cell proliferation, death, the nitric oxide biosynthetic process, hypoxia response, and synaptic transmission. Neuroactive ligand-receptor interaction, serotonergic synapse, calcium signaling, cAMP signaling, FoxO signaling, Ras signaling, and PI3K-AKT signaling were among the most key regulatory pathways. The compound-target-GO-route network found EGFR, MAPK1, MAPK14, AKT1, CASP3, and PRKACA as key genes, with PI3K-AKT signaling being the most important pathway for ginseng's anti-AD activity. Conclusion Ginseng exerts neuroprotective effects in AD patients through multicomponent, multitarget, and multipathway modes, providing novel insight into the pharmacological and experimental research on ginseng against AD.
Collapse
|
8
|
Morén C, Treder N, Martínez-Pinteño A, Rodríguez N, Arbelo N, Madero S, Gómez M, Mas S, Gassó P, Parellada E. Systematic Review of the Therapeutic Role of Apoptotic Inhibitors in Neurodegeneration and Their Potential Use in Schizophrenia. Antioxidants (Basel) 2022; 11:2275. [PMID: 36421461 PMCID: PMC9686909 DOI: 10.3390/antiox11112275] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 09/15/2023] Open
Abstract
Schizophrenia (SZ) is a deleterious brain disorder affecting cognition, emotion and reality perception. The most widely accepted neurochemical-hypothesis is the imbalance of neurotransmitter-systems. Depleted GABAergic-inhibitory function might produce a regionally-located dopaminergic and glutamatergic-storm in the brain. The dopaminergic-release may underlie the positive psychotic-symptoms while the glutamatergic-release could prompt the primary negative symptoms/cognitive deficits. This may occur due to excessive synaptic-pruning during the neurodevelopmental stages of adolescence/early adulthood. Thus, although SZ is not a neurodegenerative disease, it has been suggested that exaggerated dendritic-apoptosis could explain the limited neuroprogression around its onset. This apoptotic nature of SZ highlights the potential therapeutic action of anti-apoptotic drugs, especially at prodromal stages. If dysregulation of apoptotic mechanisms underlies the molecular basis of SZ, then anti-apoptotic molecules could be a prodromal therapeutic option to halt or prevent SZ. In fact, risk alleles related in apoptotic genes have been recently associated to SZ and shared molecular apoptotic changes are common in the main neurodegenerative disorders and SZ. PRISMA-guidelines were considered. Anti-apoptotic drugs are commonly applied in classic neurodegenerative disorders with promising results. Despite both the apoptotic-hallmarks of SZ and the widespread use of anti-apoptotic targets in neurodegeneration, there is a strikingly scarce number of studies investigating anti-apoptotic approaches in SZ. We analyzed the anti-apoptotic approaches conducted in neurodegeneration and the potential applications of such anti-apoptotic therapies as a promising novel therapeutic strategy, especially during early stages.
Collapse
Affiliation(s)
- Constanza Morén
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- U722 Group, Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Carlos III Health Institute, 28029 Madrid, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Nina Treder
- Faculty of Psychology and Neuroscience, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Albert Martínez-Pinteño
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Natàlia Rodríguez
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Néstor Arbelo
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Santiago Madero
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Marta Gómez
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
- Department of Psychiatry, Servizo Galego de Saúde (SERGAS), 36001 Pontevedra, Spain
| | - Sergi Mas
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Patricia Gassó
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Eduard Parellada
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
9
|
Oh YM, Lee SW, Kim WK, Chen S, Church VA, Cates K, Li T, Zhang B, Dolle RE, Dahiya S, Pak SC, Silverman GA, Perlmutter DH, Yoo AS. Age-related Huntington's disease progression modeled in directly reprogrammed patient-derived striatal neurons highlights impaired autophagy. Nat Neurosci 2022; 25:1420-1433. [PMID: 36303071 PMCID: PMC10162007 DOI: 10.1038/s41593-022-01185-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/19/2022] [Indexed: 01/13/2023]
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder with adult-onset clinical symptoms, but the mechanism by which aging drives the onset of neurodegeneration in patients with HD remains unclear. In this study we examined striatal medium spiny neurons (MSNs) directly reprogrammed from fibroblasts of patients with HD to model the age-dependent onset of pathology. We found that pronounced neuronal death occurred selectively in reprogrammed MSNs from symptomatic patients with HD (HD-MSNs) compared to MSNs derived from younger, pre-symptomatic patients (pre-HD-MSNs) and control MSNs from age-matched healthy individuals. We observed age-associated alterations in chromatin accessibility between HD-MSNs and pre-HD-MSNs and identified miR-29b-3p, whose age-associated upregulation promotes HD-MSN degeneration by impairing autophagic function through human-specific targeting of the STAT3 3' untranslated region. Reducing miR-29b-3p or chemically promoting autophagy increased the resilience of HD-MSNs against neurodegeneration. Our results demonstrate miRNA upregulation with aging in HD as a detrimental process driving MSN degeneration and potential approaches for enhancing autophagy and resilience of HD-MSNs.
Collapse
Affiliation(s)
- Young Mi Oh
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Seong Won Lee
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Woo Kyung Kim
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shawei Chen
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Victoria A Church
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kitra Cates
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tiandao Li
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bo Zhang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Roland E Dolle
- Department of Biochemistry, Washington University School of Medicine, St. Louis, MO, USA
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephen C Pak
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Gary A Silverman
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Perlmutter
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
10
|
Reddy DS, Abeygunaratne HN. Experimental and Clinical Biomarkers for Progressive Evaluation of Neuropathology and Therapeutic Interventions for Acute and Chronic Neurological Disorders. Int J Mol Sci 2022; 23:11734. [PMID: 36233034 PMCID: PMC9570151 DOI: 10.3390/ijms231911734] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/27/2022] Open
Abstract
This article describes commonly used experimental and clinical biomarkers of neuronal injury and neurodegeneration for the evaluation of neuropathology and monitoring of therapeutic interventions. Biomarkers are vital for diagnostics of brain disease and therapeutic monitoring. A biomarker can be objectively measured and evaluated as a proxy indicator for the pathophysiological process or response to therapeutic interventions. There are complex hurdles in understanding the molecular pathophysiology of neurological disorders and the ability to diagnose them at initial stages. Novel biomarkers for neurological diseases may surpass these issues, especially for early identification of disease risk. Validated biomarkers can measure the severity and progression of both acute neuronal injury and chronic neurological diseases such as epilepsy, migraine, Alzheimer's disease, Parkinson's disease, Huntington's disease, traumatic brain injury, amyotrophic lateral sclerosis, multiple sclerosis, and other brain diseases. Biomarkers are deployed to study progression and response to treatment, including noninvasive imaging tools for both acute and chronic brain conditions. Neuronal biomarkers are classified into four core subtypes: blood-based, immunohistochemical-based, neuroimaging-based, and electrophysiological biomarkers. Neuronal conditions have progressive stages, such as acute injury, inflammation, neurodegeneration, and neurogenesis, which can serve as indices of pathological status. Biomarkers are critical for the targeted identification of specific molecules, cells, tissues, or proteins that dramatically alter throughout the progression of brain conditions. There has been tremendous progress with biomarkers in acute conditions and chronic diseases affecting the central nervous system.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Institute of Pharmacology and Neurotherapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Intercollegiate School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Hasara Nethma Abeygunaratne
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Institute of Pharmacology and Neurotherapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| |
Collapse
|
11
|
Ashraf GM, Rehan M, Alsayed AO, Somvanshi P, Haque S. Drug repurposing against galectin-3 using simulation-based studies. J Biomol Struct Dyn 2022:1-8. [PMID: 36184598 DOI: 10.1080/07391102.2022.2120538] [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: 10/07/2022]
Abstract
The protein galectin, which binds to carbohydrates and is involved in a number of therapeutic processes including cell proliferation, inflammatory responses, apoptosis, etc., has been discovered as a potential therapeutic target. Galectin-3 is a stable biomarker that exhibits both increased and decreased expression in a variety of illnesses and infections, regardless of sex, age, or body mass index. The goal of the current study is to apply bioinformatics techniques to examine the possibility of cardiovascular medications to inhibit Galectin-3-related biological activities. Unsupervised clustering techniques, molecular docking, and guided molecular dynamics (MD) simulation were used to create a computational pipeline that was used to screen potential chemical compounds from a library of chemical compounds with related molecular fingerprints. Utilizing input factors such as gene expression, mode of action, and chemical descriptors, clustering enables prioritization of medicinal molecules. Twenty-four compounds were screened and repurposed against Galectin-3 utilizing molecular docking as part of the cluster-facilitated virtual screening technique. The polar interactions that Arg144, Glu184, Arg162, His158, and Asn174 have with Bufalin, Cymarin, and Ouabalin have the highest binding affinities, according to docking studies. Studies using MD simulations confirm the tested compounds' ability to inhibit Galectin-3. Galactin-3 targeted experimental and in vivo animal model-based validation studies using Bufalin, Cymarin, and Ouabalin are also necessary.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohd Rehan
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alhuseen O Alsayed
- Deanship of Scientific Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Pallavi Somvanshi
- School of Computational & Integrative Sciences (SC&IS), Jawaharlal Nehru University, New Delhi, India.,Special Centre of Systems Medicine (SCSM), Jawaharlal Nehru University, New Delhi, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
12
|
Restored Fyn Levels in Huntington’s Disease Contributes to Enhanced Synaptic GluN2B-Composed NMDA Receptors and CREB Activity. Cells 2022; 11:cells11193063. [PMID: 36231023 PMCID: PMC9563007 DOI: 10.3390/cells11193063] [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: 09/03/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 11/20/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are important postsynaptic receptors that contribute to normal synaptic function and cell survival; however, when overactivated, as in Huntington’s disease (HD), NMDARs cause excitotoxicity. HD-affected striatal neurons show altered NMDAR currents and augmented ratio of surface to internal GluN2B-containing NMDARs, with augmented accumulation at extrasynaptic sites. Fyn protein is a member of the Src kinase family (SKF) with an important role in NMDARs phosphorylation and synaptic localization and function; recently, we demonstrated that Fyn is reduced in several HD models. Thus, in this study, we aimed to explore the impact of HD-mediated altered Fyn levels at post-synaptic density (PSD), and their role in distorted NMDARs function and localization, and intracellular neuroprotective pathways in YAC128 mouse primary striatal neurons. We show that reduced synaptic Fyn levels and activity in HD mouse striatal neurons is related to decreased phosphorylation of synaptic GluN2B-composed NMDARs; this occurs concomitantly with augmented extrasynaptic NMDARs activity and currents and reduced cAMP response element-binding protein (CREB) activation, along with induction of cell death pathways. Importantly, expression of a constitutive active form of SKF reestablishes NMDARs localization, phosphorylation, and function at PSD in YAC128 mouse neurons. Enhanced SKF levels and activity also promotes CREB activation and reduces caspase-3 activation in YAC128 mouse striatal neurons. This work supports, for the first time, a relevant role for Fyn protein in PSD modulation, controlling NMDARs synaptic function in HD, and favoring neuroprotective pathways and cell survival. In this respect, Fyn Tyr kinase constitutes an important potential HD therapeutic target directly acting at PSD.
