1
|
Ghanbari A, Danaie Fard P, Azarmehr N, Mahmoudi R, Roozbehi A, Zibara K, Hosseini E. D-galactose Induces Senescence in Adult Mouse Neural Stem Cells by Imbalanced Oxidant and Antioxidant Activity and Differential Expression of Specific Hub Genes. Mol Neurobiol 2024:10.1007/s12035-024-04546-1. [PMID: 39425831 DOI: 10.1007/s12035-024-04546-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Cellular senescence is a permanent state of cell cycle arrest that occurs in proliferating cells under various stresses causing age-related disorders. This study investigated the role of D-galactose in inducing premature senescence of neural stem cells (NSCs) and the genes involved in this process. After NSC isolation and proliferation, senescence was induced with 10, 20, or 30 µM concentrations of D-galactose for 24 h. Cell viability was tested using the MTT assay, and senescent cells were identified based on increased lysosomal β-galactosidase activity. In addition, levels of NO and malondialdehyde (MDA) oxidative biomarkers but also total thiols (T-SH) and total antioxidant FRAP, as well as inflammatory cytokines, were investigated. Besides, RNA-Seq was performed on the various groups, the gene network was mapped, and genes with the most significant changes were examined. Treatment of NSCs with 20 or 30 µM concentrations of D-galactose caused a significant decrease in cell survival, a number of neurospheres, and a number of neurosphere-derived cells, compared to the control group. In addition, the number of BrdU + cells significantly decreased after induction of NSC senescence with 10 or 20 μM D-galactose, whereas aging-related β-galactosidase (SA-β-gal) increased significantly. Moreover, treatment with 10 or 20 μM D-galactose showed a significant increase in NO production, but not malondialdehyde (MDA). However, the levels of total thiol (T-SH) and antioxidant FRAP levels decreased significantly. Furthermore, TNF-α and IL-6 cytokines significantly increased in NSCs treated with 20 μM, but not 10 μM, D-galactose. Finally, a gene network consisting of 860 gene orthologs was mapped using RNA-Seq. Protein interactions were obtained in 11 hub genes which were classified using gene ontology based on molecular function, biological processes, or cellular processes. Genes with the most significant changes in aging included Fn1, Itga2, Itga3, Itga6, Itga8, Ptk2b, Grin2b, Cacna2d3, Pde4d, Shisa6, and Stac3. This study showed that D-galactose reduces NSC proliferation and antioxidant activity while increasing oxidative stress and inflammatory cytokines. A survey of the genes involved in premature aging may be used as therapeutic candidates for aging-related disorders.
Collapse
Affiliation(s)
- Amir Ghanbari
- Cellular and Molecular Research Center, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Parisa Danaie Fard
- Cellular and Molecular Research Center, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Nahid Azarmehr
- Medicinal Plants Research Center, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Reza Mahmoudi
- Cellular and Molecular Research Center, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Amrollah Roozbehi
- Cellular and Molecular Research Center, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Kazem Zibara
- ER045, Laboratory of Stem Cells, Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.
| | - Ebrahim Hosseini
- Medicinal Plants Research Center, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran.
| |
Collapse
|
2
|
Sarma MK, Saha D, Das BK, Das T, Azizov S, Kumar D. A delve into the pharmacological targets and biological mechanisms of Paederia foetida Linn.: a rather invaluable traditional medicinal plant. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2217-2240. [PMID: 37099165 DOI: 10.1007/s00210-023-02496-4] [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: 02/01/2023] [Accepted: 04/13/2023] [Indexed: 04/27/2023]
Abstract
Drug development from herbal medicines or botanical sources is believed to have a prominent role in the exploration of novel counteractive drugs that has sparked much interest in recent times. Paederia foetida is one such medicinal plant used in both traditional and folkloric medicine. Several parts of the herb are locally utilised as a natural curative agent for several ailments since time immemorial. Paederia foetida indeed possesses anti-diabetic, anti-hyperlipidaemic, antioxidant, nephro-protective, anti-inflammatory, antinociceptive, antitussive, thrombolytic, anti-diarrhoeal, sedative-anxiolytic, anti-ulcer, hepatoprotective activity, anthelmintic and anti-diarrhoeal activity. Furthermore, growing evidence shows many of its active constituents to be effective in cancer, inflammatory diseases, wound healing and spermatogenesis as well. These investigations shed light on possible pharmacological targets and attempts to establish a mechanism of action for these pharmacological effects. These findings contrast the significance of this medicinal plant for further research and for the exploration of novel counteractive drugs to establish a mechanism of action before being employed to healthcare. Pharmacological activities of Paederia foetida and their mechanism of action.
Collapse
Affiliation(s)
- Mrinal Kashyap Sarma
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science, Assam Science and Technology University, Guwahati, 781 017, Assam, India
| | - Dipankar Saha
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science, Assam Science and Technology University, Guwahati, 781 017, Assam, India.
