1
|
Zhang W, Zhang X, Wang K, Liu Z, Zhang L, Liu S, He K, Wang H, Wang J, Wang Y, Wang Y, Yang Y, Wu H. Single-nucleus transcriptome profiling provides insights into the pathophysiology of adhesive arachnoiditis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167655. [PMID: 39755217 DOI: 10.1016/j.bbadis.2024.167655] [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: 05/15/2024] [Revised: 12/08/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025]
Abstract
Adhesive arachnoiditis (AA) is a rare form of chronic degenerative pathology associated with persistent inflammation in the arachnoid matter of the spinal cord. Despite the existing knowledge, the detailed pathological mechanisms underlying AA are not fully understood. This study aimed to elucidate through comprehensive single nuclei RNA sequencing (snRNA-seq) to delineate the transcriptomic landscape of AA. From six arachnoid membrane samples, a total of 52,886 cells met the quality control standards for analysis. The main cell populations identified with specific gene markers were as follows: fibroblasts, glial cells, microglial cells, endothelial cells, mural cells, plasma cells, and T cells. Downstream analysis of fibroblasts, glial cells, and microglial cells was performed. Notably, fibroblast subsets 1 and 3 demonstrated a strong association with AA. Among them, subcluster 3 demonstrated elevated expression of genes COL1A1, COL3A1, and FN1, indicative of enhanced Wnt/β-catenin and extracellular matrix (ECM) synthesis pathways. Subcluster 3 was predicted to progressively transform into subcluster 1. In subcluster 1, there was a significant upregulation of genes such as BMP and ALPL, signaling enhanced activation of calcification-related pathways. This was highly relevant to end-stage arachnoid ossification formation. After being activated, microglial cells transformed into inflammatory disease-associated microglial cells and continued to express high levels of chemokines CCL2, CCL4, IL-1β, and other inflammatory factors NAMPT, INPP5D and NLRP3. This might be the main reason why AA recurrence is frequently observed in patients. These insights enhance our understanding of the pathological progression of AA and may contribute to the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Weikang Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiangyu Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Kai Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Zhenlei Liu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lei Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shaocheng Liu
- Beijing Mentougou District Hospital, Beijing 102300, China
| | - Kun He
- Department of Neurosurgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - He Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Junyi Wang
- Beijing Science and Technology Innovation Group, Beijing 100101, China
| | - Yaobin Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yutian Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yuhua Yang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Hao Wu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
2
|
Kolotyeva NA, Groshkov AA, Rozanova NA, Berdnikov AK, Novikova SV, Komleva YK, Salmina AB, Illarioshkin SN, Piradov MA. Pathobiochemistry of Aging and Neurodegeneration: Deregulation of NAD+ Metabolism in Brain Cells. Biomolecules 2024; 14:1556. [PMID: 39766263 PMCID: PMC11673498 DOI: 10.3390/biom14121556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
NAD+ plays a pivotal role in energy metabolism and adaptation to external stimuli and stressful conditions. A significant reduction in intracellular NAD+ levels is associated with aging and contributes to the development of chronic cardiovascular, neurodegenerative, and metabolic diseases. It is of particular importance to maintain optimal levels of NAD+ in cells with high energy consumption, particularly in the brain. Maintaining the tissue level of NAD+ with pharmacological tools has the potential to slow down the aging process, to prevent the development of age-related diseases. This review covers key aspects of NAD+ metabolism in terms of brain metabolic plasticity, including NAD+ biosynthesis and degradation in different types of brain cells, as well as its contribution to the development of neurodegeneration and aging, and highlights up-to-date approaches to modulate NAD+ levels in brain cells.
Collapse
|
3
|
Sekiya M, Sakakibara Y, Hirota Y, Ito N, Chikamatsu S, Takei K, Nishijima R, Iijima KM. Decreased plasma nicotinamide and altered NAD + metabolism in glial cells surrounding Aβ plaques in a mouse model of Alzheimer's disease. Neurobiol Dis 2024; 202:106694. [PMID: 39374707 DOI: 10.1016/j.nbd.2024.106694] [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: 02/20/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/09/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and a leading cause of senile dementia. Amyloid-β (Aβ) accumulation triggers chronic neuroinflammation, initiating AD pathogenesis. Recent clinical trials for anti-Aβ immunotherapy underscore that blood-based biomarkers have significant advantages and applicability over conventional diagnostics and are an unmet clinical need. To further advance ongoing clinical trials and identify novel therapeutic targets for AD, developing additional plasma biomarkers closely associated with pathogenic mechanisms downstream of Aβ accumulation is critically important. To identify plasma metabolites reflective of neuroinflammation caused by Aβ pathology, we performed untargeted metabolomic analyses of the plasma by capillary electrophoresis time-of-flight mass spectrometry (CE-TOFMS) and analyzed the potential roles of the identified metabolic changes in the brain neuroinflammatory response using the female App knock-in (AppNLGF) mouse model of Aβ amyloidosis. The CE-TOFMS analysis of plasma samples from female wild-type (WT) and AppNLGF mice revealed that plasma levels of nicotinamide, a nicotinamide adenine dinucleotide (NAD+) precursor, were decreased in AppNLGF mice, and altered metabolite profiles were enriched for nicotinate/nicotinamide metabolism. In AppNLGF mouse brains, NAD+ levels were unaltered, but mRNA levels of NAD+-synthesizing nicotinate phosphoribosyltransferase (Naprt) and NAD+-degrading Cd38 genes were increased. These enzymes were induced in reactive astrocytes and microglia surrounding Aβ plaques in the cortex and hippocampus of female AppNLGF mouse brains, suggesting neuroinflammation increases NAD+ metabolism. This study suggests plasma nicotinamide could be indicative of the neuroinflammatory response and that nicotinate and nicotinamide metabolism are potential therapeutic targets for AD, by targeting both neuroinflammation and neuroprotection.
Collapse
Affiliation(s)
- Michiko Sekiya
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Yasufumi Sakakibara
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Yu Hirota
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Reseach Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Naoki Ito
- Brain-Skeletal Muscle Connection in Aging Project Team, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Sachie Chikamatsu
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Kimi Takei
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Risa Nishijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Koichi M Iijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| |
Collapse
|
4
|
Zhao N, Xu B, Xia J, Wang J, Zhang X, Yan Q. Effect of alternating nicotinamide phosphoribosyltransferase expression levels on mitophagy in Alzheimer's disease mouse models. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167288. [PMID: 38862096 DOI: 10.1016/j.bbadis.2024.167288] [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: 01/08/2024] [Revised: 05/17/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024]
Abstract
AD is the abbreviation for Alzheimer's Disease, which is a neurodegenerative disorder that features progressive dysfunction in cognition. Previous research has reported that mitophagy impairment and mitochondrial dysfunction have been crucial factors in the AD's pathogenesis. More recently, literature has emerged which offers findings suggesting that the nicotinamide adenine dinucleotide (short for NAD+) augmentation eliminates the defective mitochondria and restores mitophagy. Meanwhile, as an enzyme which is rate-limiting, the Nicotinamide phosphoribosyltransferase, or NAMPT, is part of the salvage pathway of NAD+ synthesis. Therefore, the aim of the research project has been to produce proof for how the NAMPT-NAD +-silent information-regulated transcription factors1/3 (short for SIRT1/3) axis function in mediating mitophagy in APP/PS1 mice aged six months. The results revealed that the NAMPT-NAD+-SIRT1/3 axis in the APP/PS1 mice's hippocampus was considerably declined. Surprisingly, P7C3 (an NAMPT activator) noticeably promoted the NAD+-SIRT1/3 axis, improved mitochondrial structure and function, enhanced mitophagy activity along with the ability of learning and memory. While FK866 (an NAMPT inhibitor) reversed the decreased NAD+-SIRT1/3 axis, and even exacerbated Aβ plaque deposition level in the APP/PS1 mice's hippocampus. The findings observed in this study indicate two main points: avoiding downregulation of the NAMPT activity can prevent AD-related mitophagy impairment; on the other hand, NAMPT characterizes a potential therapeutic intervention regarding AD pathogenesis.