Collapse
|
13
|
Salama A, Elgohary R, M Amin M, Elwahab SA. Immunomodulatory effect of protocatechuic acid on cyclophosphamide induced brain injury in rat: Modulation of inflammosomes NLRP3 and SIRT1. Eur J Pharmacol 2022; 932:175217. [PMID: 36007603 DOI: 10.1016/j.ejphar.2022.175217] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/13/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022]
Abstract
Modulation of the inflammasome NLRP3 and SIRT1 are new combat strategy for brain injury protection. The inflammasome activates proinflammatory cytokines releasing interleukin-1β and interleukin-18 which in turn affect the toxins release from immune cells. In addition, SIRT1 controls many biological functions, such as immune response and oxidative stress. Protocatechuic has versatile biological activities and possesses antioxidant, anti-inflammatory and neuroprotective effects. So this work aims to study immunomodulatory effect of protocatechuic acid on cyclophosphamide chemotherapy drug-induced brain injury via modulation of inflammosomes NLRP3 and SIRT1. Rats were randomly assigned to four experimental groups. Normal control group was injected with a single i.p injection of saline. Cyclophosphamide group was injected with a single i.p injection of cyclophosphamide (200 mg/kg). Protocatechuic acid groups were orally administered (50 &100 mg/kg) once daily for 10 consecutive days after cyclophosphamide injection. Protocatechuic acid administration exhibited improvements of the cognition function and memory, a reduction in brain contents of MDA, NLRP3, IL-1 β, NF-κB, IKBKB and Galectin 3 and an elevation of GSH and SIRT1 compared to cyclophosphamide group. In addition, protocatechuic acid administration ameliorated the elevation of caspase 3 and iNOS gene expression and alleviated the neuron degeneration caused by cyclophosphamide. In conclusion, the therapeutic action of protocatechuic acid and its cellular and molecular mechanisms are new insights against various human ailments, especially, neuroprotective disease as brain injury induced by cyclophosphamide chemotherapy drug in rats through modulation of inflammosomes NLRP3 and SIRT1.
Collapse
Affiliation(s)
- Abeer Salama
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El Buhouth St. (Former El-Tahrir St.), 12622, Dokki, Cairo, Egypt
| | - Rania Elgohary
- Narcotics, Ergogenics and Poisons Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El Buhouth St. (Former El-Tahrir St.), 12622, Dokki, Cairo, Egypt.
| | - Mohamed M Amin
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El Buhouth St. (Former El-Tahrir St.), 12622, Dokki, Cairo, Egypt
| | - Sahar Abd Elwahab
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
14
|
Shabbir A, Rehman K, Akbar M, Hamid Akash MS. Neuroprotective potential of curcuminoids in modulating Alzheimer's Disease via multiple signaling pathways. Curr Med Chem 2022; 29:5560-5581. [PMID: 35674299 DOI: 10.2174/0929867329666220607161328] [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: 11/16/2021] [Revised: 02/12/2022] [Accepted: 03/15/2022] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a progressive and frequent neurodegenerative disease of elderly people. In the 21st century, owing to the increasing prevalence of AD, there is a crucial need for finding better and effective pharmacotherapeutic approaches. This review article demonstrated the various sources and possible metabolic pathways of curcuminoids obtained from Curcuma longa herb, to prevent and treat AD but the information related to the metabolic fate of curcuminoids is deficient. Different in vitro and in vivo research studies demonstrating the mechanisms by which curcuminoids attenuated AD have been summarized. Administration of curcuminoids has been indicated to inhibit hyperphosphorylation of tau protein, deposition, and oligomerization of amyloid beta plaques in several AD models. Curcuminoids also chelate metals and form complexes, have antioxidant properties, mediates neuroinflammatory signaling pathways by modifying microglial cells activity, inhibit acetylcholinesterase activities and also modulates other associated signaling pathways including insulin signaling pathways and heme-oxygenase pathway. Briefly curcuminoids exhibit the capability to be more productive and efficacious compared to many recent treatments due to their antioxidant, delayed neuron degeneration and anti-inflammatory potential. Although their effectiveness as a curative agent is considered to be reduced due to their low bioavailability, If the issue of curcuminoids' low bioavailability is resolved then curcuminoid-based medications are hopefully on the horizon against AD.
Collapse
Affiliation(s)
- Anam Shabbir
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Moazzama Akbar
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
15
|
Eskandari E, Eaves CJ. Paradoxical roles of caspase-3 in regulating cell survival, proliferation, and tumorigenesis. J Cell Biol 2022; 221:213213. [PMID: 35551578 PMCID: PMC9106709 DOI: 10.1083/jcb.202201159] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 11/22/2022] Open
Abstract
Caspase-3 is a widely expressed member of a conserved family of proteins, generally recognized for their activated proteolytic roles in the execution of apoptosis in cells responding to specific extrinsic or intrinsic inducers of this mode of cell death. However, accumulating evidence indicates that caspase-3 also plays key roles in regulating the growth and homeostatic maintenance of both normal and malignant cells and tissues in multicellular organisms. Given that yeast possess an ancestral caspase-like gene suggests that the caspase-3 protein may have acquired different functions later during evolution to better meet the needs of more complex multicellular organisms, but without necessarily losing all of the functions of its ancestral yeast precursor. This review provides an update on what has been learned about these interesting dichotomous roles of caspase-3, their evolution, and their potential relevance to malignant as well as normal cell biology.
Collapse
Affiliation(s)
- Ebrahim Eskandari
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Connie J. Eaves
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada,Correspondence to Connie J. Eaves:
| |
Collapse
|
16
|
Ahmed YH, AbuBakr HO, Ahmad IM, Ahmed ZSO. Histopathological, Immunohistochemical, And Molecular Alterations In Brain Tissue And Submandibular Salivary Gland Of Atrazine-Induced Toxicity In Male Rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:30697-30711. [PMID: 34994930 DOI: 10.1007/s11356-021-18399-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/25/2021] [Indexed: 06/14/2023]
Abstract
Atrazine (ATZ) is herbicide that has been widely used for different crops. This extensive use has resulted in severe deleterious effects in different species. In this work, we investigated the potentially harmful effect of atrazine herbicide on the brain and submandibular salivary gland. Our investigation was carried out on 20 adult male albino rats that were equally divided into two groups. The first group received distilled water as control, while the second group received ATZ at 200 mg/kg body weight/ day via stomach gavage for 30 successive days of the experiment; the oral LD50 for ATZ is 3090 mg/kg. Our findings revealed the ability of ATZ to cause damage to the cerebrum, hippocampus, and submandibular salivary gland. This damage resulted from the induced oxidative stress, which was indicated by a significant elevation in malondialdehyde (MDA) concentration, DNA fragmentation, tumor necrotic factor-alpha (TNF-α) expression, with a significant decrease in reduced glutathione (GSH) level and reduction of B cell lymphoma 2 (BCL2), dopamine receptor D1 (Drd1), cAMP-responsive element-binding protein 1 (Creb1) genes expression after ATZ exposure. Moreover, degeneration of cells, cytoplasmic vacuolation, congestion of blood vessels, a strong immune reaction to caspase 3, and negligible immune expression of a glial fibrillary acidic protein (GFAP) were also noticed in the ATZ-treated group. We concluded that ATZ induces oxidative stress and has a toxic and apoptotic effects on the cerebrum, hippocampus, and salivary gland of adult male albino rats.