| | - Bhrigu Kumar Das
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science, Assam Science and Technology University, Guwahati, 781 017, Assam, India
| | - Trishna Das
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science, Assam Science and Technology University, Guwahati, 781 017, Assam, India
| | - Shavkatjon Azizov
- Laboratory of Biological Active Macromolecular Systems, Institute of Bioorganic Chemistry, Uzbekistan Academy of Sciences, 100125, Tashkent, Uzbekistan
- Faculty of Life Sciences, Pharmaceutical Technical University, 100084, Tashkent, Uzbekistan
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| |
Collapse
|
3
|
Lunesu MF, Battacone G, Mellino MR, Carta S, Pulina G, Nudda A. The heavy suckling lamb of Sarda dairy sheep and its crossbreed with Dorper rams: Performance, meat quality and consumer perceptions. Meat Sci 2023; 204:109234. [PMID: 37295034 DOI: 10.1016/j.meatsci.2023.109234] [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: 03/09/2023] [Revised: 05/12/2023] [Accepted: 05/28/2023] [Indexed: 06/11/2023]
Abstract
The increase of meat production in dairy sheep farms, has been evaluated by the extension of the suckling period from the traditional 28 days to 75 days to obtain a new product, the heavy suckling lamb. Nineteen single-born Sarda (S) lambs (10 male, 9 female) and 20 single-born Dorper x Sarda (DS) lambs (9 male, 11 female), randomly selected from autumn lambing season, were fed exclusively with maternal milk until slaughtering at 20 ± 0.28 kg of body weight (BW, mean ± standard deviation, SD) and approximately 11 weeks of age. Body weight were recorded at birth and every 15 days until slaughter to estimate average daily gain (ADG). At slaughter, carcass measurements, pH and colour traits were recorded from the left side of the carcass. Proximate composition, fatty acid (FA) profile, cooking and drip losses were evaluated using the Longissimus thoracis et lumborum (LTL) muscle. In addition, Visual Panel Test (VPT) and Taste Panel Test (TPT) were performed. Experimental results evidenced that ADG did not differ between pure and crossbreed lambs and between sexes. The S lamb carcasses showed a higher fat content and rib fat thickness compared to that of crossbreed. No significant differences were found between genetic types and sex for colour and pH determinations, cooking and drip losses, whereas LTL fat of DS displayed a higher nutritional FA profile with higher content of 22:5n-3, 22:6n-3, branched-chain FA, and odd- and branched chain FA. No differences emerged during VPT and TPT, demonstrating that both DS and S lamb meats present no distinguishable visual and eating quality characteristics. The production of heavy suckling lambs from Sarda and Dorper crossbreed by the extension of suckling period appeared a promising strategy for producing meat of high quality, well appreciated by the consumers.
Collapse
Affiliation(s)
| | - Gianni Battacone
- Dipartimento di Agraria, University of Sassari, Viale Italia 39a, 07100 Sassari, Italy.
| | - Maria Rita Mellino
- Dipartimento di Agraria, University of Sassari, Viale Italia 39a, 07100 Sassari, Italy
| | - Silvia Carta
- Dipartimento di Agraria, University of Sassari, Viale Italia 39a, 07100 Sassari, Italy
| | - Giuseppe Pulina
- Dipartimento di Agraria, University of Sassari, Viale Italia 39a, 07100 Sassari, Italy
| | - Anna Nudda
- Dipartimento di Agraria, University of Sassari, Viale Italia 39a, 07100 Sassari, Italy.
| |
Collapse
|
4
|
Abdolmaleky HM, Zhou JR. Underlying Mechanisms of Brain Aging and Neurodegenerative Diseases as Potential Targets for Preventive or Therapeutic Strategies Using Phytochemicals. Nutrients 2023; 15:3456. [PMID: 37571393 PMCID: PMC10473240 DOI: 10.3390/nu15153456] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
During aging, several tissues and biological systems undergo a progressive decline in function, leading to age-associated diseases such as neurodegenerative, inflammatory, metabolic, and cardiovascular diseases and cancer. In this review, we focus on the molecular underpinning of senescence and neurodegeneration related to age-associated brain diseases, in particular, Alzheimer's and Parkinson's diseases, along with introducing nutrients or phytochemicals that modulate age-associated molecular dysfunctions, potentially offering preventive or therapeutic benefits. Based on current knowledge, the dysregulation of microglia genes and neuroinflammation, telomere attrition, neuronal stem cell degradation, vascular system dysfunction, reactive oxygen species, loss of chromosome X inactivation in females, and gut microbiome dysbiosis have been seen to play pivotal roles in neurodegeneration in an interactive manner. There are several phytochemicals (e.g., curcumin, EGCG, fucoidan, galangin, astin C, apigenin, resveratrol, phytic acid, acacetin, daucosterol, silibinin, sulforaphane, withaferin A, and betulinic acid) that modulate the dysfunction of one or several key genes (e.g., TREM2, C3, C3aR1, TNFA, NF-kb, TGFB1&2, SIRT1&6, HMGB1, and STING) affected in the aged brain. Although phytochemicals have shown promise in slowing down the progression of age-related brain diseases, more studies to identify their efficacy, alone or in combinations, in preclinical systems can help to design novel nutritional strategies for the management of neurodegenerative diseases in humans.
Collapse
Affiliation(s)
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA;
| |
Collapse
|
5
|
Shang Q, Chen G, Zhang P, Zhao W, Chen H, Yu D, Yu F, Liu H, Zhang X, He J, Yu X, Zhang Z, Tan R, Wu Z, Tang J, Liang D, Shen G, Jiang X, Ren H. Myristic acid alleviates hippocampal aging correlated with GABAergic signaling. Front Nutr 2022; 9:907526. [PMID: 36159502 PMCID: PMC9493098 DOI: 10.3389/fnut.2022.907526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Previous studies have shown that myristic acid (MA), a saturated fatty acid, could promote the proliferation and differentiation of neural stem cells in vitro. However, the effect of MA on hippocampal neurons aging has not been reported in vivo. Here we employed 22-month-old naturally aged C57BL/6 mice to evaluate the effect and mechanism of MA on hippocampal aging. First, we examined a decreased exploration and spatial memory ability in aging mice using the open field test and Morris water maze. Consistently, aging mice showed degenerative hippocampal histomorphology by H&E and Nissl staining. In terms of mechanism, imbalance of GABRB2 and GABRA2 expression in aging mice might be involved in hippocampus aging by mRNA high throughput sequencing (mRNA-seq) and immunohistochemistry (IHC) validation. Then, we revealed that MA alleviated the damage of exploration and spatial memory ability and ameliorated degeneration and aging of hippocampal neurons. Meanwhile, MA downregulated GABRB2 and upregulated GABRA2 expression, indicating MA might alleviate hippocampal aging correlated with GABAergic signaling. In conclusion, our findings revealed MA alleviated hippocampal aging correlated with GABAergic signaling, which might provide insight into the treatment of aging-associated diseases.