Collapse
Affiliation(s)
- Na Zhao
- College of Sports and Health, Shandong Sport University, Jinan 250102, China.
| | - Bo Xu
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Jie Xia
- Department of Physical Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Wang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong, China
| | - Xianliang Zhang
- School of Physical Education, Shandong University, Jinan, China
| | - Qingwei Yan
- School of Physical Education, Xizang Minzu University, Xianyang 712082, Shanxi, China
| |
Collapse
|
5
|
Ozcan M, Ayar A. Endocrine Aspects of Pain Pathophysiology: Focus on Adipose Tissue. Neuroendocrinology 2024; 114:894-906. [PMID: 38801814 DOI: 10.1159/000539531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Multiple factors, including neurobiological, hormonal, psychological, and social/cultural norms, influence the manner in which individuals experience pain. Adipose tissue, once considered solely an energy storage site, has been recognized as a significant endocrine organ that produces and releases a range of hormones and cytokines. In recent years, research has highlighted the role of adipose tissue and its endocrine factors in the pathophysiology of pain. SUMMARY This narrative review aimed to provide a comprehensive overview of the current knowledge on the endocrine aspects of pain pathophysiology, with a specific focus on adipose tissue. We examine the role of adipokines released by adipose tissue, such as leptin, adiponectin, resistin, visfatin, asprosin in pain perception and response. We also explore the clinical implications of these findings, including the potential for personalized pain management based on endocrine factors and adipose tissue. KEY MESSAGES Overall, given this background, this review intended to highlight the importance of understanding the endocrine aspects of pain pathophysiology, particularly focusing on the role of adipose tissue, in the development of chronic pain and adipokines. Better understanding the role of adipokines in pain modulation might have therapeutic implications by providing novel targets for addressing underlying mechanism rather than directly focusing on symptoms for chronic pain, particularly in obese individuals.
Collapse
Affiliation(s)
- Mete Ozcan
- Department of Biophysics, Firat University Medical Faculty, Elazig, Turkey
| | - Ahmet Ayar
- Department of Physiology, Karadeniz Technical University Medical Faculty, Trabzon, Turkey
| |
Collapse
|
6
|
Liu Q, Gao K, Ding X, Mo D, Guo H, Chen B, Xia B, Ye C, Chen G, Guo C. NAMPT inhibition relieves intestinal inflammation by regulating macrophage activation in experimental necrotizing enterocolitis. Biomed Pharmacother 2023; 165:115012. [PMID: 37329710 DOI: 10.1016/j.biopha.2023.115012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/07/2023] [Accepted: 06/11/2023] [Indexed: 06/19/2023] Open
Abstract
Nicotinamide phosphoribosyl transferase (NAMPT) is associated with various NAD+ -consuming enzymatic reactions. The precise role in intestinal mucosal immunity in necrotizing enterocolitis (NEC) is not well defined. Here, we examined whether NAMPT inhibition by the highly specific inhibitor FK866 could alleviate intestinal inflammation during the pathogenesis of NEC. In the present study, we showed that NAMPT expression was upregulated in the human terminal ileum of human infants with NEC. FK866 administration attenuated M1 macrophage polarization and relieved the symptoms of experimental NEC pups. FK866 inhibited intercellular NAD+ levels, macrophage M1 polarization, and the expression of NAD+ -dependent enzymes, such as poly (ADP ribose) polymerase 1 (PARP1) and Sirt6. Consistently, the capacity of macrophages to phagocytose zymosan particles, as well as antibacterial activity, were impaired by FK866, whereas NMN supplementation to restore NAD+ levels reversed the changes in phagocytosis and antibacterial activity. In conclusion, FK866 reduced intestinal macrophage infiltration and skewed macrophage polarization, which is implicated in intestinal mucosal immunity, thereby promoting the survival of NEC pups.
Collapse
Affiliation(s)
- Qianyang Liu
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China; Department of Pediatric Surgery, Chongqing Health Center for Women and Children, Chongqing Medical University, Chongqing 400054, China; Department of Obstetrics and Gynecology, Women and Chidren's Hospital, Chongqing Medical University, Chongqing, China
| | - Kai Gao
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China; Department of Pediatric Surgery, Chongqing Health Center for Women and Children, Chongqing Medical University, Chongqing 400054, China
| | - Xionghui Ding
- Department of Burn, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Dandan Mo
- Department of Pediatric Surgery, Chongqing Health Center for Women and Children, Chongqing Medical University, Chongqing 400054, China
| | - Hongjie Guo
- Department of anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Bailin Chen
- Department of General Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Bingshan Xia
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China; Department of Pediatric Surgery, Chongqing Health Center for Women and Children, Chongqing Medical University, Chongqing 400054, China; Department of Obstetrics and Gynecology, Women and Chidren's Hospital, Chongqing Medical University, Chongqing, China
| | - Cuilian Ye
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Gongli Chen
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China; Department of Pediatric Surgery, Chongqing Health Center for Women and Children, Chongqing Medical University, Chongqing 400054, China; Department of Obstetrics and Gynecology, Women and Chidren's Hospital, Chongqing Medical University, Chongqing, China.
| | - Chunbao Guo
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China; Department of Pediatric Surgery, Chongqing Health Center for Women and Children, Chongqing Medical University, Chongqing 400054, China; Department of Obstetrics and Gynecology, Women and Chidren's Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
7
|
Narne P, Phanithi PB. Role of NAD + and FAD in Ischemic Stroke Pathophysiology: An Epigenetic Nexus and Expanding Therapeutic Repertoire. Cell Mol Neurobiol 2023; 43:1719-1768. [PMID: 36180651 PMCID: PMC11412205 DOI: 10.1007/s10571-022-01287-4] [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] [Received: 04/03/2022] [Accepted: 09/15/2022] [Indexed: 11/03/2022]
Abstract
The redox coenzymes viz., oxidized β-nicotinamide adenine dinucleotide (NAD+) and flavin adenine dinucleotide (FAD) by way of generation of optimal reducing power and cellular energy currency (ATP), control a staggering array of metabolic reactions. The prominent cellular contenders for NAD+ utilization, inter alia, are sirtuins (SIRTs) and poly(ADP-ribose) polymerase (PARP-1), which have been significantly implicated in ischemic stroke (IS) pathogenesis. NAD+ and FAD are also two crucial epigenetic enzyme-required metabolites mediating histone deacetylation and poly(ADP-ribosyl)ation through SIRTs and PARP-1 respectively, and demethylation through FAD-mediated lysine specific demethylase activity. These enzymes and post-translational modifications impinge on the components of neurovascular unit, primarily neurons, and elicit diverse functional upshots in an ischemic brain. These could be circumstantially linked with attendant cognitive deficits and behavioral outcomes in post-stroke epoch. Parsing out the contribution of NAD+/FAD-synthesizing and utilizing enzymes towards epigenetic remodeling in IS setting, together with their cognitive and behavioral associations, combined with possible therapeutic implications will form the crux of this review.
Collapse
Affiliation(s)
- Parimala Narne
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana State, 500046, India.
| | - Prakash Babu Phanithi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana State, 500046, India.
| |
Collapse
|
8
|
Tribble JR, Hagström A, Jusseaume K, Lardner E, Wong RCB, Stålhammar G, Williams PA. NAD salvage pathway machinery expression in normal and glaucomatous retina and optic nerve. Acta Neuropathol Commun 2023; 11:18. [PMID: 36681854 PMCID: PMC9867855 DOI: 10.1186/s40478-023-01513-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/10/2023] [Indexed: 01/23/2023] Open
Abstract
Glaucoma is the leading cause of irreversible blindness and is a major health and economic burden. Current treatments do not address the neurodegenerative component of glaucoma. In animal models of glaucoma, the capacity to maintain retinal nicotinamide adenine dinucleotide (NAD) pools declines early during disease pathogenesis. Treatment with nicotinamide, an NAD precursor through the NAD salvage pathway, robustly protects against neurodegeneration in a number of glaucoma models and improves vision in existing glaucoma patients. However, it remains unknown in humans what retinal cell types are able to process nicotinamide to NAD and how these are affected in glaucoma. To address this, we utilized publicly available RNA-sequencing data (bulk, single cell, and single nucleus) and antibody labelling in highly preserved enucleated human eyes to identify expression of NAD synthesizing enzyme machinery. This identifies that the neural retina favors expression of the NAD salvage pathway, and that retinal ganglion cells are particularly enriched for these enzymes. NMNAT2, a key terminal enzyme in the salvage pathway, is predominantly expressed in retinal ganglion cell relevant layers of the retina and declines in glaucoma. These findings suggest that human retinal ganglion cells can directly utilize nicotinamide and could maintain a capacity to do so in glaucoma, showing promise for ongoing clinical trials.