Collapse
Affiliation(s)
- Yasmine H Ahmed
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Huda O AbuBakr
- Biochemistry and Chemistry of Nutrition Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Ismail M Ahmad
- Biochemistry and Chemistry of Nutrition Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Zainab Sabry Othman Ahmed
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
- King Salman International University, Ras Sudr, South Sinai, Egypt.
| |
Collapse
|
17
|
Dhani S, Zhao Y, Zhivotovsky B. A long way to go: caspase inhibitors in clinical use. Cell Death Dis 2021; 12:949. [PMID: 34654807 PMCID: PMC8519909 DOI: 10.1038/s41419-021-04240-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/15/2021] [Accepted: 09/28/2021] [Indexed: 12/19/2022]
Abstract
Caspases are an evolutionary conserved family of cysteine-dependent proteases that are involved in many vital cellular processes including apoptosis, proliferation, differentiation and inflammatory response. Dysregulation of caspase-mediated apoptosis and inflammation has been linked to the pathogenesis of various diseases such as inflammatory diseases, neurological disorders, metabolic diseases, and cancer. Multiple caspase inhibitors have been designed and synthesized as a potential therapeutic tool for the treatment of cell death-related pathologies. However, only a few have progressed to clinical trials because of the consistent challenges faced amongst the different types of caspase inhibitors used for the treatment of the various pathologies, namely an inadequate efficacy, poor target specificity, or adverse side effects. Importantly, a large proportion of this failure lies in the lack of understanding various caspase functions. To overcome the current challenges, further studies on understanding caspase function in a disease model is a fundamental requirement to effectively develop their inhibitors as a treatment for the different pathologies. Therefore, the present review focuses on the descriptive properties and characteristics of caspase inhibitors known to date, and their therapeutic application in animal and clinical studies. In addition, a brief discussion on the achievements, and current challenges faced, are presented in support to providing more perspectives for further development of successful therapeutic caspase inhibitors for various diseases.
Collapse
Affiliation(s)
- Shanel Dhani
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden
| | - Yun Zhao
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden
| | - Boris Zhivotovsky
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden.
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia.
| |
Collapse
|
18
|
Bashir DW, Rashad MM, Ahmed YH, Drweesh EA, Elzahany EAM, Abou-El-Sherbini KS, El-Leithy EMM. The ameliorative effect of nanoselenium on histopathological and biochemical alterations induced by melamine toxicity on the brain of adult male albino rats. Neurotoxicology 2021; 86:37-51. [PMID: 34216684 DOI: 10.1016/j.neuro.2021.06.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/16/2021] [Accepted: 06/25/2021] [Indexed: 01/01/2023]
Abstract
Melamine is a chemical substance used as a food adulterant because of its high nitrogen content; it is known to induce neurotoxicity, thereby adversely affecting the central nervous system. The biocompatibility, bioavailability, lower toxicity, and the large surface area of nanosized selenium relative to its other forms indicate that selenium nanoparticles (SeNPs) have a potential ameliorative effect against melamine-induced neurotoxicity. In this study, we tested this hypothesis using 40 adult male albino rats that were randomly assigned into four groups (n = 10 per group): group I rats served as the untreated negative controls and were fed with standard diet and distilled water; group II rats were orally treated with melamine (300 mg/kg body weight/d); group III rats orally received melamine (300 mg/kg body weight/d) and SeNPs (2 mg/kg body weight/d); and group IV rats received SeNPs only (2 mg/kg body weight/d) for 28 days. Blood and brain samples were collected from all rats and processed for biochemical, histopathological, and immunohistochemical investigations. SeNPs were encapsulated in starch as a natural stabilizer and a size-controlling agent (SeNP@starch). The prepared SeNPs were characterized using different techniques. Inductively coupled plasma-optical emission spectrometry (ICP-OES) indicated that the percentage of selenium loaded in starch was 1.888 %. Powder x-ray diffractometer (XRD) was used to investigate the crystalline structure of the Se-NP@starch, to be tubular and composed of amorphous starch as well as metallic selenium. Thermogravimetric analysis confirmed the thermal stability of the product and determined the interactions among the different components. Transmission electron microscope demonstrated the spherical shape of SeNPs and their dispersion into starch surface as well as evaluating their size in nanoscale (range 20-140 nm). Our results revealed that the melamine- exposed rats had significantly elevated in malondialdehyde levels, significantly reduced in total antioxidant capacity, down-regulated expression of the antioxidant related genes Nrf2 (nuclear factor erythroid 2-related factor 2) and GPx (glutathione peroxidase), as well as up-regulated expression of the apoptosis-related gene Bax (B-cell lymphoma 2-associated X protein), with down regulation of Bcl-2 (B-cell lymphoma 2). Histopathological examination exhibited several alterations in the cerebrum, cerebellum, and hippocampus of the treated rats compared with the controls. Neuronal degeneration, vacuolation of the neuropils, and pericellular and perivascular spaces were observed. In addition, the pyramidal and granular cell layers of the hippocampus and cerebellum, respectively, were found to have significantly reduced thickness. Furthermore, a significant decrease in the percentage area of the glial fibrillary acidic protein and a significant increase in the percentage area of caspase-3 were noted. On the other hand, co-treatment with SeNPs partially ameliorated these alterations. A significant reduction in malondialdehyde levels; a non- significant elevation in total antioxidant capacity; up-regulation, upregulation of Nrf2, GPx, and Bcl-2 and downregulation of Bax were recorded. Neuronal degeneration, vacuolation of neuropils, and pericellular spaces were reduced. The pyramidal and granular cell layers restored their normal thickness. The percentage area of the glial fibrillary acidic protein significantly increased, whereas that of caspase-3 significantly decreased. In conclusion, SeNPs have an ameliorative effect against melamine-induced neurotoxicity in albino rats.
Collapse
Affiliation(s)
- Dina W Bashir
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Maha M Rashad
- Biochemistry and Chemistry of Nutrition Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Yasmine H Ahmed
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| | - Elsayed A Drweesh
- Department of Inorganic Chemistry, National Research Centre, Giza, Egypt
| | - Eman A M Elzahany
- Department of Inorganic Chemistry, National Research Centre, Giza, Egypt
| | | | - Ebtihal M M El-Leithy
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
19
|
Association of dietary intake, medication and anthropometric indices with serum levels of advanced glycation end products, caspase-3, and matrix metalloproteinase-9 in diabetic patients. J Diabetes Metab Disord 2021; 20:719-725. [PMID: 34222087 DOI: 10.1007/s40200-021-00803-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
Background and objective Increased serum levels of advanced glycation end products (AGEs), caspase-3 (Cas-3) and matrix metalloproteinase-9 (MMP-9) have been reported in diabetic patients. This study aimed to evaluate association of anthropometric, dietary, and therapeutic factors with serum levels of methylglyoxal (MGO), carboxymethyl lysine (CML), pentosidine (Pen), Cas-3, and MMP-9 in diabetic patients. Methods The current study included 36 diabetic subjects. Dietary intake of the participants was assessed using three-day 24-h recall survey and anthropometric indices were measured. Demographic factors and medication intake of every subject were obtained. Serum levels of CML, MGO, Pen, MMP-9, and Cas-3 were measured using ELISA method. Results Gliclazide consumption was positively correlated with MMP-9 and Cas-3, but not AGEs levels. Females had higher MGO level compared with males. Further, CML levels were negatively correlated with BMI and WHR. Dietary protein intake was positively correlated with MMP-9, Cas-3, and MGO levels. As well as dietary energy and fat intake had significant positive relationship with serum Cas-3 concentration. Conclusion It is concluded that anthropometric characteristics, dietary intake, and therapeutic medications are possible factors that may determine the circulating levels of AGEs, MMP-9, and Cas-3 in patients with diabetes.
Collapse
|
20
|
Maitra U, Harding T, Liang Q, Ciesla L. GardeninA confers neuroprotection against environmental toxin in a Drosophila model of Parkinson's disease. Commun Biol 2021; 4:162. [PMID: 33547411 PMCID: PMC7864937 DOI: 10.1038/s42003-021-01685-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 01/11/2021] [Indexed: 02/08/2023] Open
Abstract
Parkinson’s disease is an age-associated neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons from the midbrain. Epidemiological studies have implicated exposures to environmental toxins like the herbicide paraquat as major contributors to Parkinson’s disease etiology in both mammalian and invertebrate models. We have employed a paraquat-induced Parkinson’s disease model in Drosophila as an inexpensive in vivo platform to screen therapeutics from natural products. We have identified the polymethoxyflavonoid, GardeninA, with neuroprotective potential against paraquat-induced parkinsonian symptoms involving reduced survival, mobility defects, and loss of dopaminergic neurons. GardeninA-mediated neuroprotection is not solely dependent on its antioxidant activities but also involves modulation of the neuroinflammatory and cellular death responses. Furthermore, we have successfully shown GardeninA bioavailability in the fly heads after oral administration using ultra-performance liquid chromatography and mass spectrometry. Our findings reveal a molecular mechanistic insight into GardeninA-mediated neuroprotection against environmental toxin-induced Parkinson’s disease pathogenesis for novel therapeutic intervention. Maitra and colleagues identify the neuroprotective properties of GardeninA against environmental toxin-induced neurodegeneration in Drosophila. This study has the potential to influence future research into toxin-induced Parkinson’s disease pathogenesis.