Collapse
Affiliation(s)
- Qi Shang
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guifeng Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenhua Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Honglin Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Die Yu
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fuyong Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huiwen Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuelai Zhang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiahui He
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiang Yu
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhida Zhang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Riwei Tan
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zixian Wu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjing Tang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - De Liang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gengyang Shen
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Nanshan Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine (Shenzhen Nanshan Hospital of Chinese Medicine), Guangzhou, China
- Gengyang Shen
| | - Xiaobing Jiang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Xiaobing Jiang
| | - Hui Ren
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Hui Ren
| |
Collapse
|
6
|
Ahuja A, Tyagi PK, Kumar M, Sharma N, Prakash S, Radha, Chandran D, Dhumal S, Rais N, Singh S, Dey A, Senapathy M, Saleena LAK, Shanavas A, Mohankumar P, Rajalingam S, Murugesan Y, Vishvanathan M, Sathyaseelan SK, Viswanathan S, Kumar KK, Natta S, Mekhemar M. Botanicals and Oral Stem Cell Mediated Regeneration: A Paradigm Shift from Artificial to Biological Replacement. Cells 2022; 11:2792. [PMID: 36139367 PMCID: PMC9496740 DOI: 10.3390/cells11182792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/03/2022] [Accepted: 09/04/2022] [Indexed: 11/23/2022] Open
Abstract
Stem cells are a well-known autologous pluripotent cell source, having excellent potential to develop into specialized cells, such as brain, skin, and bone marrow cells. The oral cavity is reported to be a rich source of multiple types of oral stem cells, including the dental pulp, mucosal soft tissues, periodontal ligament, and apical papilla. Oral stem cells were useful for both the regeneration of soft tissue components in the dental pulp and mineralized structure regeneration, such as bone or dentin, and can be a viable substitute for traditionally used bone marrow stem cells. In recent years, several studies have reported that plant extracts or compounds promoted the proliferation, differentiation, and survival of different oral stem cells. This review is carried out by following the PRISMA guidelines and focusing mainly on the effects of bioactive compounds on oral stem cell-mediated dental, bone, and neural regeneration. It is observed that in recent years studies were mainly focused on the utilization of oral stem cell-mediated regeneration of bone or dental mesenchymal cells, however, the utility of bioactive compounds on oral stem cell-mediated regeneration requires additional assessment beyond in vitro and in vivo studies, and requires more randomized clinical trials and case studies.
Collapse
Affiliation(s)
- Anami Ahuja
- Department of Biotechnology, Dr. A.P.J. Abdul Kalam Technical University, Lucknow 226031, India
- Department of Biotechnology, Meerut Institute of Engineering and Technology, Meerut 250005, India
| | - Pankaj Kumar Tyagi
- Department of Biotechnology, Noida Institute of Engineering & Technology, Greater Noida 201306, India
| | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR–Central Institute for Research on Cotton Technology, Mumbai 400019, India
| | - Naveen Sharma
- Division of Biomedical Informatics, Indian Council of Medical Research, New Delhi 110029, India
| | - Suraj Prakash
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India
| | - Radha
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India
| | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sci-ences, Amrita Vishwa Vidyapeetham University, Coimbatore 642109, India
| | - Sangram Dhumal
- Division of Horticulture, RCSM College of Agriculture, Kolhapur 416004, India
| | - Nadeem Rais
- Department of Pharmacy, Bhagwant University, Ajmer 305004, India
| | - Surinder Singh
- Dr. S. S. Bhatnagar University Institute of Chemical Engineering and Technology, Panjab University, Chandigarh 160014, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, India
| | - Marisennayya Senapathy
- Department of Rural Development and Agricultural Extension, College of Agriculture, Wolaita Sodo University, Wolaita Sodo P.O. Box 138, Ethiopia
| | - Lejaniya Abdul Kalam Saleena
- Department of Food Science and Nutrition, Faculty of Applied Sciences, UCSI University, Kuala Lampur 56000, Malaysia
| | - Arjun Shanavas
- Division of Medicine, Indian Veterinary Research Institute, Bareilly 243122, India
| | - Pran Mohankumar
- School of Agriculture and Biosciences, Karunya Institute of Technology and Sciences, Coimbatore 641114, India
| | - Sureshkumar Rajalingam
- Department of Agronomy, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore 642109, India
| | - Yasodha Murugesan
- Department of Agronomy, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore 642109, India
| | - Marthandan Vishvanathan
- Department of Seed Science and Technology, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore 642109, India
| | | | - Sabareeshwari Viswanathan
- Department of Soil Science and Agricultural Chemistry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore 642109, India
| | - Keerthana Krishna Kumar
- Department of Soil Science and Agricultural Chemistry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore 642109, India
| | - Suman Natta
- ICAR—National Research Centre for Orchids, Pakyong 737106, India
| | - Mohamed Mekhemar
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Chris-tian-Albrecht’s University, 24105 Kiel, Germany
| |
Collapse
|
7
|
Lin SK, Wu YF, Chang WJ, Feng SW, Huang HM. The Treatment Efficiency and Microbiota Analysis of Sapindus mukorossi Seed Oil on the Ligature-Induced Periodontitis Rat Model. Int J Mol Sci 2022; 23:8560. [PMID: 35955695 PMCID: PMC9369273 DOI: 10.3390/ijms23158560] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 02/05/2023] Open
Abstract
Periodontitis is a common oral disease mainly caused by bacterial infection and inflammation of the gingiva. In the prevention or treatment of periodontitis, anti-bacterial agents are used to inhibit pathogen growth, despite increasing levels of bacterial resistance. Sapindus mukorossi Gaertn (SM) seed oil has proven anti-bacterial and anti-inflammation properties. However, the possibility of using this plant to prevent or treat periodontitis has not been reported previously. The aim of this study was to evaluate the effects of SM oil on experimental periodontitis in rats by using micro-CT and microbiota analysis. The distance between cementoenamel junction (CEJ) and alveolar bone crest (ABC) on the sagittal micro-CT slide showed that total bone loss (TBL) was significantly lower in CEJ-ABC distances between SM oil and SM oil-free groups on Day 14. Histology data also showed less alveolar bone resorption, a result consistent result with micro-CT imaging. The microbiota analyzed at phylum and class levels were compared between the SM oil and SM oil-free groups on Day 7 and Day 14. At the phylum level, Proteobacteria, Firmicutes, Bacteroidetes, and Actinobacteria were the dominant bacterium. Firmicutes in box plot analysis was significantly less in the SM oil group than in the SM oil-free group on Day 7. At the class level, Bacteroidia, Gammaproteobacteria, Bacilli, Clostridia, and Erysipelotrichia were the dominant bacteria. The bacteria composition proportion of Bacilli, Clostridiay, and Erysipelotrichia could be seen in the SM oil group significantly less than in t SM oil-free group on Day 7. Overall, the present results show that topical application of SM oil can reduce bone resorption and change bacteria composition in the ligature-induced periodontitis model. According to these results, it is reasonable to suggest SM oil as a potential material for preventing oral disease.