Collapse
Affiliation(s)
- James R Tribble
- Division of Eye and Vision, Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Anna Hagström
- Division of Eye and Vision, Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Kenza Jusseaume
- Division of Eye and Vision, Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Emma Lardner
- Division of Eye and Vision, Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Raymond Ching-Bong Wong
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Gustav Stålhammar
- Division of Eye and Vision, Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Pete A Williams
- Division of Eye and Vision, Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
9
|
Zhang Z, Xiao K, Wang S, Ansari AR, Niu X, Yang W, Lu M, Yang Z, Rehman ZU, Zou W, Bei W, Song H. Visfatin is a multifaceted molecule that exerts regulation effects on inflammation and apoptosis in RAW264.7 cells and mice immune organs. Front Immunol 2022; 13:1018973. [PMID: 36532047 PMCID: PMC9753570 DOI: 10.3389/fimmu.2022.1018973] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
Visfatin, a multifunctional adipocytokine, is particularly important in the regulation of apoptosis and inflammation through an unidentified mechanism. Clarifying the control mechanisms of visfatin on inflammation and apoptosis in RAW264.7 cells and mice immunological organs was the goal of the current investigation. In order to create a pathophysiological model, the RAW264.7 cells were stimulated with 200 ng/mL visfatin and 20 μg/mL lipopolysaccharide (LPS), either separately or combined. The effects of exogenous visfatin on inflammation and apoptosis in RAW264.7 cells were investigated by flow cytometry assay, RNA-seq analysis and fluorescence quantitative PCR. According to the findings, exogenous visfatin exhibits dual effects on inflammation by modulating the expression of IL-1α, TNFRSF1B, and LIF as well as taking part in various signaling pathways, including the MAPK and Rap1 signaling pathways. By controlling the expression levels of Bcl2l1, Bcl2a1a, and Fas and primarily participating in the PI3K/AKT signaling pathway and Hippo signaling pathway, exogenous visfatin can inhibit apoptosis in RAW264.7 cells. The visfatin inhibitor FK866 was used to further confirm the effects of visfatin on inflammation and apoptosis in mice immune organs. Subsequently, mice spleen and thymus were collected. It is interesting to note that in LPS-treated mice, suppression of endogenous visfatin might worsen the immune system's inflammatory response and even result in rapid mortality. Additionally, endogenous visfatin promotes the apoptosis in mice immune organs by regulating the expression levels of Bcl2l1, Fas, Caspase 3, Bcl2a1a, and Bax. Together, these results imply that visfatin is a multifaceted molecule that regulates inflammation and apoptosis in RAW264.7 cells and mice immunological organs by taking part in a variety of biological processes and regulating the amounts of associated cytokines expression. Our findings offer additional understandings of how visfatin affects apoptosis and inflammation.
Collapse
Affiliation(s)
- Zhewei Zhang
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ke Xiao
- The Brain Cognition and Brain Disease Institute of Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Sheng Wang
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Abdur Rahman Ansari
- Section of Anatomy and Histology, Department of Basic Sciences, College of Veterinary & Animal Sciences, Jhang University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Xiaoyu Niu
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wenjie Yang
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Mengqi Lu
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhi Yang
- Animal Health Supervision Institute of Taihe County, Fuyang, China
| | - Zia ur Rehman
- College of Veterinary Sciences, Faculty of Animal Husbandry and Veterinary Sciences, University of Agriculture, Peshawar, Pakistan
| | - Weihua Zou
- Wuhan Keqian Biology Company Limited, Wuhan, China
| | - Weicheng Bei
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Hui Song
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China,*Correspondence: Hui Song,
| |
Collapse
|
10
|
Xu Q, Liu X, Mohseni G, Hao X, Ren Y, Xu Y, Gao H, Wang Q, Wang Y. Mechanism research and treatment progress of NAD pathway related molecules in tumor immune microenvironment. Cancer Cell Int 2022; 22:242. [PMID: 35906622 PMCID: PMC9338646 DOI: 10.1186/s12935-022-02664-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/19/2022] [Indexed: 11/21/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is the core of cellular energy metabolism. NAMPT, Sirtuins, PARP, CD38, and other molecules in this classic metabolic pathway affect many key cellular functions and are closely related to the occurrence and development of many diseases. In recent years, several studies have found that these molecules can regulate cell energy metabolism, promote the release of related cytokines, induce the expression of neoantigens, change the tumor immune microenvironment (TIME), and then play an anticancer role. Drugs targeting these molecules are under development or approved for clinical use. Although there are some side effects and drug resistance, the discovery of novel drugs, the development of combination therapies, and the application of new technologies provide solutions to these challenges and improve efficacy. This review presents the mechanisms of action of NAD pathway-related molecules in tumor immunity, advances in drug research, combination therapies, and some new technology-related therapies.
Collapse
Affiliation(s)
- QinChen Xu
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Xiaoyan Liu
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Ghazal Mohseni
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Xiaodong Hao
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Yidan Ren
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Yiwei Xu
- Marine College, Shandong University, 264209, Weihai, China
| | - Huiru Gao
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China
| | - Qin Wang
- Department of Anesthesiology, Cheeloo College of Medicine, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| | - Yunshan Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Jinan, Shandong, China.
| |
Collapse
|
11
|
Zhang F, Wang Z, Sun B, Huang Y, Chen C, Hu J, Li L, Xia P, Ye Z. Propofol rescued astrocytes from LPS-induced inflammatory response via blocking LncRNA-MEG3/NF-κB axis. Curr Neurovasc Res 2022; 19:5-18. [PMID: 35297349 DOI: 10.2174/1567202619666220316112509] [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: 12/13/2021] [Revised: 12/27/2021] [Accepted: 12/31/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Evidences had demonstrated that propofol attenuated neuro-inflammation following brain ischemia. Moreover, LncRNA-MEG3 was identified as an independent prognostic marker for ischemic stroke patients, and was found to be correlated with cerebral ischemia in animal models. Therefore, the current study explored the role of propofol on lipopolysaccharide (LPS)-mediated inflammation in cultured astrocytes, along with the molecular mechanism involved in LncRNA-MEG3/NF-κB axis. METHODS The primary cultured astrocytes isolated from rats were used to establish an inflammatory model, which were treated with LPS. Propofol was administrated to the primary cultured astrocytes during LPS treatment. The effect of propofol on pro-inflammatory cytokines and the LncRNA-MEG3/NF-κB pathway were detected by ELISA, qRT-PCR and Western Blot assay, respectively. Then, dual-luciferase assay, chromatin immunoprecipitation and RNA immunoprecipitation were used to determine the interaction between LncRNA-MEG3 and NF-κB. RESULTS Our study found that propofol significantly reduced LncRNA-MEG3 expression, which was elevated in LPS-stimulated astrocytes. Moreover, both propofol and LncRNA-MEG3 knockdown remarkably alleviated LPS-induced cytotoxicity by suppressing expressions and release of pro-inflammatory cytokines. Loss of LncRNA-MEG3 notably suppressed the NF-κB activity and its phosphorylated activation. Additionally, it was also observed that LncRNA-MEG3 could bind nuclear p65/p50, and promote the binding of NF-κB to IL-6 and TNF-α promoters in the nucleus, subsequently stimulating the production of inflammatory cytokines in LPS-treated astrocytes. Furthermore, a specific inhibitor of NF-κB, PDTC rescued astrocytes from LPS exposure without affecting LncRNA-MEG3 expression. CONCLUSION These findings demonstrated that LncRNA-MEG3 acted as a positive regulator of NF-κB, mediated the neuroprotection of propofol in LPS-triggered astrocytes injury.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Zhihua Wang
- Department of Anesthesiology, Hainan General Hospital, Haikou, Hainan Province, China
| | - Bei Sun
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Yan Huang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Cheng Chen
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Jie Hu
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Longyan Li
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Pingping Xia
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| |
Collapse
|
12
|
Ostrakhovitch EA, Song ES, Macedo JKA, Gentry MS, Quintero JE, van Horne C, Yamasaki TR. Analysis of circulating metabolites to differentiate Parkinson's disease and essential tremor. Neurosci Lett 2021; 769:136428. [PMID: 34971771 DOI: 10.1016/j.neulet.2021.136428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/24/2021] [Accepted: 12/24/2021] [Indexed: 12/26/2022]
Abstract
Parkinson disease (PD) and essential tremor (ET) are two common adult-onset tremor disorders in which prevalence increases with age. PD is a neurodegenerative condition with progressive disability. In ET, neurodegeneration is not an established etiology. We sought to determine whether an underlying metabolic pattern may differentiate ET from PD. Circulating metabolites in plasma and cerebrospinal fluid were analyzed using gas chromatography-mass spectroscopy. There were several disrupted pathways in PD compared to ET plasma including glycolysis, tyrosine, phenylalanine, tyrosine biosynthesis, purine and glutathione metabolism. Elevated α-synuclein levels in plasma and CSF distinguished PD from ET. The perturbed metabolic state in PD was associated with imbalance in the pentose phosphate pathway, deficits in energy production, and change in NADPH, NADH and nicotinamide phosphoribosyltransferase levels. This work demonstrates significant metabolic differences in plasma and CSF of PD and ET patients.