Collapse
Affiliation(s)
- Urmila Maitra
- Department of Biological Sciences, University of Alabama, 2320 Science and Engineering Complex, Tuscaloosa, AL, 35487-0344, USA.
| | - Thomas Harding
- Department of Biological Sciences, University of Alabama, 2320 Science and Engineering Complex, Tuscaloosa, AL, 35487-0344, USA
| | - Qiaoli Liang
- Mass Spectrometry Facility, Department of Chemistry and Biochemistry, University of Alabama, 2004 Shelby Hall, Tuscaloosa, AL, 35487-0336, USA
| | - Lukasz Ciesla
- Department of Biological Sciences, University of Alabama, 2320 Science and Engineering Complex, Tuscaloosa, AL, 35487-0344, USA.
| |
Collapse
|
21
|
Malankhanova T, Suldina L, Grigor’eva E, Medvedev S, Minina J, Morozova K, Kiseleva E, Zakian S, Malakhova A. A Human Induced Pluripotent Stem Cell-Derived Isogenic Model of Huntington's Disease Based on Neuronal Cells Has Several Relevant Phenotypic Abnormalities. J Pers Med 2020; 10:jpm10040215. [PMID: 33182269 PMCID: PMC7712151 DOI: 10.3390/jpm10040215] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/30/2022] Open
Abstract
Huntington's disease (HD) is a severe neurodegenerative disorder caused by a CAG triplet expansion in the first exon of the HTT gene. Here we report the introduction of an HD mutation into the genome of healthy human embryonic fibroblasts through CRISPR/Cas9-mediated homologous recombination. We verified the specificity of the created HTT-editing system and confirmed the absence of undesirable genomic modifications at off-target sites. We showed that both mutant and control isogenic induced pluripotent stem cells (iPSCs) derived by reprogramming of the fibroblast clones can be differentiated into striatal medium spiny neurons. We next demonstrated phenotypic abnormalities in the mutant iPSC-derived neural cells, including impaired neural rosette formation and increased sensitivity to growth factor withdrawal. Moreover, using electron microscopic analysis, we detected a series of ultrastructural defects in the mutant neurons, which did not contain huntingtin aggregates, suggesting that these defects appear early in HD development. Thus, our study describes creation of a new isogenic iPSC-based cell system that models HD and recapitulates HD-specific disturbances in the mutant cells, including some ultrastructural features implemented for the first time.
Collapse
|
22
|
Mahyudin F, Yazid H, Edward M, Basuki MH, Bari YA, Rantam FA. The enhancement apoptosis of osteosarcoma mesenchymal stem cells co-cultivation with peripheral blood mononuclear cells sensitized by secretome and granulocyte macrophage colony-stimulating factor. J Adv Pharm Technol Res 2020; 11:213-219. [PMID: 33425707 PMCID: PMC7784941 DOI: 10.4103/japtr.japtr_52_20] [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: 04/25/2020] [Revised: 06/06/2020] [Accepted: 08/25/2020] [Indexed: 12/22/2022] Open
Abstract
The advanced, metastasis, and reccurent of osteosarcoma (OS) patients have a poor prognosis postaggresive surgery and chemotherapy. Peripheral blood mononuclear cells (PBMCs) as cell-based immunotherapy may successful in the OS treatment. To investigate the enhancement apoptosis of OS-mesenchymal stem cells (OS-MSCs) co-cultivated with PBMCs sensitized using the secretome and granulocyte macrophage colony-stimulating factor (GMCSF). This true experimental study with posttest only control group design and in vitro study. The sample was cultured OS-MSCs which confirmed by Cluster of Differentiation-133 using immunocytochemistry (ICC) and histopathology analysis. The sample divided into six groups accordingly: OS-MSC, OS-MSC + PMBC, OS-MSC + PMBC + Secretome, OS-MSC + PMBC + GMCSF, OS-MSC + PBMC + Secretome + GMCSF (n = 5/N = 30). The enhancement of OS-MSCs apoptosis was analyzed through Interleukin-2 (IL-2) level through the Enyzme-Linked Immunosorbent Assay examination, expression of Signal Transducers and Activators of Transcription (STAT)-3 and caspase-3 by ICC. One-way analysis of variance test and Tukey Honestly Significant Difference to analyze the difference between the groups (P < 0.05). The highest of IL-2 level was found in the PBMC + Secretome + GMCSF group. The highest expression of caspase-3 was found in OS-MSC + PBMC + Secretome + GMCSF group with significant different between groups (P < 0.05). There was insignificant difference of STAT-3 epxression and IL-2 level between groups (P > 0.05). The co-cultivation of OS-MSCs and PBMSCs activated using secretome and GMCSF has a great ability to enhance OS-MSCs apoptosis.
Collapse
Affiliation(s)
- Ferdiansyah Mahyudin
- Orthopedic and Traumatology Department, Faculty of Medicine, Dr Soetomo General Hospital, Airlangga University, Surabaya, Indonesia
| | - Hizbillah Yazid
- Orthopedic and Traumatology Department, Faculty of Medicine, Dr Soetomo General Hospital, Airlangga University, Surabaya, Indonesia
| | - Mouli Edward
- Orthopedic and Traumatology Department, Faculty of Medicine, Dr Soetomo General Hospital, Airlangga University, Surabaya, Indonesia
| | - Mohammad Hardian Basuki
- Orthopedic and Traumatology Department, Faculty of Medicine, Dr Soetomo General Hospital, Airlangga University, Surabaya, Indonesia
| | - Yunus Abdul Bari
- Orthopedic and Traumatology Department, Faculty of Medicine, Dr Soetomo General Hospital, Airlangga University, Surabaya, Indonesia
| | - Fedik Abdul Rantam
- Stem Cell Research And Development Center, Airlangga University, Surabaya, Indonesia
| |
Collapse
|
23
|
Wang T, Wu X, Al Rudaisat M, Song Y, Cheng H. Curcumin induces G2/M arrest and triggers autophagy, ROS generation and cell senescence in cervical cancer cells. J Cancer 2020; 11:6704-6715. [PMID: 33046993 PMCID: PMC7545669 DOI: 10.7150/jca.45176] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022] Open
Abstract
Our study explored the tumor-suppressive effect of curcumin on cervical cancer cells. Cervical cancer is one of the most common cancers among women worldwide. Acquired resistance to chemotherapeutics and toxicity of such drugs has undermined the effectiveness of cervical cancer treatments. Therefore, the identification of novel chemotherapeutics is key to improving the survival of patients with cervical cancer. Curcumin has been shown to have various bioactivities, including antioxidant and antitumor effects; however, its effect on cervical cancer remains elusive. Here, we used the SiHa human cervical cancer cell line to test various concentrations of curcumin on the proliferation and apoptosis of cervical cancer cells. The involvement of autophagy and reactive oxygen species (ROS) in these effects were also tested by using specific autophagy inhibitors and ROS scavengers. Our results showed that curcumin induced ROS accumulation, apoptosis, autophagy, cell cycle arrest, and cellular senescence accompanied by upregulation of p53 and p21 proteins in SiHa cells.
Collapse
Affiliation(s)
- Tuan Wang
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 3 Qingchun Road, Zhejiang, 310016, P.R. China
| | - Xia Wu
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 3 Qingchun Road, Zhejiang, 310016, P.R. China
| | - Mus'ab Al Rudaisat
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 3 Qingchun Road, Zhejiang, 310016, P.R. China
| | - Yinjing Song
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 3 Qingchun Road, Zhejiang, 310016, P.R. China
| | - Hao Cheng
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 3 Qingchun Road, Zhejiang, 310016, P.R. China
| |
Collapse
|
24
|
Cankara FN, Günaydın C, Bilge SS, Özmen Ö, Kortholt A. The neuroprotective action of lenalidomide on rotenone model of Parkinson's Disease: Neurotrophic and supportive actions in the substantia nigra pars compacta. Neurosci Lett 2020; 738:135308. [PMID: 32932183 DOI: 10.1016/j.neulet.2020.135308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/06/2020] [Accepted: 08/12/2020] [Indexed: 01/13/2023]
Abstract
Lenalidomide is a centrally active thalidomide analog that has potent anti-inflammatory and antiangiogenic activities. Currently, it is primarily used in the treatment of multiple myeloma and myelodysplastic syndromes. However, recent studies have revealed in addition to neuroprotection and neuromodulation of lenalidomide. Because of this combination of inflammation and neuro-immunogenic properties, lenalidomide is considered as a high potential compound for the treatment of neurodegenerative diseases. Despite intensive research during the last decade, the role of neurotrophic elements in the effect of lenalidomide is still not well understood. Therefore, in the current study, the effects of lenalidomide on neurodegeneration were investigated in a rotenone model of Parkinson's disease (PD) rat model. The PD rat model was generated by rotenone injection into the substantia nigra pars compacta (SNpc). After validation of the PD model, the rats were treated with lenalidomide (100 mg/kg) for 28 days. Our data shows that lenalidomide alleviated rotenone-induced motor impairments and deficits in dopamine-related behaviors and resulted in increased levels of tumor necrosis factor-α and calcium-binding protein B in the SNpc. Moreover, chronic lenalidomide treatment resulted increase in transforming growth factor immunoreactivity and brain derived neurotrophic factor expression in the SNPc. In addition, chronic treatment mitigated tyrosine hydroxylase expression prevented the rotenone-induced decrease in dopamine levels, and consequently a decrease in caspase-3/9 immunoreactivity. This thus shows that chronic lenalidomide treatment improves neuronal survival. Together with our data demonstrate that lenalidomide, in addition to its anti-inflammatory and immunomodulatory actions, is also capable of increasing neurotrophic factors in the SNpc, thereby preventing rotenone-induced motor impairments.