Collapse
Affiliation(s)
- Shih-Kai Lin
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Fan Wu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Jen Chang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Dentistry, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235041, Taiwan
| | - Sheng-Wei Feng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
8
|
Fatty Acids: A Safe Tool for Improving Neurodevelopmental Alterations in Down Syndrome? Nutrients 2022; 14:nu14142880. [PMID: 35889838 PMCID: PMC9323400 DOI: 10.3390/nu14142880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023] Open
Abstract
The triplication of chromosome 21 causes Down syndrome (DS), a genetic disorder that is characterized by intellectual disability (ID). The causes of ID start in utero, leading to impairments in neurogenesis, and continue into infancy, leading to impairments in dendritogenesis, spinogenesis, and connectivity. These defects are associated with alterations in mitochondrial and metabolic functions and precocious aging, leading to the early development of Alzheimer’s disease. Intense efforts are currently underway, taking advantage of DS mouse models to discover pharmacotherapies for the neurodevelopmental and cognitive deficits of DS. Many treatments that proved effective in mouse models may raise safety concerns over human use, especially at early life stages. Accumulating evidence shows that fatty acids, which are nutrients present in normal diets, exert numerous positive effects on the brain. Here, we review (i) the knowledge obtained from animal models regarding the effects of fatty acids on the brain, by focusing on alterations that are particularly prominent in DS, and (ii) the progress recently made in a DS mouse model, suggesting that fatty acids may indeed represent a useful treatment for DS. This scenario should prompt the scientific community to further explore the potential benefit of fatty acids for people with DS.
Collapse
|
9
|
Ma WP, Yin SN, Chen JP, Geng XC, Liu MF, Li HH, Liu M, Liu HB. Stimulating the Hematopoietic Effect of Simulated Digestive Product of Fucoidan from Sargassum fusiforme on Cyclophosphamide-Induced Hematopoietic Damage in Mice and Its Protective Mechanisms Based on Serum Lipidomics. Mar Drugs 2022; 20:201. [PMID: 35323500 PMCID: PMC8950290 DOI: 10.3390/md20030201] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/26/2022] Open
Abstract
Hematopoietic damage is a serious side effect of cytotoxic drugs, and agents promoting hematopoiesis are quite important for decreasing the death rate in cancer patients. In our previous work, we prepared the simulated digestive product of fucoidan from Sargassum fusiforme, DSFF, and found that DSFF could activate macrophages. However, more investigations are needed to further evaluate whether DSFF could promote hematopoiesis in the chemotherapy process. In this study, the protective effect of DSFF (1.8-7.2 mg/kg, i.p.) on cyclophosphamide-induced hematopoietic damage in mice and the underlying mechanisms were investigated. Our results show that DSFF could restore the numbers of white blood cells, neutrophils, and platelets in the peripheral blood, and could also retard bone marrow cell decrease in mice with cyclophosphamide-induced hematopoietic damage. UPLC/Q-Extraction Orbitrap/MS/MS-based lipidomics results reveal 16 potential lipid biomarkers in a serum that responded to hematopoietic damage in mice. Among them, PC (20:1/14:0) and SM (18:0/22:0) were the key lipid molecules through which DSFF exerted protective actions. In a validation experiment, DSFF (6.25-100 μg/mL) could also promote K562 cell proliferation and differentiation in vitro. The current findings indicated that DSFF could affect the blood cells and bone marrow cells in vivo and thus showed good potential and application value in alleviating the hematopoietic damage caused by cyclophosphamide.
Collapse
Affiliation(s)
- Wei-Ping Ma
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (W.-P.M.); (J.-P.C.); (X.-C.G.); (M.-F.L.); (H.-H.L.)
| | - Shi-Ning Yin
- Qingdao Institute for Food and Drug Control, Qingdao 266000, China;
- NMPA Key Laboratory for Quality Research and Evaluation of Marine Traditional Chinese Medicine, Qingdao 266000, China
| | - Jia-Peng Chen
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (W.-P.M.); (J.-P.C.); (X.-C.G.); (M.-F.L.); (H.-H.L.)
| | - Xi-Cheng Geng
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (W.-P.M.); (J.-P.C.); (X.-C.G.); (M.-F.L.); (H.-H.L.)
| | - Ming-Fei Liu
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (W.-P.M.); (J.-P.C.); (X.-C.G.); (M.-F.L.); (H.-H.L.)
| | - Hai-Hua Li
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (W.-P.M.); (J.-P.C.); (X.-C.G.); (M.-F.L.); (H.-H.L.)
| | - Ming Liu
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (W.-P.M.); (J.-P.C.); (X.-C.G.); (M.-F.L.); (H.-H.L.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Hong-Bing Liu
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (W.-P.M.); (J.-P.C.); (X.-C.G.); (M.-F.L.); (H.-H.L.)