Collapse
Affiliation(s)
| | - Eun-Suk Song
- Department of Neurology, University of Kentucky, Lexington, KY, 40536, USA
| | - Jessica K A Macedo
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Matthew S Gentry
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Jorge E Quintero
- Department of Neurosurgery, University of Kentucky, Lexington, KY, 40536, USA
| | - Craig van Horne
- Department of Neurosurgery, University of Kentucky, Lexington, KY, 40536, USA
| | - Tritia R Yamasaki
- Department of Neurology, University of Kentucky, Lexington, KY, 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA; Veterans Affairs Medical Center, Lexington, KY, 40536, USA
| |
Collapse
|
13
|
Adipokines as Immune Cell Modulators in Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms221910845. [PMID: 34639186 PMCID: PMC8509121 DOI: 10.3390/ijms221910845] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS), a chronic inflammatory and demyelinating disease of the central nervous system (CNS), is a major clinical and societal problem, which has a tremendous impact on the life of patients and their proxies. Current immunomodulatory and anti-inflammatory therapies prove to be relatively effective; however, they fail to concomitantly stop ongoing neurological deterioration and do not reverse acquired disability. The proportion to which genetic and environmental factors contribute to the etiology of MS is still incompletely understood; however, a recent association between MS etiology and obesity was shown, with obesity greatly increasing the risk of developing MS. An altered balance of adipokines, which are white adipose tissue (WAT) hormones, plays an important role in the low-grade chronic inflammation during obesity by their pervasive modification of local and systemic inflammation. Vice versa, inflammatory factors secreted by immune cells affect adipokine function. To explore the role of adipokines in MS pathology, we will here review the reciprocal effects of adipokines and immune cells and summarize alterations in adipokine levels in MS patient cohorts. Finally, we will discuss proof-of-concept studies demonstrating the therapeutic potential of adipokines to target both neuroinflammation and neurodegeneration processes in MS.
Collapse
|
14
|
Zeng M, Wei TF, Chen C, Shen C, Gao TY, Xie X, Wu M, Lu YB, Zhang WP. Nicotinamide phosphoribosyltransferase inhibitor ameliorates mouse aging-induced cognitive impairment. J Cereb Blood Flow Metab 2021; 41:2510-2523. [PMID: 33818184 PMCID: PMC8504953 DOI: 10.1177/0271678x211006291] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is the key enzyme for the synthesis of nicotinamide adenine dinucleotide (NAD) in the salvaging pathway. Though NAMPT inhibitors such as FK866 were originally developed as anti-cancer drugs, they also display neuroprotective effects. Here we show that the administration of FK866 at 0.5 mg/kg (ip, qod) for four weeks, i.e., ∼1% of the dose used for the treatment of cancer, significantly alleviates the aging-induced impairment of cognition and locomotor activity. Mechanistically, FK866 enhanced autophagy, reduced protein aggregation, and inhibited neuroinflammation indicated by decreasing TNFα, IL-6, GFAP, and Iba1 levels in the aged mouse brain. Though FK866 did not affect the total NAD and nicotinamide mononucleotide (NMN) levels in the mouse brain at the dose we used, FK866 increased nicotinamide (NAM) level in the young mouse brain and decreased NAM level in the aged mouse brain. On the other hand, FK866 did not affect the serum glucose, cholesterol, and triglyceride of young and aged mice and exhibited no effects on the various indices of young mice. Thus, the NAMPT inhibitor can be repurpose to counteract the cognitive impairment upon aging. We also envision that NAMPT inhibitor can be used for the treatment of age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Min Zeng
- Departments of Pharmacology and Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
| | - Tao-Feng Wei
- Departments of Pharmacology and Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
| | - Cong Chen
- Departments of Pharmacology and Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
| | - Chen Shen
- Departments of Pharmacology and Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
| | - Tong-Yao Gao
- Departments of Pharmacology and Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
| | - Xian Xie
- Hospital of Stomatology, Zhejiang University School of Medicine, Zhejiang, China
| | - Ming Wu
- Department of Thoracic Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
| | - Yun-Bi Lu
- Departments of Pharmacology and Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
| | - Wei-Ping Zhang
- Departments of Pharmacology and Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
| |
Collapse
|
15
|
He Y, Dai J, Niu M, Li B, Chen C, Jiang M, Wu Z, Bao J, Zhang X, Li L, Husain SZ, Hu G, Wen L. Inhibition of nicotinamide phosphoribosyltransferase protects against acute pancreatitis via modulating macrophage polarization and its related metabolites. Pancreatology 2021; 21:870-883. [PMID: 33810973 DOI: 10.1016/j.pan.2021.03.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/03/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & OBJECTIVES Acute pancreatitis is a common inflammatory disorder of the exocrine pancreas with no specific therapy. Intracellular nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in nicotinamide adenine dinucleotide (NAD) salvage pathway, is involved in many inflammatory disorders. In this study, we investigated the role of NAMPT in experimental acute pancreatitis. METHODS Acute pancreatitis was induced in mice using three disparate models: (1) caerulein hyperstimulation, (2) ethanol plus palmitoleic acid, and (3) retrograde biliopancreatic ductal infusion of sodium taurocholate. The NAMPT inhibitor FK866 and NAMPT downstream product nicotinamide mononucleotide (NMN) was administered. Serum and pancreas were collected and analyzed biochemically and histologically. Bone marrow derived macrophages were isolated, cultured with cytokines or pancreatic acini, then analyzed by quantitative PCR and non-targeted metabolomics. RESULTS The levels of pancreatic NAMPT and NAD were down-regulated upon acute pancreatitis. NAMPT inhibitor FK866 suppressed M1 macrophage polarization while NMN boosted it. In co-culture of macrophages with acinar cells, inhibition of NAMPT prevented M1-like macrophage differentiation induced by injured pancreatic acini. The injured pancreatic acinar milieu induced a unique metabolic signature linked to macrophage polarization, and inhibition of NAMPT reversed these metabolites changes. Furthermore, NMN supplementation aggravated caerulein hyperstimulation pancreatitis and alcoholic pancreatitis, and inhibition of NAMPT protected against caerulein hyperstimulation, alcoholic and biliary acute pancreatitis and reducing pancreatic macrophage infiltration in vivo. CONCLUSIONS NAMPT inhibition protects against acute pancreatitis via preventing M1 macrophage polarization and restoring the metabolites related to macrophage polarization and that NAMPT could be a promising therapeutic target for acute pancreatitis.
Collapse
Affiliation(s)
- Yan He
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Dai
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengya Niu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Congying Chen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingjie Jiang
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zengkai Wu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingpiao Bao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuli Zhang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sohail Z Husain
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, United States
| | - Guoyong Hu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Li Wen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
16
|
Huffaker TB, Ekiz HA, Barba C, Lee SH, Runtsch MC, Nelson MC, Bauer KM, Tang WW, Mosbruger TL, Cox JE, Round JL, Voth WP, O'Connell RM. A Stat1 bound enhancer promotes Nampt expression and function within tumor associated macrophages. Nat Commun 2021; 12:2620. [PMID: 33976173 PMCID: PMC8113251 DOI: 10.1038/s41467-021-22923-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 03/30/2021] [Indexed: 02/03/2023] Open
Abstract
Tumor associated macrophage responses are regulated by distinct metabolic states that affect their function. However, the ability of specific signals in the local tumor microenvironment to program macrophage metabolism remains under investigation. Here, we identify NAMPT, the rate limiting enzyme in NAD salvage synthesis, as a target of STAT1 during cellular activation by interferon gamma, an important driver of macrophage polarization and antitumor responses. We demonstrate that STAT1 occupies a conserved element within the first intron of Nampt, termed Nampt-Regulatory Element-1 (NRE1). Through disruption of NRE1 or pharmacological inhibition, a subset of M1 genes is sensitive to NAMPT activity through its impact on glycolytic processes. scRNAseq is used to profile in vivo responses by NRE1-deficient, tumor-associated leukocytes in melanoma tumors through the creation of a unique mouse strain. Reduced Nampt and inflammatory gene expression are present in specific myeloid and APC populations; moreover, targeted ablation of NRE1 in macrophage lineages results in greater tumor burden. Finally, elevated NAMPT expression correlates with IFNγ responses and melanoma patient survival. This study identifies IFN and STAT1-inducible Nampt as an important factor that shapes the metabolic program and function of tumor associated macrophages.