Collapse
Affiliation(s)
- Fatma Nihan Cankara
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey; Innovative Technologies Application and Research Center, Suleyman Demirel University, Isparta, Turkey.
| | - Caner Günaydın
- Department of Pharmacology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey.
| | - Süleyman Sırrı Bilge
- Department of Pharmacology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey.
| | - Özlem Özmen
- Department of Pathology, Faculty of Veterinary Medicine, Mehmet Akif Ersoy University, Burdur, Turkey.
| | - Arjan Kortholt
- Department of Cell Biochemistry, Groningen Institute of Biomolecular Sciences & Biotechnology, University of Groningen, Groningen, The Netherlands; Innovative Technologies Application and Research Center, Suleyman Demirel University, Isparta, Turkey.
| |
Collapse
|
25
|
Abbaszadeh F, Fakhri S, Khan H. Targeting apoptosis and autophagy following spinal cord injury: Therapeutic approaches to polyphenols and candidate phytochemicals. Pharmacol Res 2020; 160:105069. [PMID: 32652198 DOI: 10.1016/j.phrs.2020.105069] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/28/2020] [Accepted: 07/03/2020] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) is a neurological disorder associated with the loss of sensory and motor function. Understanding the precise dysregulated signaling pathways, especially apoptosis and autophagy following SCI, is of vital importance in developing innovative therapeutic targets and treatments. The present study lies in the fact that it reveals the precise dysregulated signaling mediators of apoptotic and autophagic pathways following SCI and also examines the effects of polyphenols and other candidate phytochemicals. It provides new insights to develop new treatments for post-SCI complications. Accordingly, a comprehensive review was conducted using electronic databases including, Scopus, Web of Science, PubMed, and Medline, along with the authors' expertise in apoptosis and autophagy as well as their knowledge about the effects of polyphenols and other phytochemicals on SCI pathogenesis. The primary mechanical injury to spinal cord is followed by a secondary cascade of apoptosis and autophagy that play critical roles during SCI. In terms of pharmacological mechanisms, caspases, Bax/Bcl-2, TNF-α, and JAK/STAT in apoptosis along with LC3 and Beclin-1 in autophagy have shown a close interconnection with the inflammatory pathways mainly glutamatergic, PI3K/Akt/mTOR, ERK/MAPK, and other cross-linked mediators. Besides, apoptotic pathways have been shown to regulate autophagy mediators and vice versa. Prevailing evidence has highlighted the importance of modulating these signaling mediators/pathways by polyphenols and other candidate phytochemicals post-SCI. The present review provides dysregulated signaling mediators and therapeutic targets of apoptotic and autophagic pathways following SCI, focusing on the modulatory effects of polyphenols and other potential phytochemical candidates.
Collapse
Affiliation(s)
- Fatemeh Abbaszadeh
- Department of Neuroscience, Faculty of Advanced Technologies in Medical Sciences, Iran University of Medical Sciences, Tehran, Iran; Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| |
Collapse
|
26
|
Akomolafe SF, Olasehinde TA, Oyeleye SI, Aluko TB, Adewale OO, Ijomone OM. Curcumin Administration Mitigates Cyclophosphamide-Induced Oxidative Damage and Restores Alteration of Enzymes Associated with Cognitive Function in Rats' Brain. Neurotox Res 2020; 38:199-210. [PMID: 32405958 DOI: 10.1007/s12640-020-00205-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 03/19/2020] [Accepted: 04/08/2020] [Indexed: 12/18/2022]
Abstract
The use of chemotherapeutic drugs is associated with oxidative damage, cognitive dysfunction, and brain damage. This study sought to investigate the neuroprotective effect of curcumin against cognitive problems associated with treatment with cyclophosphamide via assessment of biomolecules associated with cognitive function in rats' brain homogenates. Rats were divided in to five groups: Control (vehicle), CUR (curcumin [20 mg/kg]), CPA (cyclophosphamide [150 mg/kg]), CUR1 + CPA (curcumin [20 mg/kg] and cyclophosphamide [150 mg/kg]), and CPA + CUR2 (cyclophosphamide [150 mg/kg] and curcumin [20 mg/kg]). After the treatment, cognitive behavior was assessed and enzymes [cholinesterases, purinergic enzymes, arginase, and angiotensin I-converting enzyme] associated with cognitive function were examined. Oxidative stress parameters [total thiol, non-protein thiol, malondialdehyde, and nitric oxide] including the expression of caspase-3 were also assessed in rats' brain. Our results showed that curcumin improved cognitive behavior, attenuated cholinergic deficit as revealed by the inhibition of cholinesterases, and improved purinergic signaling in cyclophosphamide-treated rats. Furthermore, curcumin reduced angiotensin-I-converting enzyme and arginase activities before and after treatment with cyclophosphamide. Curcumin also improved redox balance and showed protection against cyclophosphamide-induced oxidative damage to rats' brain via an increase in protein and non-protein thiols and nitric oxide levels as well as a significant reduction in malondialdehyde levels. Curcumin also prevented neuronal degeneration in different brain regions and reduced caspase-3 expression. Hence this study suggests that pre and post-treatment with curcumin improved neurobehavior, modulates some biomarkers associated with cognitive function and exhibit neuroprotection against cyclophosphamide-induced neurotoxicity in rats.
Collapse
Affiliation(s)
| | - Tosin A Olasehinde
- Nutrition and Toxicology Division, Food Technology Department, Federal Institute of Industrial Research Oshodi, Lagos, Nigeria. .,Department of Biochemistry and Microbiology, University of Fort Hare, Alice, Eastern Cape, South Africa.
| | - Sunday Idowu Oyeleye
- Department of Biomedical Technology, Federal University of Technology Akure, Akure, Ondo State, Nigeria
| | - Tola B Aluko
- Department of Biochemistry, Faculty of Basic and Applied Sciences, Osun State University, Osogbo, Nigeria
| | - Omowumi O Adewale
- Department of Biochemistry, Faculty of Basic and Applied Sciences, Osun State University, Osogbo, Nigeria
| | - Omamuyovwi M Ijomone
- The Neuro-Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Akure, Ondo State, Nigeria
| |
Collapse
|
27
|
Eskandari H, Ehsanpour AA, Al-Mansour N, Bardania H, Sutherland D, Mohammad-Beigi H. Rosmarinic acid inhibits programmed cell death in Solanum tuberosum L. calli under high salinity. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2020; 147:54-65. [PMID: 31841962 DOI: 10.1016/j.plaphy.2019.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/10/2019] [Accepted: 12/02/2019] [Indexed: 06/10/2023]
Abstract
Oxidative stress induced by salinity is a prime cause of cell death when Na+ toxicity becomes unbearable. We explored the effect of rosmarinic acid (RA) on the Solanum tuberosum L. cv. Desiree calli against salt-induced programmed cell death (PCD). We showed that PCD events were triggered in calli under 250 mM NaCl by the loss of plasma membrane integrity as measured by the amount of malondialdehyde (MDA) in the cytoplasm, the degree of DNA degradation resulting from the cleavage of nuclear DNA into oligonucleosomal fragments in apoptotic cells, the presence of TUNEL-positive nuclei (90 ± 0.005%) damage in genomic DNA, and activation of caspase 3-like protease. Callus Formation Medium (CFM) supplemented with RA led to the suppression of salt-induced cell death and a dramatic decrease in the MDA level and frequency of TUNEL-positive nuclei under salinity to 4 ± and 7.3 ± % in the presence of 50 and 350 μM RA, respectively. The application of RA also resulted in an increase in GSH content and maintenance of a high GSH/GSSG ratio. Interestingly, these reductions in PCD were accompanied by inhibiting caspase 3-like protease activities due to RA under salinity. Molecular docking predicted high binding energies of RA for binding to subtilisin-like protease (StSCTc-3), which has caspase-3 like activity in Solanum tuberosum, near the active site. This finding supports the notion of a role for RA in PCD protection in plants, which is consistent with earlier reports in animal cells.
Collapse
Affiliation(s)
- Hoda Eskandari
- Department of Biology, University of Isfahan, Isfahan, Iran; Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | | | - Naemah Al-Mansour
- Faculty of Science, Department of Biological Sciences, University of Kuwait, Kuwait.
| | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Duncan Sutherland
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | - Hossein Mohammad-Beigi
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| |
Collapse
|
28
|
Tappe D, Schmidt-Chanasit J, Rauch J, Allartz P, Herden C. Immunopathology of Fatal Human Variegated Squirrel Bornavirus 1 Encephalitis, Germany, 2011-2013. Emerg Infect Dis 2019; 25:1058-1065. [PMID: 31107210 PMCID: PMC6537742 DOI: 10.3201/eid2506.181082] [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] [Indexed: 12/20/2022] Open
Abstract
Variegated squirrel bornavirus 1 (VSBV-1) is a zoonotic virus that causes fatal encephalitis in humans who are infected after contact with exotic squirrels. We analyzed the brain lesions and the immune responses in all 4 known human cases that showed panencephalitis. Inflammatory infiltrates in areas positive for VSBV-1 RNA and antigen consisted of CD4+ and CD8+ T cells, with perivascular B-cell accumulation. Strong microglial response and bizarre astroglial expansion were present. Areas of malacia contained neutrophils and foamy microglia and macrophages. Immunopathologic examination during infection showed cleavage of caspase 3 in brain cells adjacent to CD8+ cells and widespread p53 expression, hallmarks of apoptosis. Cerebrospinal fluid analyses over time demonstrated increasing protein concentrations and cell counts, paralleled by pathologic lactate elevations in all patients. The most severe cerebrospinal fluid and histologic changes occurred in the patient with the highest viral load, shortest duration of disease, and most medical preconditions.