- NMPA Key Laboratory for Quality Research and Evaluation of Marine Traditional Chinese Medicine, Qingdao 266000, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
10
|
Shen J, Li J, Hua Y, Ding B, Zhou C, Yu H, Xiao R, Ma W. Association between the Erythrocyte Membrane Fatty Acid Profile and Cognitive Function in the Overweight and Obese Population Aged from 45 to 75 Years Old. Nutrients 2022; 14:nu14040914. [PMID: 35215564 PMCID: PMC8878599 DOI: 10.3390/nu14040914] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 12/21/2022] Open
Abstract
Dietary fatty acid intake is closely related to the cognitive function of the overweight and obese population. However, few studies have specified the correlation between exact fatty acids and cognitive functions in different body mass index (BMI) groups. We aimed to explain these relationships and reference guiding principles for the fatty acid intake of the overweight and obese population. Normal weight, overweight, and obese participants were recruited to receive a cognitive function assessment and dietary survey, dietary fatty acids intake was calculated, and the erythrocyte membrane fatty acid profile was tested by performing a gas chromatography analysis. The percentages of saturated fatty acids (SFAs) in the obese group were higher, while monounsaturated fatty acids (MUFAs) and polyunsaturated fatty acids (PUFAs) were lower than in the normal weight and overweight groups. In the erythrocyte membrane, the increase of n-3 PUFAs was accompanied by cognitive decline in the overweight group, which could be a protective factor for cognitive function in the obese group. High n-6 PUFAs intake could exacerbate the cognitive decline in the obese population. Dietary fatty acid intake had different effects on the cognitive function of overweight and obese people, especially the protective effect of n-3 PUFAs; more precise dietary advice is needed to prevent cognitive impairment.
Collapse
Affiliation(s)
- Jingyi Shen
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (J.S.); (J.L.); (Y.H.); (C.Z.); (H.Y.); (R.X.)
| | - Jinchen Li
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (J.S.); (J.L.); (Y.H.); (C.Z.); (H.Y.); (R.X.)
| | - Yinan Hua
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (J.S.); (J.L.); (Y.H.); (C.Z.); (H.Y.); (R.X.)
| | - Bingjie Ding
- Department of Clinical Nutrition, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China;
| | - Cui Zhou
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (J.S.); (J.L.); (Y.H.); (C.Z.); (H.Y.); (R.X.)
| | - Huiyan Yu
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (J.S.); (J.L.); (Y.H.); (C.Z.); (H.Y.); (R.X.)
| | - Rong Xiao
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (J.S.); (J.L.); (Y.H.); (C.Z.); (H.Y.); (R.X.)
| | - Weiwei Ma
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; (J.S.); (J.L.); (Y.H.); (C.Z.); (H.Y.); (R.X.)
- Correspondence:
| |
Collapse
|
11
|
Calabrese EJ. Hormesis and embryonic stem cells. Chem Biol Interact 2021; 352:109783. [PMID: 34932953 DOI: 10.1016/j.cbi.2021.109783] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 02/07/2023]
Abstract
This paper provides an identification and detailed assessment of hormetic dose responses of embryonic stem cells (ESCs) with particular emphasis on cell renewal (proliferation) and differentiation, underlying mechanistic foundations and potential therapeutic implications. Hormetic dose responses were commonly reported, being induced by a broad range of chemicals, including pharmaceuticals (e.g., atorvastatin, isoproterenol, lithium, nicotine, ouabain), dietary supplements (e.g., curcumin, multiple ginsenosides, resveratrol), endogenous agents (e.g., estrogen, hydrogen peroxide, melatonin), and physical stressor agents (e.g., hypoxia, ionizing radiation). ESC-hormetic dose responses are similar for other stem cell types (e.g., adipose-derived stem cells, apical papilla, bone marrow stem cells, dental pulp stem cells, endothelial stem cells, muscle stem cells, periodontal ligament stem cells, neural stem cells), indicating a high degree of generality for the hormetic-stem cells response. The widespread occurrence of hormetic dose responses shown by ESCs and other stem cells suggests that the hormetic dose response may represent a fundamental and highly conserved evolutionary strategy.
Collapse
Affiliation(s)
- Edward J Calabrese
- School of Public Health and Health Sciences, Department of Environmental Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
12
|
Study on the Mechanism of Liuwei Dihuang Pills in Treating Parkinson's Disease Based on Network Pharmacology. BIOMED RESEARCH INTERNATIONAL 2021; 2021:4490081. [PMID: 34746302 PMCID: PMC8568527 DOI: 10.1155/2021/4490081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/26/2021] [Accepted: 09/16/2021] [Indexed: 11/17/2022]
Abstract
Background Parkinson's disease (PD) is a common neurodegenerative disease in middle-aged and elderly people. Liuwei Dihuang (LWDH) pills have a good effect on PD, but its mechanism remains unclear. Network pharmacology is the result of integrating basic theories and research methods of medicine, biology, computer science, bioinformatics, and other disciplines, which can systematically and comprehensively reflect the mechanism of drug intervention in disease networks. Methods The main components and targets of herbs in LWDH pills were obtained through Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). Its active components were screened based on absorption, distribution, metabolism, and excretion (ADME); the PD-related targets were obtained from the Genecards, OMIM, TTD, and DRUGBANK databases. We used R to take the intersection of LWDH- and PD-related targets and Cytoscape software to construct the drug-component-target network. Moreover, STRING and Cytoscape software was used to analyze protein-protein interactions (PPI), construct a PPI network, and explore potential protein functional modules in the network. The Metascape platform was used to perform KEGG pathway and GO function enrichment analyses. Finally, molecular docking was performed to verify whether the compound and target have good binding activity. Results After screening and deduplication, 210 effective active ingredients, 204 drug targets, 4333 disease targets, and 162 drug-disease targets were obtained. We consequently constructed a drug-component-targets network and a PPI-drug-disease-targets network. The results showed that the hub components of LWDH pills were quercetin, stigmasterol, kaempferol, and beta-sitosterol; the hub targets were AKT1, VEGFA, and IL6. GO and KEGG enrichment analyses showed that these targets are involved in neuronal death, G protein-coupled amine receptor activity, reactive oxygen species metabolic processes, membrane rafts, MAPK signaling pathways, cellular senescence, and other biological processes. Molecular docking showed that the hub components were in good agreement with the hub targets. Conclusion LWDH pills have implications for the treatment of PD since they contain several active components, target multiple ligands, and activate various pathways. The hub components possibly include quercetin, stigmasterol, kaempferol, and beta-sitosterol and act through pairing with hub targets, such as AKT1, VEGFA, and IL6, to regulate neuronal death, G protein-coupled amine receptor activity, reactive oxygen species metabolic process, membrane raft, MAPK signaling pathway, and cellular senescence for the treatment of PD.