Collapse
Affiliation(s)
- Thomas B Huffaker
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - H Atakan Ekiz
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Cindy Barba
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Soh-Hyun Lee
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Marah C Runtsch
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Morgan C Nelson
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Kaylyn M Bauer
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - William W Tang
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | | | - James E Cox
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
| | - June L Round
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Warren P Voth
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA.
| | - Ryan M O'Connell
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
17
|
Xu Y, Yu L, Liu Y, Tang X, Wang X. Lipopolysaccharide-Induced Microglial Neuroinflammation: Attenuation by FK866. Neurochem Res 2021; 46:1291-1304. [PMID: 33713324 DOI: 10.1007/s11064-021-03267-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/21/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
Alleviating microglia-mediated neuroinflammation bears great promise to reduce neurodegeneration. Nicotinamide phosphoribosyltransferase (NAMPT) may exert cytokine-like effect in the brain. However, it remains unclear about role of NAMPT in microglial inflammation. Also, it remains unknown about effect of NAMPT inhibition on microglial inflammation. In the present study, we observed that FK866 (a specific noncompetitive NAMPT inhibitor) dose-dependently inhibited lipopolysaccharide (LPS)-induced proinflammatory mediator (interleukin (IL)-6, IL-1β, inducible nitric oxide synthase, nitric oxide and reactive species) level increase in BV2 microglia cultures. FK866 also significantly inhibited LPS-induced polarization change in microglia. Furthermore, LPS significantly increased NAMPT expression and nuclear factor kappa B (NF-κB) phosphorylation in microglia. FK866 significantly decreased NAMPT expression and NF-κB phosphorylation in LPS-treated microglia. Finally, conditioned medium from microglia cultures co-treated with FK866 and LPS significantly increased SH-SY5Y and PC12 cell viability compared with conditioned medium from microglia cultures treated with LPS alone. Our study strongly indicates that NAMPT may be a promising target for microglia modulation and NAMPT inhibition may attenuate microglial inflammation.
Collapse
Affiliation(s)
- Yaling Xu
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Lijia Yu
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Ying Liu
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Xiaohui Tang
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Xijin Wang
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China.
| |
Collapse
|
18
|
Annie L, Gurusubramanian G, Roy VK. Changes in the localization of ovarian visfatin protein and its possible role during estrous cycle of mice. Acta Histochem 2020; 122:151630. [PMID: 32992122 DOI: 10.1016/j.acthis.2020.151630] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022]
Abstract
Visfatin is a crucial adipokine, which also regulates ovarian functions in many animals. Mice estrous cycle is characterized by a dynamic complex physiological process in the reproductive system. Expression of various factors changes during the estrous cycle in the ovary. To the best of our knowledge, no previous study has been conducted on the expression of visfatin in mice ovaries during the estrous cycle. Therefore, we investigated the localization and expression of visfatin protein in the ovary of mice during the estrous cycle. Western blot analysis showed the elevated expression of visfatin in proestrus and lowest in diestrus. Immunohistochemical localization of visfatin showed intense staining in the corpus luteum of proestrus and diestrus ovaries. Thecal cells, granulosa cells, and oocytes also showed the presence of visfatin. Expression of ovarian visfatin was correlated to BCL2 and active caspase3 expression and exhibited a significant positive correlation. Furthermore, in vivo inhibition of visfatin by FK866 in the proestrus ovary down-regulated active caspase3 and PCNA expression, and up-regulated the BCL2 expression. These results suggest the role of visfatin in the proliferation and apoptosis of the follicles and specific localization of visfatin in the corpus luteum also indicate its role in corpus luteum function, which may be in progesterone biosynthesis and regression of old corpus luteum. However, further study is required to support these findings. In conclusion, visfatin may also be regulating follicular growth during the estrous cycle by regulating proliferation and apoptosis.
Collapse
|
19
|
Liu J, Zhu T, Niu Q, Yang X, Suo H, Zhang H. Dendrobium nobile Alkaloids Protects against H 2O 2-Induced Neuronal Injury by Suppressing JAK-STATs Pathway Activation in N2A Cells. Biol Pharm Bull 2020; 43:716-724. [PMID: 32238714 DOI: 10.1248/bpb.b19-01083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to investigate the preventive effect and mechanism of Dendrobium alkaloids (DNLA) on oxidative stress-related death in neuronal cells. Our results demonstrated that DNLA has a direct neuroprotective effect through oxidative stress in N2A cells induced by hydrogen peroxide (H2O2). CCK8, lactate dehydrogenase (LDH), intracellular Ca2+, intracellular reactive oxygen species (ROS), and mitochondrial membrane potential (MMP) were used to evaluate the mechanism of DNLA neutralization by H2O2-induced injury. Results presented in the paper indicate that treatment with DNLA (35 ng/mL) significantly attenuated decreases in cell viability, release of LDH, and apoptosis after H2O2-induced neuronal injury. Furthermore, DNLA significantly reduced intracellular Ca2+ up-regulation, ROS production, and inhibited mitochondrial depolarization. Moreover, DNLA treatment significantly downregulated expressions of interleukin (IL)-1β, tumor necrosis factor (TNF)-α, IL-6, nitric oxide synthase, janus kinase-signal transducer and activators of transcription (JAK-STATs) signaling in N2A cells, all of which were H2O2-induced. Taken together, our findings suggested that DNLA may inhibit the expression of pro-inflammatory and pro-apoptotic factors by blocking JAK-STATs signaling after oxidative stress injury. This research provides a potential experimental basis for further application of DNLA to prevent various human nervous system diseases caused by oxidative stress.
Collapse
Affiliation(s)
- Jing Liu
- School of Life Science and Bioengineering, Henan University of Urban Construction
| | - Tao Zhu
- School of Life Science and Bioengineering, Henan University of Urban Construction
| | - Qingqing Niu
- School of Life Science and Bioengineering, Henan University of Urban Construction
| | - Xiaoxing Yang
- School of Life Science and Bioengineering, Henan University of Urban Construction
| | - Hao Suo
- School of Life Science and Bioengineering, Henan University of Urban Construction
| | - Hao Zhang
- School of Life Science and Bioengineering, Henan University of Urban Construction.,Institute of Biomedical and Pharmaceutical sciences, Guangdong University of Technology
| |
Collapse
|
20
|
Notoginseng Leaf Triterpenes Ameliorates OGD/R-Induced Neuronal Injury via SIRT1/2/3-Foxo3a-MnSOD/PGC-1 α Signaling Pathways Mediated by the NAMPT-NAD Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7308386. [PMID: 33149812 PMCID: PMC7603631 DOI: 10.1155/2020/7308386] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/14/2020] [Accepted: 09/25/2020] [Indexed: 12/11/2022]
Abstract
Background Cerebral ischemic stroke (CIS) is a common cerebrovascular disease whose main risks include necrosis, apoptosis, and cerebral infarction. But few therapeutic advances and prominent drugs seem to be of value for ischemic stroke in the clinic yet. In the previous study, notoginseng leaf triterpenes (PNGL) from Panax notoginseng stem and leaf have been confirmed to have neuroprotective effects against mitochondrial damages caused by cerebral ischemia in vivo. However, the potential mechanisms of mitochondrial protection have not been fully elaborated yet. Methods The oxygen and glucose deprivation and reperfusion (OGD/R)-induced SH-SY5Y cells were adopted to explore the neuroprotective effects and the potential mechanisms of PNGL in vitro. Cellular cytotoxicity was measured by MTT, viable mitochondrial staining, and antioxidant marker detection in vitro.Mitochondrial functions were analyzed by ATP content measurement, MMP determination, ROS, NAD, and NADH kit in vitro. And the inhibitor FK866 was adopted to verify the regulation of PNGL on the target NAMPT and its key downstream. Results In OGD/R models, treatment with PNGL strikingly alleviated ischemia injury, obviously preserved redox balance and excessive oxidative stress, inhibited mitochondrial damage, markedly alleviated energy metabolism dysfunction, improved neuronal mitochondrial functions, obviously reduced neuronal loss and apoptosis in vitro, and thus notedly raised neuronal survival under ischemia and hypoxia. Meanwhile, PNGL markedly increased the expression of nicotinamide phosphoribosyltransferase (NAMPT) in the ischemic regions and OGD/R-induced SH-SY5Y cells and regulated the downstream SIRT1/2-Foxo3a and SIRT1/3-MnSOD/PGC-1α pathways. And FK866 further verified that the protective effects of PNGL might be mediated by the NAMPT in vitro. Conclusions The mitochondrial protective effects of PNGL are, at least partly, mediated via the NAMPT-NAD+ and its downstream SIRT1/2/3-Foxo3a-MnSOD/PGC-1α signaling pathways. PNGL, as a new drug candidate, has a pivotal role in mitochondrial homeostasis and energy metabolism therapy via NAMPT against OGD-induced SH-SY5Y cell injury.