Collapse
|
29
|
Kumi RO, Issahaku AR, Soremekun OS, Agoni C, Olotu FA, Soliman MES. From the Explored to the Unexplored: Computer-Tailored Drug Design Attempts in the Discovery of Selective Caspase Inhibitors. Comb Chem High Throughput Screen 2019; 22:432-444. [PMID: 31560284 DOI: 10.2174/1386207322666190927143026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/19/2019] [Accepted: 08/01/2019] [Indexed: 01/09/2023]
Abstract
The pathophysiological roles of caspases have made them attractive targets in the treatment and amelioration of neurologic diseases. In normal conditions, the expression of caspases is regulated in the brain, while at the onset of neurodegeneration, such as in Alzheimer's disease, they are typically overexpressed. Till date, several therapeutic efforts that include the use of small endogenous binders have been put forward to curtail dysfunctionalities that drive aberrant death in neuronal cells. Caspases are highly homologous, both in structure and in sequence, which leaves us with the question: is it possible to specifically and individually target caspases, while multiple therapeutic attempts to achieve selective targeting have failed! Based on antecedent events, the use of Computer-Aided Drug Design (CADD) methods has significantly contributed to the design of small molecule inhibitors, especially with selective target ability and reduced off-target therapeutic effects. Interestingly, we found out that there still exists an enormous room for the integration of structure/ligand-based drug design techniques towards the development of highly specific reversible and irreversible caspase inhibitors. Therefore, in this review, we highlight drug discovery approaches that have been directed towards caspase inhibition in addition to an insightful focus on applicable CADD techniques for achieving selective targeting in caspase research.
Collapse
Affiliation(s)
- Ransford O Kumi
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Abdul R Issahaku
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Opeyemi S Soremekun
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Clement Agoni
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Fisayo A Olotu
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| |
Collapse
|
30
|
Wu X, Lv YG, Du YF, Hu M, Reed MN, Long Y, Suppiramaniam V, Hong H, Tang SS. Inhibitory effect of INT-777 on lipopolysaccharide-induced cognitive impairment, neuroinflammation, apoptosis, and synaptic dysfunction in mice. Prog Neuropsychopharmacol Biol Psychiatry 2019; 88:360-374. [PMID: 30144494 DOI: 10.1016/j.pnpbp.2018.08.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/02/2018] [Accepted: 08/19/2018] [Indexed: 01/09/2023]
Abstract
Neuroinflammation plays an important role in the pathophysiology of Alzheimer's disease (AD) and memory impairment. Herein, we evaluated the neuroprotective effects of 6-ethyl-23(S)-methyl-cholic acid (INT-777), a specific G-protein coupled bile acid receptor 1 (TGR5) agonist, in the LPS-treated mouse model of acute neurotoxicity. Single intracerebroventricular (i.c.v.) injection of LPS remarkably induced mouse behavioral impairments in Morris water maze, novel object recognition, and Y-maze avoidance tests, which were ameliorated by INT-777 (1.5 or 3.0 μg/mouse, i.c.v.) treatment. Importantly, INT-777 treatment reversed LPS-induced TGR5 down-regulation, suppressed the increase of nuclear NF-κB p65, and mitigated neuroinflammation, evidenced by lower proinflammatory cytokines, less activation of microglia, and increased the ratio of p-CREB/CREB or mBDNF/proBDNF in the hippocampus and frontal cortex. In addition, INT-777 treatment also suppressed neuronal apoptosis, as indicated by the reduction of TUNEL-positive cells, decreased activation of caspase-3, increased the ratio of Bcl-2/Bax, and ameliorated synaptic dysfunction as evidenced by the upregulation of PSD95 and synaptophysin in the hippocampus and frontal cortex. Taken together, this study showed the potential neuroprotective effects of INT-777 against LPS-induced cognitive impairment, neuroinflammation, apoptosis, and synaptic dysfunction in mice.
Collapse
Affiliation(s)
- Xian Wu
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Yang-Ge Lv
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Yi-Feng Du
- Department of Drug Discovery and Development, School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Mei Hu
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Miranda N Reed
- Department of Drug Discovery and Development, School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Yan Long
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Hao Hong
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China.
| | - Su-Su Tang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
31
|
Bell K, Und Hohenstein-Blaul NVT, Teister J, Grus F. Modulation of the Immune System for the Treatment of Glaucoma. Curr Neuropharmacol 2018; 16:942-958. [PMID: 28730968 PMCID: PMC6120111 DOI: 10.2174/1570159x15666170720094529] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/17/2017] [Accepted: 07/18/2017] [Indexed: 12/20/2022] Open
Abstract
Background: At present intraocular pressure (IOP) lowering therapies are the only approach to treat glaucoma. Neuroprotective strategies to protect the retinal ganglion cells (RGC) from apoptosis are lacking to date. Substantial amount of research concerning the role of the immune system in glaucoma has been performed in the recent years. This review aims to analyse changes found in the peripheral immune system, as well as selected local changes of retina immune cells in the glaucomatous retina. Methods: By dividing the immune system into the innate and the adaptive immune system, a systematic literature research was performed to find recent approaches concerning the modulation of the immune system in the context of glaucoma. Also ClinicalTrials.gov was assessed to identify studies with a translational context. Results: We found that some aspects of the immune system, such as changes in antibody levels, changes in toll like receptor signalling, T cells and retinal microglial cells, experience more research activity than other areas such as changes in dendritic cells or macrophages. Briefly, results from clinical studies revealed altered immunoreactivities against retinal and optic nerve antigens in sera and aqueous humor of glaucoma patients and point toward an autoimmune involvement in glaucomatous neurodegeneration and RGC death. IgG accumulations along with plasma cells were found localised in human glaucomatous retinae in a pro-inflammatory environment possibly maintained by microglia. Animal studies show that antibodies (e.g. anti- heat shock protein 60 and anti-myelin basic protein) elevated in glaucoma patients provoke autoaggressive RGC loss and are associated with IgG depositions and increased microglial cells. Also, studies addressing changes in T lymphocytes, macrophages but also local immune responses in the retina have been performed and also hold promising results. Conclusions: This recapitulation of recent literature demonstrates that the immune system definitely plays a role in the pathogenesis of glaucoma. Multiple changes in the peripheral innate as well as adaptive immune system have been detected and give room for further research concerning valuable therapeutic targets. We conclude that there still is a great need to bring together the results derived from basic research analysing different aspects of the immune system in glaucoma to understand the immune context of the disease. Furthermore local immune changes in the retina of glaucoma patients still leave room for further therapeutic targets
Collapse
Affiliation(s)
- Katharina Bell
- Experimental and Translational Ophthalmology Mainz, Department of Ophthalmology, Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstrasse 1, 55101 Mainz, Germany
| | - Nadine von Thun Und Hohenstein-Blaul
- Experimental and Translational Ophthalmology Mainz, Department of Ophthalmology, Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstrasse 1, 55101 Mainz, Germany
| | - Julia Teister
- Experimental and Translational Ophthalmology Mainz, Department of Ophthalmology, Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstrasse 1, 55101 Mainz, Germany
| | - Franz Grus
- Experimental and Translational Ophthalmology Mainz, Department of Ophthalmology, Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstrasse 1, 55101 Mainz, Germany
| |
Collapse
|
32
|
Izadpanah M, Seddigh A, Ebrahimi Barough S, Fazeli SAS, Ai J. Potential of Extracellular Vesicles in Neurodegenerative Diseases: Diagnostic and Therapeutic Indications. J Mol Neurosci 2018; 66:172-179. [PMID: 30140997 DOI: 10.1007/s12031-018-1135-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 07/20/2018] [Indexed: 01/09/2023]
Abstract
Extracellular vesicles (EVs) are membrane-bound vesicles, including exosomes and microvesicles. EVs are nanometer sized, found in physiological fluids such as urine, blood, cerebro-spinal fluid (CSF), with a capacity of transferring various biological materials such as microRNAs, proteins, and lipids among cells without direct cell-to-cell contact. Many cells in the nervous system have been shown to release EVs. These vesicles are involved in intercellular communication and a variety of biological processes such as modulation of immune response, signal transduction, and transport of genetic materials with low immunogenicity; therefore, they have also been recently investigated for the delivery of therapeutic molecules such as siRNAs and drugs in the treatment of diseases. In addition, since EV components reflect the physiological status of the cells and tissues producing them, they can be utilized as biomarkers for early detection of various diseases. In this review, we summarize EV application, in diagnosis as biomarker sources and as a carrier tool for drug delivery in EV-based therapies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Mehrnaz Izadpanah
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P. O. Box: 1417755469, Tehran, Iran.,Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| | - Arshia Seddigh
- Department of Neurology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Somayeh Ebrahimi Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P. O. Box: 1417755469, Tehran, Iran
| | - Seyed Abolhassan Shahzadeh Fazeli
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran.,Department of Molecular and Cellular Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P. O. Box: 1417755469, Tehran, Iran.