Collapse
|
13
|
Abstract
Ulcerative colitis (UC) is a type of inflammatory bowel disease characterized by inflammation of the large intestine, rectal bleeding, and abdominal pain. It can be alleviated by certain bioactive compounds, including α-linolenic acid (ALA), which is a bioactive component in fermented black radish (Raphanus sativus L. var. niger). The aim of this study was to evaluate the anti-inflammatory effects of ALA in dextran sulfate sodium (DSS)-induced UC in mice. UC was induced in C57BL/6 mice by allowing them to freely drink water containing 2.5% DSS for 7 days, followed by oral administration of ALA (30 and 60 mg/kg/day) or vehicle control for 7 days. DSS-induced colitis was evaluated using the Disease Activity Index (DAI) and by measuring colon length and performing a histopathological examination. Compared to the control group, the vehicle-treated group showed a higher DAI score, shorter colon, goblet cell loss, and crypt shortening. The ALA treatment mitigated clinical signs of UC and histopathological changes. Furthermore, it mitigated intestinal inflammation by reducing the expression of ionized calcium binding adaptor molecule 1-positive macrophages in the colon. These results show that ALA alleviates DSS-induced UC by suppressing colon damage, which includes goblet cell loss, crypt shortening, and a reduction of macrophages in the colon.
Collapse
|
14
|
Dodo K, Sato A, Tamura Y, Egoshi S, Fujiwara K, Oonuma K, Nakao S, Terayama N, Sodeoka M. Synthesis of deuterated γ-linolenic acid and application for biological studies: metabolic tuning and Raman imaging. Chem Commun (Camb) 2021; 57:2180-2183. [PMID: 33527102 DOI: 10.1039/d0cc07824g] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
γ-Linolenic acid (GLA) is reported to show tumor-selective cytotoxicity through unidentified mechanisms. Here, to assess the involvement of oxidized metabolites of GLA, we synthesized several deuterated GLAs and evaluated their metabolism and cytotoxicity towards normal human fibroblast WI-38 cells and VA-13 tumor cells generated from WI-38 by transformation with SV40 virus. Deuteration of GLA suppressed both metabolism and cytotoxicity towards WI-38 cells and increased the selectivity for VA-13 cells. Fully deuterated GLA was visualized by Raman imaging, which indicated that GLA is accumulated in intracellular lipid droplets of VA-13 cells. Our results suggest the tumor-selective cytotoxicity is due to GLA itself, not its oxidized metabolites.
Collapse
Affiliation(s)
- Kosuke Dodo
- Synthetic Organic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan. and Sodeoka Live Cell Chemistry Project, ERATO, Japan Sciences and Technology Agency, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan and AMED-CREST, Japan Agency for Medical Research and Development, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan and RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Ayato Sato
- Synthetic Organic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan. and Sodeoka Live Cell Chemistry Project, ERATO, Japan Sciences and Technology Agency, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yuki Tamura
- Synthetic Organic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan. and Sodeoka Live Cell Chemistry Project, ERATO, Japan Sciences and Technology Agency, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Syusuke Egoshi
- Synthetic Organic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan. and AMED-CREST, Japan Agency for Medical Research and Development, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Koichi Fujiwara
- Synthetic Organic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan. and AMED-CREST, Japan Agency for Medical Research and Development, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Kana Oonuma
- RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Shuhei Nakao
- Synthetic Organic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan. and AMED-CREST, Japan Agency for Medical Research and Development, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naoki Terayama
- Synthetic Organic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan. and AMED-CREST, Japan Agency for Medical Research and Development, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan and RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Mikiko Sodeoka
- Synthetic Organic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan. and Sodeoka Live Cell Chemistry Project, ERATO, Japan Sciences and Technology Agency, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan and AMED-CREST, Japan Agency for Medical Research and Development, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan and RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
15
|
Fatty acids promote the expansion of NK-92 cells in vitro by improving energy metabolism. Appl Microbiol Biotechnol 2021; 105:4285-4295. [PMID: 33990857 DOI: 10.1007/s00253-021-11313-y] [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: 01/15/2021] [Revised: 04/13/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
Natural killer-92 cells (NK-92 cells) need to be efficiently expanded by serum-free culture in vitro to meet clinical requirements. Fatty acids mainly provide substrates for energy production, which is of crucial importance to meet the energy demands of highly proliferating cells. This study optimized the medium (EM) for NK-92 cells by designing an experiment to expand cells efficiently. EM, an in-house designed chemically defined serum-free medium, was used as the basal medium. Fatty acids as additive ingredients were screened and optimized by the experimental design method. Three additives, arachidonic acid, myristic acid and palmitoleic acid, were screened; therefore, the optimized medium was named EM-FA. The total cell expansion of NK-92 cells in EM-FA was 72.61±11.95-fold on day 8, which was significantly higher than the 28.55±8.67-fold expansion in EM. To explore the mechanism by which fatty acids promote NK-92 cell expansion, the cell growth kinetics and metabolic characteristics in EM-FA were analyzed. The results showed that NK-92 cells in EM-FA were rapidly expanded while maintaining their cell phenotype and cytotoxicity and enhancing the oxygen consumption rate and mitochondrial function. Fatty acids promoted ATP production to elevate the energy flux for better cell expansion. This study developed an expansion strategy of NK-92 cells in vitro to facilitate their clinical application. KEY POINTS: • Arachidonic acid, myristic acid and palmitoleic acid in serum-free medium were optimized by experimental design to enable the rapid expansion of NK-92 cells in vitro. • Fatty acids upregulated oxidative phosphorylation levels and improved the energy metabolism of NK-92 cells.