Collapse
|
21
|
Zou X, Xie L, Wang W, Zhao G, Tian X, Chen M. FK866 alleviates cerebral pyroptosis and inflammation mediated by Drp1 in a rat cardiopulmonary resuscitation model. Int Immunopharmacol 2020; 89:107032. [PMID: 33045576 DOI: 10.1016/j.intimp.2020.107032] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Dynamin-related protein 1 (Drp1) mediates mitochondrial fission and triggers NLRP3 inflammasome activation. FK866 (a NAMPT inhibitor) exerts a neuroprotective effect in ischemia/reperfusion injury through the suppression of mitochondrial dysfunction. We explored the effects of FK866 on pyroptosis and inflammation mediated by Drp1 in a cardiac arrest/cardiopulmonary resuscitation (CA/CPR) rat model. METHODS Healthy male Sprague-Dawley rats were subjected to 7 min CA by trans-esophageal electrical stimulation followed by CPR. The surviving rats were treated with FK866 (a selective inhibitor of NAMPT), Mdivi-1 (Drp1 inhibitor), FK866 + Mdivi-1, or vehicle and then underwent 24 h reperfusion. Hematoxylin and eosin staining and immunohistochemistry (to detect NSE) were used to evaluate brain injury. We performed immunofluorescent staining to analyze NLRP3 and GSDMD expression in microglia or astrocytes and western blot to determine expression of NLRP3, IL-1β, GSDMD, Drp1, and Mfn2. Transmission electron microscopy was used to observe mitochondria. RESULTS FK866 significantly decreased pathological damage to brain tissue, inhibited the activation of NLRP3 in microglia or astrocytes, downregulated the expression of NLRP3, IL-1β, GSDMD, p-Drp1 protein, upregulated Mfn2 and improve mitochondrial morphology. CONCLUSIONS Our results demonstrated that FK866 protects the brain against ischemia-reperfusion injury in rats after CA/CPR by inhibiting pyroptosis and inflammation mediated by Drp1.
Collapse
Affiliation(s)
- Xinsen Zou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi, China
| | - Lu Xie
- Department of Physiology, Guangxi Medical University, Nanning 530000, Guangxi, China
| | - Wenyan Wang
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi, China
| | - Gaoyang Zhao
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi, China
| | - Xinyue Tian
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi, China
| | - Menghua Chen
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi, China.
| |
Collapse
|
22
|
Ma H, Su D, Wang Q, Chong Z, Zhu Q, He W, Wang W. Phoenixin 14 inhibits ischemia/reperfusion-induced cytotoxicity in microglia. Arch Biochem Biophys 2020; 689:108411. [PMID: 32450066 DOI: 10.1016/j.abb.2020.108411] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 05/15/2020] [Accepted: 05/17/2020] [Indexed: 12/23/2022]
Abstract
The process of ischemia/reperfusion (IR) in ischemic stroke often leads to significant cell death and permanent neuronal damage. Safe and effective treatments are urgently needed to mitigate the damage caused by IR injury. The naturally occurring pleiotropic peptide phoenixin 14 (PNX-14) has recently come to light as a potential treatment for IR injury. In the present study, we examined the effects of PNX-14 on several key processes involved in ischemic injury, such as pro-inflammatory cytokine expression, oxidative stress, and the related cascade mediated through the toll-like receptor 4 (TLR4) pathway, using BV2 microglia exposed to oxygen-glucose deprivation and reoxygenation (OGD/R). Our results demonstrate an acute ability of PNX-14 to regulate the expression levels of proinflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6). PNX-14 also prevented oxidative stress by reducing the generation of reactive oxygen species (ROS) and increasing the level of the antioxidant glutathione (GSH). Importantly, PNX-14 inhibited high-mobility group box 1 (HMGB1)/TLR4/myeloid differentiation primary response 88 (MyD88)/nuclear factor-κB (NF-κB) signaling pathway, by inhibiting the activation of TLR4 and preventing the nuclear translocation of p65 protein. We further confirmed the cerebroprotective effects of PNX-14 in an MCAO rat model, which resulted in reduced infarct volume and decreased microglia activation. Together, the results of this study implicate a possible protective role of PNX-14 against various aspects of IR injury in vitro.
Collapse
Affiliation(s)
- Hongling Ma
- Department of Neurology, Liaocheng People's Hospital of Shandong First Medical University, Liaocheng City, Shandong Province, 252000, China
| | - Daoqing Su
- Department of Neurosurgery, Liaocheng People's Hospital of Shandong First Medical University, Liaocheng City, Shandong Province, 252000, China
| | - Qingdong Wang
- Department of Neurology, Liaocheng People's Hospital of Shandong First Medical University, Liaocheng City, Shandong Province, 252000, China
| | - Zonglei Chong
- Department of Neurosurgery, Liaocheng People's Hospital of Shandong First Medical University, Liaocheng City, Shandong Province, 252000, China
| | - Qiushi Zhu
- Department of Neurosurgery, Liaocheng People's Hospital of Shandong First Medical University, Liaocheng City, Shandong Province, 252000, China
| | - Weibin He
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, Hubei Province, 430060, China
| | - Wei Wang
- Department of Neurology, Liaocheng People's Hospital of Shandong First Medical University, Liaocheng City, Shandong Province, 252000, China.
| |
Collapse
|
23
|
Galli U, Colombo G, Travelli C, Tron GC, Genazzani AA, Grolla AA. Recent Advances in NAMPT Inhibitors: A Novel Immunotherapic Strategy. Front Pharmacol 2020; 11:656. [PMID: 32477131 PMCID: PMC7235340 DOI: 10.3389/fphar.2020.00656] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is a cofactor of many enzymatic reactions as well as being a substrate for a number of NAD-consuming enzymes (e.g., PARPS, sirtuins, etc). NAD can be synthesized de novo starting from tryptophan, nicotinamide, nicotinic acid, or nicotinamide riboside from the diet. On the other hand, the nicotinamide that is liberated by NAD-consuming enzymes can be salvaged to re-form NAD. In this former instance, nicotinamide phosphoribosyltransferase (NAMPT) is the bottleneck enzyme. In the many cells in which the salvage pathway is predominant, NAMPT, therefore, represents an important controller of intracellular NAD concentrations, and as a consequence of energy metabolism. It is, therefore, not surprising that NAMPT is over expressed by tumoral cells, which take advantage from this to sustain growth rate and tumor progression. This has led to the initiation of numerous medicinal chemistry programs to develop NAMPT inhibitors in the context of oncology. More recently, however, it has been shown that NAMPT inhibitors do not solely target the tumor but also have an effect on the immune system. To add complexity, this enzyme can also be secreted by cells, and in the extracellular space it acts as a cytokine mainly through the activation of Toll like Receptor 4 (TLR4), although it has not been clarified yet if this is the only receptor responsible for its actions. While specific small molecules have been developed only against the intracellular form of NAMPT, growing evidences sustain the possibility to target the extracellular form. In this contribution, the most recent evidences on the medicinal chemistry of NAMPT will be reviewed, together with the key elements that sustain the hypothesis of NAMPT targeting and the drawbacks so far encountered.