| |
Collapse
|
33
|
Khan S, Bhardwaj T, Somvanshi P, Mandal RK, Dar SA, Jawed A, Wahid M, Akhter N, Lohani M, Alouffi S, Haque S. Inhibition of C298S mutant of human aldose reductase for antidiabetic applications: Evidence from in silico elementary mode analysis of biological network model. J Cell Biochem 2018; 119:6961-6973. [PMID: 29693278 DOI: 10.1002/jcb.26904] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 03/28/2018] [Indexed: 01/05/2023]
Abstract
Human aldose reductase (hAR) is the key enzyme in sorbitol pathway of glucose utilization and is implicated in the etiology of secondary complications of diabetes, such as, cardiovascular complications, neuropathy, nephropathy, retinopathy, and cataract genesis. It reduces glucose to sorbitol in the presence of NADPH and the major cause of diabetes complications could be the change in the osmotic pressure due to the accumulation of sorbitol. An activated form of hAR (activated hAR or ahAR) poses a potential obstacle in the development of diabetes drugs as hAR-inhibitors are ineffective against ahAR. The therapeutic efficacy of such drugs is compromised when a large fraction of the enzyme (hAR) undergoes conversion to the activated ahAR form as has been observed in the diabetic tissues. In the present study, attempts have been made to employ systems biology strategies to identify the elementary nodes of human polyol metabolic pathway, responsible for normal metabolic states, followed by the identification of natural potent inhibitors of the activated form of hAR represented by the mutant C298S for possible antidiabetic applications. Quantum Mechanical Molecular Mechanical docking strategy was used to determine the probable inhibitors of ahAR. Rosmarinic acid was found as the most potent natural ahAR inhibitor and warrants for experimental validation in the near future.
Collapse
Affiliation(s)
- Saif Khan
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, University of Ha'il, Ha'il, Saudi Arabia
| | - Tulika Bhardwaj
- Department of Biotechnology, TERI School of Advanced Studies, New Delhi, India
| | - Pallavi Somvanshi
- Department of Biotechnology, TERI School of Advanced Studies, New Delhi, India
| | - Raju K Mandal
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Sajad A Dar
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Arshad Jawed
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Mohd Wahid
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Naseem Akhter
- Faculty of Applied Medical Sciences, Department of Laboratory Medicine, Albaha University, Albaha, Saudi Arabia
| | - Mohtashim Lohani
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - S Alouffi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, University of Ha'il, Ha'il, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
34
|
Yao Q, Wang W, Jin J, Min K, Yang J, Zhong Y, Xu C, Deng J, Zhou Y. Synergistic role of Caspase-8 and Caspase-3 expressions: Prognostic and predictive biomarkers in colorectal cancer. Cancer Biomark 2018; 21:899-908. [PMID: 29355114 DOI: 10.3233/cbm-170967] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Qiang Yao
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Soochow, Jiangsu, China
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Weimin Wang
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
- Institute of Combining Chinese Traditional and Western Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Jun Jin
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Ke Min
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Jian Yang
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Yubing Zhong
- Department of General Surgery, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Chunni Xu
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Jianliang Deng
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Yan Zhou
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
- Institute of Combining Chinese Traditional and Western Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
35
|
Sehgal SA, Hammad MA, Tahir RA, Akram HN, Ahmad F. Current Therapeutic Molecules and Targets in Neurodegenerative Diseases Based on in silico Drug Design. Curr Neuropharmacol 2018; 16:649-663. [PMID: 29542412 PMCID: PMC6080102 DOI: 10.2174/1570159x16666180315142137] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 01/01/2018] [Accepted: 03/02/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND As the number of elderly persons increases, neurodegenerative diseases are becoming ubiquitous. There is currently a great need for knowledge concerning management of oldage neurodegenerative diseases; the most important of which are: Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis, and Huntington's disease. OBJECTIVE To summarize the potential of computationally predicted molecules and targets against neurodegenerative diseases. METHOD Review of literature published since 1997 against neurodegenerative diseases, utilizing as keywords: in silico, Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis ALS, and Huntington's disease was conducted. RESULTS AND CONCLUSION Due to the costs associated with experimentation and current ethical law, performing experiments directly on living organisms has become much more difficult. In this scenario, in silico techniques have been successful and have become powerful tools in the search to cure disease. Researchers use the Computer Aided Drug Design pipeline which: 1) generates 3- dimensional structures of target proteins through homology modeling 2) achieves stabilization through molecular dynamics simulation, and 3) exploits molecular docking through large compound libraries. Next generation sequencing is continually producing enormous amounts of raw sequence data while neuroimaging is producing a multitude of raw image data. To solve such pressing problems, these new tools and algorithms are required. This review elaborates precise in silico tools and techniques for drug targets, active molecules, and molecular docking studies, together with future prospects and challenges concerning possible breakthroughs in Alzheimer's, Parkinson's, Amyotrophic Lateral Sclerosis, and Huntington's disease.
Collapse
Affiliation(s)
- Sheikh Arslan Sehgal
- Address correspondence to this author at the State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences; Beijing, China; E-mail:
| | | | | | | | | |
Collapse
|
36
|
Zanforlin E, Zagotto G, Ribaudo G. The Medicinal Chemistry of Natural and Semisynthetic Compounds against Parkinson's and Huntington's Diseases. ACS Chem Neurosci 2017; 8:2356-2368. [PMID: 28862431 DOI: 10.1021/acschemneuro.7b00283] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Among the diseases affecting the central nervous system (CNS), neurodegenerations attract the interest of both the clinician and the medicinal chemist. The increasing average age of population, the growing number of patients, and the lack of long-term effective remedies push ahead the quest for novel tools against this class of pathologies. We present a review on the state of the art of the molecules (or combination of molecules) of natural origin that are currently under study against two well-defined pathologies: Parkinson's disease (PD) and Huntington's disease (HD). Nowadays, very few tools are available for preventing or counteracting the progression of such diseases. Two major parameters were considered for the preparation of this review: particular attention was reserved to these research works presenting well-defined molecular mechanisms for the studied compounds, and where available, papers reporting in vivo data were preferred. A literature search for peer-reviewed articles using PubMed, Scopus, and Reaxys databases was performed, exploiting different keywords and logical operators: 91 papers were considered (preferentially published after 2015). The review presents a brief overview on the etiology of the studied neurodegenerations and the current treatments, followed by a detailed discussion of the natural and semisynthetic compounds dividing them in different paragraphs considering their several mechanisms of action.
Collapse
Affiliation(s)
- Enrico Zanforlin
- Department of Pharmaceutical
and Pharmacological Sciences, University of Padova, Padova 35131, Italy
| | - Giuseppe Zagotto
- Department of Pharmaceutical
and Pharmacological Sciences, University of Padova, Padova 35131, Italy
| | - Giovanni Ribaudo
- Department of Pharmaceutical
and Pharmacological Sciences, University of Padova, Padova 35131, Italy
| |
Collapse
|
37
|
Chen SG, Leu YL, Cheng ML, Ting SC, Liu CC, Wang SD, Yang CH, Hung CY, Sakurai H, Chen KH, Ho HY. Anti-enterovirus 71 activities of Melissa officinalis extract and its biologically active constituent rosmarinic acid. Sci Rep 2017; 7:12264. [PMID: 28947773 PMCID: PMC5613005 DOI: 10.1038/s41598-017-12388-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 09/07/2017] [Indexed: 01/22/2023] Open
Abstract
Enterovirus 71 (EV71) infection is endemic in the Asia-Pacific region. No specific antiviral drug has been available to treat EV71 infection. Melissa officinalis (MO) is a medicinal plant with long history of usage in the European and Middle East. We investigated whether an aqueous solution of concentrated methanolic extract (MOM) possesses antiviral activity. MOM inhibited plaque formation, cytopathic effect, and viral protein synthesis in EV71-infected cells. Using spectral techniques, we identified rosmarinic acid (RA) as a biologically active constituent of MOM. RA reduced viral attachment and entry; cleavage of eukaryotic translation initiation factor 4 G (eIF4G); reactive oxygen species (ROS) generation; and translocation of heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) from nucleus to cytoplasm. It alleviated EV71-induced hyperphosphorylation of p38 kinase and EPS15. RA is likely to suppress ROS-mediated p38 kinase activation, and such downstream molecular events as hnRNP A1 translocation and EPS15-regulated membrane trafficking in EV71-infected cells. These findings suggest that MO and its constituent RA possess anti-EV71 activities, and may serve as a candidate drug for therapeutic and prophylactic uses against EV71 infection.
Collapse
Affiliation(s)
- Sin-Guang Chen
- Graduate Institute of Biomedical Science, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital at Linkou, Guishan, Taoyuan, Taiwan
- Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Mei-Ling Cheng
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Guishan, Taoyuan, Taiwan
- Healthy Aging Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan
- Metabolomics Core Laboratory, Chang Gung University, Guishan, Taoyuan, Taiwan
- Clinical Phenome Center, Chang Gung Memorial Hospital at Linkou, Guishan, Taoyuan, Taiwan
| | - Siew Chin Ting
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Chuan Liu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Shulhn-Der Wang
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Cheng-Hung Yang
- Graduate Institute of Biomedical Science, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Cheng-Yu Hung
- Healthy Aging Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan
- Metabolomics Core Laboratory, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Hiroaki Sakurai
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Kuan-Hsing Chen
- Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Hung-Yao Ho
- Healthy Aging Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan.