Collapse
|
16
|
Li Y, Won KJ, Kim DY, Kim HB, Kang HM, Lee SY, Lee HM. Positive Promoting Effects of Smilax China Flower Absolute on the Wound Healing/Skin Barrier Repair-Related Responses of HaCaT Human Skin Keratinocytes. Chem Biodivers 2021; 18:e2001051. [PMID: 33738961 DOI: 10.1002/cbdv.202001051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/26/2021] [Indexed: 12/11/2022]
Abstract
Smilax china (SC) has pharmacological effects including anti-inflammatory activity, but its effects on skin wound healing and skin barrier function have not been investigated. Here, we investigated the effects of absolute extracted from SC flowers (SCF) on skin wound healing-linked responses and functional skin barrier proteins using human epidermal keratinocytes (HaCaT cells). SCF absolute contained 20 components and was non-toxic to HaCaT cells. The absolute increased the proliferation, migration, and sprout outgrowth of HaCaT cells, and enhanced the activations of serine/threonine-specific protein kinase and extracellular signal-regulated kinase1/2. In addition, it increased the syntheses of type I and IV collagens and the expressions of skin barrier proteins (filaggrin and loricrin). These results indicate SCF absolute may has positive effects on skin wound healing by accelerating keratinocyte migration and proliferation activities and collagen synthesis, and on skin barrier function by upregulating barrier proteins in keratinocytes. We suggest SCF absolute to be considered as a potential means of promoting skin wound and barrier repair.
Collapse
Affiliation(s)
- Yali Li
- Division of Cosmetic and Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, 31499, South Korea
| | - Kyung Jong Won
- Department of Physiology and Medical Science, School of Medicine, Konkuk University, Chungju, 27478, South Korea
| | - Do Yoon Kim
- Division of Cosmetic and Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, 31499, South Korea
| | - Ha Bin Kim
- Division of Cosmetic and Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, 31499, South Korea
| | - Hye Min Kang
- Division of Cosmetic and Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, 31499, South Korea
| | - Su Yeon Lee
- Division of Cosmetic and Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, 31499, South Korea
| | - Hwan Myung Lee
- Division of Cosmetic and Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, 31499, South Korea
| |
Collapse
|
17
|
Fimbristylis ovata extract and its ability to encounter AGEs-induced neurotoxicity in SH-SY5Y. Toxicol Res 2021; 37:355-367. [PMID: 34295799 DOI: 10.1007/s43188-020-00072-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 09/28/2020] [Accepted: 11/04/2020] [Indexed: 10/22/2022] Open
Abstract
Abstract Advanced glycation end products (AGEs) upon binding to its receptor (receptor for AGEs, RAGE) trigger several pathological processes involving oxidative stress and inflammatory pathway which play a pivotal role in various degenerative diseases including Alzheimer's disease. Fimbristylis ovata (F. ovata) has long been reported to be used as a traditional herbal medicine; nonetheless, very few studies have been reported. In this study, the protective effects of F. ovata extract on neurotoxicity of hippocampal neuronal cells (SH-SY5Y) was investigated. When compared to normal control, AGEs treatment significantly induced oxidative stress level and enhanced NF-κB translocation to nucleus in the neuronal cells (p < 0.05). The increase in NF-κB translocation leads to increase in transcription level of the target genes including RAGE and pro-inflammatory cytokines which include interleukin 1 beta (IL1B), tumor necrosis factor-alpha (TNFA) and interleukin 6 (IL6). Pre-treatment of SH-SY5Y with the extracts of F. ovata shows favorable results by significantly suppressing oxidative stress level (p < 0.05) as well transcriptional level of RAGE (p < 0.05) and pro-inflammatory cytokines (p < 0.05). Chemical analysis of F. ovata extracts using High Resolution Liquid Chromatograph Mass Spectrometer (HR-LCMS) and Gas Chromatograph with high resolution Mass Spectrometer (GC-HRMS) suggested some potential active phytochemical compounds. The results from this study may provide possible alternative treatment for prevention and/or therapy of neurodegenerative disorders by targeting the above-mentioned pathways. The role of the phytochemical active ingredient (s) in inhibiting the AGEs-triggered signaling inflammatory pathway should be investigated in future study. Graphic abstract
Collapse
|
18
|
Kim HB, Won KJ, Kim DY, Kang HM, Lee HM. Stimulatory Effects of Paederia foetida Flower Absolute on the Skin Wound and Barrier Repair Activities of Keratinocytes. Chem Biodivers 2020; 17:e2000506. [PMID: 32889769 DOI: 10.1002/cbdv.202000506] [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: 06/25/2020] [Accepted: 09/04/2020] [Indexed: 12/19/2022]
Abstract
Paederia foetida (PF) has antidiarrheal, antidiabetic, and anti-inflammatory activities. However, its biological activities on skin remain unclear. In this study, we examined the effect of PF flower absolute (PFFA) on skin wound healing- and skin barrier-linked responses in human epidermal keratinocytes (HaCaT cells). PFFA contained 23 components and increased the proliferation and sprout outgrowth of HaCaT cells and modestly increased migration. PFFA enhanced the phosphorylation levels of extracellular signal-regulated kinase1/2, serine/threonine-specific protein kinase (AKT), and p38 mitogen-activated protein kinase (MAPK) in HaCaT cells, and upregulated type I and IV collagen synthesis and filaggrin (an epidermal barrier protein) expression in HaCaT cells. These findings suggest PFFA may promote skin wound repair by stimulating migratory and proliferative activities (probably through the AKT/MAPK pathway), collagen synthesis, and skin barrier repair by upregulating the expressions of filaggrin in epidermal keratinocytes. Therefore, PFFA may be useful for developing agents that enhance skin wound and barrier-repair functions.