Collapse
Affiliation(s)
- Ubaldina Galli
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Giorgia Colombo
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, University of Pavia, Pavia, Italy
| | - Gian Cesare Tron
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Ambra A Grolla
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
24
|
Tan Z, Chen L, Ren Y, Jiang X, Gao W. Neuroprotective effects of FK866 against traumatic brain injury: Involvement of p38/ERK pathway. Ann Clin Transl Neurol 2020; 7:742-756. [PMID: 32302063 PMCID: PMC7261767 DOI: 10.1002/acn3.51044] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/20/2020] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE FK866 is an inhibitor of nicotinamide phosphoribosyltransferase (NAMPT), which exhibits neuroprotective effects in ischemic brain injury. However, in traumatic brain injury (TBI), the role and mechanism of FK866 remain unclear. The present research was aimed to investigate whether FK866 could attenuate TBI and clarified the underlying mechanisms. METHODS A controlled cortical impact model was established, and FK866 at a dose of 5 mg/kg was administered intraperitoneally at 1 h and 6 h, then twice per day post-TBI until sacrifice. Brain water content, Evans blue dye extravasation, modified neurological severity scores (mNSS), Morris water maze test, enzyme-linked immunosorbent assay (ELISA), immunofluorescence staining, and western blot were performed. RESULTS The results demonstrated that FK866 significantly mitigated the brain edema, blood-brain barrier (BBB) disruption, and ameliorated the neurological function post-TBI. Moreover, FK866 decreased the number of Iba-1-positive cells, GFAP-positive astrocytes, and AQP4-positive cells. FK866 reduced the protein levels of proinflammatory cytokines and inhibited NF-κB from translocation to the nucleus. FK866 upregulated the expression of Bcl-2, diminished the expression of Bax and caspase 3, and the number of apoptotic cells. Moreover, p38 MAPK and ERK activation were significantly inhibited by FK866. INTERPRETATION FK866 attenuated TBI-induced neuroinflammation and apoptosis, at least in part, through p38/ERK MAPKs signaling pathway.
Collapse
Affiliation(s)
- Zhongju Tan
- Department of GeriatricsThe First Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Lili Chen
- Department of NeurologyXiasha CampusSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Yucheng Ren
- Department of NeurosurgeryThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Xiaohang Jiang
- Department of NeurosurgeryThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Wei Gao
- Department of NeurologyChangxing People’s HospitalThe Second Affiliated Hospital of Zhejiang University Changxing CampusChangxingZhejiangChina
| |
Collapse
|
25
|
Guia RM, Hassing AS, Skov LJ, Ratner C, Plucińska K, Madsen S, Diep TA, Dela Cruz GV, Trammell SA, Sustarsic EG, Emanuelli B, Gillum MP, Gerhart‐Hines Z, Holst B, Treebak JT. Fasting- and ghrelin-induced food intake is regulated by NAMPT in the hypothalamus. Acta Physiol (Oxf) 2020; 228:e13437. [PMID: 31900990 DOI: 10.1111/apha.13437] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 11/18/2019] [Accepted: 12/29/2019] [Indexed: 12/13/2022]
Abstract
AIM Neurons in the arcuate nucleus of the hypothalamus are involved in regulation of food intake and energy expenditure, and dysregulation of signalling in these neurons promotes development of obesity. The role of the rate-limiting enzyme in the NAD+ salvage pathway, nicotinamide phosphoribosyltransferase (NAMPT), for regulation energy homeostasis by the hypothalamus has not been extensively studied. METHODS We determined whether Nampt mRNA or protein levels in the hypothalamus of mice were affected by diet-induced obesity, by fasting and re-feeding, and by leptin and ghrelin treatment. Primary hypothalamic neurons were treated with FK866, a selective inhibitor of NAMPT, or rAAV carrying shRNA directed against Nampt, and levels of reactive oxygen species (ROS) and mitochondrial respiration were assessed. Fasting and ghrelin-induced food intake was measured in mice in metabolic cages after intracerebroventricular (ICV)-mediated FK866 administration. RESULTS NAMPT levels in the hypothalamus were elevated by administration of ghrelin and leptin. In diet-induced obese mice, both protein and mRNA levels of NAMPT decreased in the hypothalamus. NAMPT inhibition in primary hypothalamic neurons significantly reduced levels of NAD+ , increased levels of ROS, and affected the expression of Agrp, Pomc and genes related to mitochondrial function. Finally, ICV-induced NAMPT inhibition by FK866 did not cause malaise or anhedonia, but completely ablated fasting- and ghrelin-induced increases in food intake. CONCLUSION Our findings indicate that regulation of NAMPT levels in hypothalamic neurons is important for the control of fasting- and ghrelin-induced food intake.
Collapse
Affiliation(s)
- Roldan M. Guia
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Anna S. Hassing
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Louise J. Skov
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Cecilia Ratner
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Kaja Plucińska
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Søren Madsen
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Thi A. Diep
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Gelo V. Dela Cruz
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Stem Cell Biology University of Copenhagen Copenhagen Denmark
| | - Samuel A.J. Trammell
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Elahu G. Sustarsic
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Brice Emanuelli
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Matthew P. Gillum
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Zach Gerhart‐Hines
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Birgitte Holst
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Jonas T. Treebak
- Faculty of Health and Medical Sciences Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| |
Collapse
|
26
|
Tang Y, Liu HL, Min LX, Yuan HS, Guo L, Han PB, Lu YX, Zhong JF, Wang DL. Serum and cerebrospinal fluid tau protein level as biomarkers for evaluating acute spinal cord injury severity and motor function outcome. Neural Regen Res 2019; 14:896-902. [PMID: 30688276 PMCID: PMC6375043 DOI: 10.4103/1673-5374.249238] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/28/2018] [Indexed: 12/19/2022] Open
Abstract
Tau protein, a microtubule-associated protein, has a high specific expression in neurons and axons. Because traumatic spinal cord injury mainly affects neurons and axons, we speculated that tau protein may be a promising biomarker to reflect the degree of spinal cord injury and prognosis of motor function. In this study, 160 female Sprague-Dawley rats were randomly divided into a sham group, and mild, moderate, and severe spinal cord injury groups. A laminectomy was performed at the T8 level to expose the spinal cord in all groups. A contusion lesion was made with the NYU-MASCIS impactor by dropping a 10 g rod from heights of 12.5 mm (mild), 25 mm (moderate) and 50 mm (severe) upon the exposed dorsal surface of the spinal cord. Tau protein levels were measured in serum and cerebrospinal fluid samples at 1, 6, 12, 24 hours, 3, 7, 14 and 28 days after operation. Locomotor function of all rats was assessed using the Basso, Beattie and Bresnahan locomotor rating scale. Tau protein concentration in the three spinal cord injury groups (both in serum and cerebrospinal fluid) rapidly increased and peaked at 12 hours after spinal cord injury. Statistically significant positive linear correlations were found between tau protein level and spinal cord injury severity in the three spinal cord injury groups, and between the tau protein level and Basso, Beattie, and Bresnahan locomotor rating scale scores. The tau protein level at 12 hours in the three spinal cord injury groups was negatively correlated with Basso, Beattie, and Bresnahan locomotor rating scale scores at 28 days (serum: r = -0.94; cerebrospinal fluid: r = -0.95). Our data suggest that tau protein levels in serum and cerebrospinal fluid might be a promising biomarker for predicting the severity and functional outcome of traumatic spinal cord injury.