- Clinical Phenome Center, Chang Gung Memorial Hospital at Linkou, Guishan, Taoyuan, Taiwan.
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
38
|
Liang Y, Huang M, Jiang X, Liu Q, Chang X, Guo Y. The neuroprotective effects of Berberine against amyloid β-protein-induced apoptosis in primary cultured hippocampal neurons via mitochondria-related caspase pathway. Neurosci Lett 2017; 655:46-53. [DOI: 10.1016/j.neulet.2017.06.048] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/24/2017] [Accepted: 06/26/2017] [Indexed: 11/24/2022]
|
39
|
Carvacrol promotes neuroprotection in the mouse hemiparkinsonian model. Neuroscience 2017; 356:176-181. [DOI: 10.1016/j.neuroscience.2017.05.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 12/12/2022]
|
40
|
Chen F, Ghosh A, Wu F, Tang S, Hu M, Sun H, Kong L, Hong H. Preventive effect of genetic knockdown and pharmacological blockade of CysLT 1R on lipopolysaccharide (LPS)-induced memory deficit and neurotoxicity in vivo. Brain Behav Immun 2017; 60:255-269. [PMID: 27810377 DOI: 10.1016/j.bbi.2016.10.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/17/2016] [Accepted: 10/30/2016] [Indexed: 12/19/2022] Open
Abstract
Previously we reported that cysteinyl leukotrienes (Cys-LTs) and the type 1 receptor for Cys-LTs (CysLT1R) are related to amyloid β (Aβ)-induced neurotoxicity. The aim of the current study was to find out the role of CysLT1R on lipopolysaccharide (LPS)-induced cognitive deficit and neurotoxicity. shRNA-mediated knockdown or pharmacological blockade (by pranlukast) of CysLT1R were performed in ICR mice for 21days prior to systemic infusion of LPS. From day 22, LPS was administered for 7days and then a set of behavioral, histopathological and biochemical tests were employed to test memory, neuroinflammation and apoptotic responses in the mouse hippocampus. LPS (only)-treated mice showed poor performance in both Morris water maze (MWM) and Y-maze tests. However, shRNA-mediated knockdown or pranlukast-treated blockade of CysLT1R improved performance of the mice in these tests. To find out the possible underlying mechanisms, we assessed several parameters such as microglial activation (by immunohistochemistry), level of CysLT1R (by WB and qRT-PCR) and the inflammatory/apoptotic pathways (by ELISA or TUNEL or WB) in the mouse hippocampus. LPS-induced memory impairment was accompanied by activation of microglia, higher level of CysLT1R, IL-1β, TNF-α and nuclear NF-κB p65. LPS also caused apoptosis in the hippocampus as detected by TUNEL staining, further supplemented by detection of increased Caspase-3 and a reduced Bcl-2/Bax ratio. All of these adverse changes in the mouse hippocampus were inhibited by pretreatment with CysLT1R-shRNA and pranlukast. Through this study we suggest that CysLT1R shares a strong correlation with LPS-associated memory deficit, neuroinflammation and apoptosis and CysLT1R could be a novel target for preventive measures to intervene the progression of Alzheimer's disease (AD)-like phenotypes.
Collapse
Affiliation(s)
- Fang Chen
- Department of Pharmacology, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, and State Key Laboratory for Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Arijit Ghosh
- Department of Pharmacology, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, and State Key Laboratory for Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Feng Wu
- School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Susu Tang
- Department of Pharmacology, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, and State Key Laboratory for Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Mei Hu
- Department of Pharmacology, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, and State Key Laboratory for Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Hongbin Sun
- Department of Pharmacology, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, and State Key Laboratory for Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Lingyi Kong
- Department of Pharmacology, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, and State Key Laboratory for Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Hao Hong
- Department of Pharmacology, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, and State Key Laboratory for Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu, China.
| |
Collapse
|
41
|
Soodi M, Dashti A, Hajimehdipoor H, Akbari S, Ataei N. Melissa officinalis Acidic Fraction Protects Cultured Cerebellar Granule Neurons Against Beta Amyloid-Induced Apoptosis and Oxidative Stress. CELL JOURNAL 2016; 18:556-564. [PMID: 28042540 PMCID: PMC5086334 DOI: 10.22074/cellj.2016.4722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 04/25/2016] [Indexed: 12/18/2022]
Abstract
Objective Extracellular deposition of the beta-amyloid (Aβ) peptide, which is the main finding in the pathophysiology of Alzheimer’s disease (AD), leads to oxidative damage and apoptosis in neurons. Melissa officinalis (M. officinalis) is a medicinal plant from the Lamiaceae
family that has neuroprotective activity. In the present study we have investigated the protective effect of the acidic fraction of M. officinalis on Aβ-induced oxidative stress and apoptosis
in cultured cerebellar granule neurons (CGN). Additionally, we investigated a possible role of
the nicotinic receptor.
Materials and Methods This study was an in vitro experimental study performed on
mice cultured CGNs. CGNs were pre-incubated with different concentrations of the acidic
fraction of M. officinalis for 24 hours, followed by incubation with Aβ for an additional 48
hours. CGNs were also pre-incubated with the acidic fraction of M. officinalis and mecamylamin, followed by incubation with Aβ. We used the 3-(4,5-dimethylthiazol-2-yl)-2,5-
diphenyltetrazolium bromide (MTT) assay to measure cell viability. Acetylcholinesterase
(AChE) activity, reactive oxygen species (ROS) production, lipidperoxidation, and caspase-3 activity were measured after incubation. Hochst/annexin Vfluorescein isothiocyanate (FITC)/propidium iodide (PI) staining was performed to detect apoptotic cells.
Results The acidic fraction could protect CGNs from Aβ-induced cytotoxicity. Mecamylamine did not abolish the protective effect of the acidic fraction. AChE activity, ROS
production, lipid peroxidation, and caspase-3 activity increased after Aβ incubation. Preincubation with the acidic fraction of M. officinalis ameliorated these factors and decreased
the number of apoptotic cells.
Conclusion Our results indicated that the protective effect of the acidic fraction of M.
officinalis was not mediated through nicotinic receptors. This fraction could protect CGNs
through antioxidant and anti-apoptotic activities.
Collapse
Affiliation(s)
- Maliheh Soodi
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abolfazl Dashti
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Homa Hajimehdipoor
- Department of Traditional Pharmacy, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shole Akbari
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nasim Ataei
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
42
|
RNAseq Analyses Identify Tumor Necrosis Factor-Mediated Inflammation as a Major Abnormality in ALS Spinal Cord. PLoS One 2016; 11:e0160520. [PMID: 27487029 PMCID: PMC4972368 DOI: 10.1371/journal.pone.0160520] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/20/2016] [Indexed: 12/11/2022] Open
Abstract
ALS is a rapidly progressive, devastating neurodegenerative illness of adults that produces disabling weakness and spasticity arising from death of lower and upper motor neurons. No meaningful therapies exist to slow ALS progression, and molecular insights into pathogenesis and progression are sorely needed. In that context, we used high-depth, next generation RNA sequencing (RNAseq, Illumina) to define gene network abnormalities in RNA samples depleted of rRNA and isolated from cervical spinal cord sections of 7 ALS and 8 CTL samples. We aligned >50 million 2X150 bp paired-end sequences/sample to the hg19 human genome and applied three different algorithms (Cuffdiff2, DEseq2, EdgeR) for identification of differentially expressed genes (DEG's). Ingenuity Pathways Analysis (IPA) and Weighted Gene Co-expression Network Analysis (WGCNA) identified inflammatory processes as significantly elevated in our ALS samples, with tumor necrosis factor (TNF) found to be a major pathway regulator (IPA) and TNFα-induced protein 2 (TNFAIP2) as a major network "hub" gene (WGCNA). Using the oPOSSUM algorithm, we analyzed transcription factors (TF) controlling expression of the nine DEG/hub genes in the ALS samples and identified TF's involved in inflammation (NFkB, REL, NFkB1) and macrophage function (NR1H2::RXRA heterodimer). Transient expression in human iPSC-derived motor neurons of TNFAIP2 (also a DEG identified by all three algorithms) reduced cell viability and induced caspase 3/7 activation. Using high-density RNAseq, multiple algorithms for DEG identification, and an unsupervised gene co-expression network approach, we identified significant elevation of inflammatory processes in ALS spinal cord with TNF as a major regulatory molecule. Overexpression of the DEG TNFAIP2 in human motor neurons, the population most vulnerable to die in ALS, increased cell death and caspase 3/7 activation. We propose that therapies targeted to reduce inflammatory TNFα signaling may be helpful in ALS patients.
Collapse
|
43
|
Genetic Variants of Microtubule Actin Cross-linking Factor 1 (MACF1) Confer Risk for Parkinson’s Disease. Mol Neurobiol 2016; 54:2878-2888. [DOI: 10.1007/s12035-016-9861-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/17/2016] [Indexed: 01/12/2023]
|