Collapse
Affiliation(s)
- Ha Bin Kim
- Division of Cosmetic and Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, 31499, South Korea.,National Institute of Forest Science, Seoul, 02455, South Korea
| | - Kyung Jong Won
- Department of Physiology and Medical Science, School of Medicine, Konkuk University, Chungju, 27478, South Korea
| | - Do Yoon Kim
- Division of Cosmetic and Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, 31499, South Korea
| | - Hye Min Kang
- Division of Cosmetic and Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, 31499, South Korea
| | - Hwan Myung Lee
- Division of Cosmetic and Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, 31499, South Korea
| |
Collapse
|
19
|
Shiu ST, Lew WZ, Lee SY, Feng SW, Huang HM. Effects of Sapindus mukorossi Seed Oil on Proliferation, Osteogenetic/Odontogenetic Differentiation and Matrix Vesicle Secretion of Human Dental Pulp Mesenchymal Stem Cells. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E4063. [PMID: 32933188 PMCID: PMC7560370 DOI: 10.3390/ma13184063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/07/2020] [Accepted: 09/11/2020] [Indexed: 12/18/2022]
Abstract
Stem cells have attracted great interest in the development of tissue engineering. However, the self-regeneration and multi-differentiation capabilities of stem cells are easily impaired during cell transplantation. Recent studies have demonstrated that Sapindus mukorossi (S. mukorossi) seed oil has various positive biological effects. However, it is not yet clear whether S. mukorossi seed oil can increase the growth and differentiation of dental pulp mesenchymal stem cells (DPSCs). The aim of this study is to investigate the effects of S. mukorossi seed oil on the proliferation and differentiation of DPSCs. DPSCs with and without S. mukorossi seed oil, respectively, were evaluated and compared. The viabilities of the cells were assessed by MTT tests. The osteogenetic and odontogenetic capacities of the DPSCs were tested using Alizarin red S staining and alkaline phosphatase (ALP) activity assays. In addition, real-time PCR was performed to examine the gene expression of ALP, BMP-2 and DMP-1. Finally, extracellular matrix vesicle secretion was detected via scanning electron microscopy. No significant difference was observed in the viabilities of the DPSCs with and without S. mukorossi seed oil, respectively. However, under osteogenic and odontogenic induction, S. mukorossi seed oil increased the secretion of mineralized nodules and the ALP activity of the DPSCs (p < 0.05). The ALP gene expression of the differentiation-induced DPSCs was also enhanced. Finally, a greater secretion of extracellular matrix vesicles was detected in the DPSCs following odontogenic induction complemented with S. mukorossi seed oil. Overall, the present results show that S. mukorossi seed oil promotes the osteogenic/odontogenic differentiation and matrix vesicle secretion of DPSCs.
Collapse
Affiliation(s)
- Shiau-Ting Shiu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (S.-T.S.); (W.-Z.L.); (S.-Y.L.)
- Department of Dentistry, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Wei-Zhen Lew
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (S.-T.S.); (W.-Z.L.); (S.-Y.L.)
| | - Sheng-Yang Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (S.-T.S.); (W.-Z.L.); (S.-Y.L.)
- Department of Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei 11031, Taiwan
| | - Sheng-Wei Feng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (S.-T.S.); (W.-Z.L.); (S.-Y.L.)
- Division of Prosthodontics, Department of Dentistry, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (S.-T.S.); (W.-Z.L.); (S.-Y.L.)
- Graduate Institute of Biomedical Optomechatronics, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
20
|
Network Pharmacology-Based Strategy to Investigate the Pharmacological Mechanisms of Ginkgo biloba Extract for Aging. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8508491. [PMID: 32802136 PMCID: PMC7403930 DOI: 10.1155/2020/8508491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022]
Abstract
Aging is a main risk factor for a number of debilitating diseases and contributes to an increase in mortality. Previous studies have shown that Ginkgo biloba extract (EGb) can prevent and treat aging-related diseases, but its pharmacological effects need to be further clarified. This study aimed to propose a network pharmacology-based method to identify the therapeutic pathways of EGb for aging. The active components of EGb and targets of sample chemicals were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) database. Information on aging-related genes was obtained from the Human Ageing Genomic Resources database and JenAge Ageing Factor Database. Subsequently, a network containing the interactions between the putative targets of EGb and known therapeutic targets of aging was established, which was used to investigate the pharmacological mechanisms of EGb for aging. A total of 24 active components, 154 targets of active components of EGb, and 308 targets of aging were obtained. Network construction and pathway enrichment were conducted after data integration. The study found that flavonoids (quercetin, luteolin, and kaempferol) and beta-sitosterol may be the main active components of EGb. The top eight candidate targets, namely, PTGS2, PPARG, DPP4, GSK3B, CCNA2, AR, MAPK14, and ESR1, were selected as the main therapeutic targets of EGb. Pathway enrichment results in various pathways were associated with inhibition of oxidative stress, inhibition of inflammation, amelioration of insulin resistance, and regulation of cellular biological processes. Molecular docking results showed that PPARG had better binding capacity with beta-sitosterol, and PTGS2 had better binding capacity with kaempferol and quercetin. The main components of EGb may act on multiple targets, such as PTGS2, PPARG, DPP4, and GSK3B, to regulate multiple pathways, and play an antiaging role by inhibiting oxidative stress, inhibiting inflammation, and ameliorating insulin resistance.
Collapse
|