Collapse
Affiliation(s)
- Ying Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, China
| | - Hong-Liang Liu
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ling-Xia Min
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hao-Shi Yuan
- Department of Spine Surgery, Second Affiliated Hospital of Xi’an Medical University, Xi’an, Shanxi Province, China
| | - Lei Guo
- Department of Orthopedics, Chinese PLA Beijing Army General Hospital, Beijing, China
| | - Peng-Bo Han
- Department of Spine Surgery, Second Affiliated Hospital of Xi’an Medical University, Xi’an, Shanxi Province, China
| | - Yu-Xin Lu
- Department of Spine Surgery, Second Affiliated Hospital of Xi’an Medical University, Xi’an, Shanxi Province, China
| | - Jian-Feng Zhong
- Department of Spine Surgery, Second Affiliated Hospital of Xi’an Medical University, Xi’an, Shanxi Province, China
| | - Dong-Lin Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
27
|
Lu N, Li X, Tan R, An J, Cai Z, Hu X, Wang F, Wang H, Lu C, Lu H. HIF-1α/Beclin1-Mediated Autophagy Is Involved in Neuroprotection Induced by Hypoxic Preconditioning. J Mol Neurosci 2018; 66:238-250. [PMID: 30203298 PMCID: PMC6182618 DOI: 10.1007/s12031-018-1162-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/20/2018] [Indexed: 02/07/2023]
Abstract
Hypoxic preconditioning (HPC) exerts a protective effect against hypoxic/ischemic brain injury, and one mechanism explaining this effect may involve the upregulation of hypoxia-inducible factor-1 (HIF-1). Autophagy, an endogenous protective mechanism against hypoxic/ischemic injury, is correlated with the activation of the HIF-1α/Beclin1 signaling pathway. Based on previous studies, we hypothesize that the protective role of HPC may involve autophagy occurring via activation of the HIF-1α/Beclin1 signaling pathway. To test this hypothesis, we evaluated the effects of HPC on oxygen-glucose deprivation/reperfusion (OGD/R)-induced apoptosis and autophagy in SH-SY5Y cells. HPC significantly attenuated OGD/R-induced apoptosis, and this effect was suppressed by the autophagy inhibitor 3-methyladenine and mimicked by the autophagy agonist rapamycin. In control SH-SY5Y cells, HPC upregulated the expression of HIF-1α and downstream molecules such as BNIP3 and Beclin1. Additionally, HPC increased the LC3-II/LC3-I ratio and decreased p62 levels. The increase in the LC3-II/LC3-I ratio was inhibited by the HIF-1α inhibitor YC-1 or by Beclin1-short hairpin RNA (shRNA). In OGD/R-treated SH-SY5Y cells, HPC also upregulated the expression levels of HIF-1α, BNIP3, and Beclin1, as well as the LC3-II/LC3-I ratio. Furthermore, YC-1 or Beclin1-shRNA attenuated the HPC-mediated cell viability in OGD/R-treated cells. Taken together, our results demonstrate that HPC protects SH-SY5Y cells against OGD/R via HIF-1α/Beclin1-regulated autophagy.
Collapse
Affiliation(s)
- Na Lu
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
- Key Laboratory for the Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Xingxing Li
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Ruolan Tan
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Jing An
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Zhenlu Cai
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Xiaoxuan Hu
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Feidi Wang
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Haoruo Wang
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Chengbiao Lu
- Key Laboratory for the Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Haixia Lu
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
28
|
Gao J, Xiong B, Zhang B, Li S, Huang N, Zhan G, Jiang R, Yang L, Wu Y, Miao L, Zhu B, Yang C, Luo A. Sulforaphane Alleviates Lipopolysaccharide-induced Spatial Learning and Memory Dysfunction in Mice: The Role of BDNF-mTOR Signaling Pathway. Neuroscience 2018; 388:357-366. [PMID: 30086367 DOI: 10.1016/j.neuroscience.2018.07.052] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/29/2018] [Accepted: 07/30/2018] [Indexed: 12/12/2022]
Abstract
Peripheral immune activation could cause neuroinflammation, leading to a series of central nervous system (CNS) disorders, such as spatial learning and memory dysfunction. However, its pathogenic mechanism and therapeutic strategies are not yet determined. The present study aimed to investigate the therapeutic effects of sulforaphane (SFN) on lipopolysaccharide (LPS)-induced spatial learning and memory dysfunction, and tried to elucidate its relationship with the role of hippocampal brain-derived neurotrophic factor (BDNF)-mammalian target of rapamycin (mTOR) signaling pathway. Intraperitoneal injection of LPS for consecutive 7 days to mice caused abnormal behaviors in Morris water maze test (MWMT), while systemic administration of SFN notably reversed the abnormal behaviors. In addition, hippocampal levels of inflammatory cytokines, synaptic proteins, BDNF-tropomyosin receptor kinase B (TrkB) and mTOR signaling pathways were altered in the processes of LPS-induced cognitive dysfunction and SFN's therapeutic effects. Furthermore, we found that ANA-12 (a TrkB inhibitor) or rapamycin (a mTOR inhibitor) could block the beneficial effects of SFN on LPS-induced cognitive dysfunction, and that hippocampal levels of synaptic proteins, BDNF-TrkB and mTOR signaling pathways were also notably changed. In conclusion, the results of the present study suggest that SFN could elicit improving effects on LPS-induced spatial learning and memory dysfunction, which is likely related to the regulation of hippocampal BDNF-mTOR signaling pathway.
Collapse
Affiliation(s)
- Jie Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingrui Xiong
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Niannian Huang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaofeng Zhan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Riyue Jiang
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ling Yang
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yeshun Wu
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Liying Miao
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Bin Zhu
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Chun Yang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ailin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
29
|
Travelli C, Colombo G, Mola S, Genazzani AA, Porta C. NAMPT: A pleiotropic modulator of monocytes and macrophages. Pharmacol Res 2018; 135:25-36. [PMID: 30031171 DOI: 10.1016/j.phrs.2018.06.022] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 06/20/2018] [Indexed: 12/11/2022]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is the bottleneck enzyme of the NAD salvage pathway and thereby is a controller of intracellular NAD concentrations. It has been long known that the same enzyme can be secreted by a number of cell types and acts as a cytokine, although its receptor is at present unknown. Investigational compounds have been developed that target the enzymatic activity as well as the extracellular action (i.e. neutralizing antibodies). The present contribution reviews the evidence that links intracellular and extracellular NAMPT to myeloid biology, for example governing monocyte/macrophage differentiation, polarization and migration. Furthermore, it reviews the evidence that links this protein to some disorders in which myeloid cells have a prominent role (acute infarct, inflammatory bowel disease, acute lung injury and rheumatoid arthritis) and the data showing that inhibition of the enzymatic activity or the neutralization of the cytokine is beneficial in preclinical animal models.
Collapse
Affiliation(s)
- Cristina Travelli
- Department of Pharmacological Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Giorgia Colombo
- Department of Pharmacological Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Silvia Mola
- Department of Pharmacological Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Armando A Genazzani
- Department of Pharmacological Sciences, Università del Piemonte Orientale, Novara, Italy.
| | - Chiara Porta
- Department of Pharmacological Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
30
|
Jiang M, Liu X, Zhang D, Wang Y, Hu X, Xu F, Jin M, Cao F, Xu L. Celastrol treatment protects against acute ischemic stroke-induced brain injury by promoting an IL-33/ST2 axis-mediated microglia/macrophage M2 polarization. J Neuroinflammation 2018. [PMID: 29540209 PMCID: PMC5853059 DOI: 10.1186/s12974-018-1124-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Acute ischemic stroke (AIS) is the most common type of cerebrovascular disease and is a leading cause of disability and death worldwide. Recently, a study suggested that transformation of microglia from the pro-inflammatory M1 state to the anti-inflammatory and tissue-reparative M2 phenotype may be an effective therapeutic strategy for ischemic stroke. Celastrol, a traditional oriental medicine, may have anti-inflammatory and neuroprotective effects. However, the underlying mechanisms remain unknown. Methods We first determined the expression levels of inflammatory factors in patients and rodent models associated with AIS; we then determined the anti-inflammatory effects of celastrol in AIS, both in vivo and in vitro, using animal models of middle cerebral artery occlusion (MCAO) and cell models of oxygen-glucose deprivation (OGD) treatment with or without celastrol, respectively. Results The results indicated that expression of both inflammatory (interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α) cytokines, as well as the anti-inflammatory cytokine, IL-33, and IL-10, were increased following AIS in patients and in animal models. Furthermore, in vitro experiments confirmed that celastrol treatment decreased inflammatory cytokine expression induced by OGD through an IL-33/ST2 axis-mediated M2 microglia/macrophage polarization. Finally, celastrol is protected against ischemic-induced nerve injury, both in vivo and in vitro. Conclusions Taken together, these data suggest that celastrol post-treatment reduces ischemic stroke-induced brain damage, suggesting celastrol may represent a novel potent pharmacological therapy.
Collapse
Affiliation(s)
- Mei Jiang
- Department of neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Xinghui Liu
- Department of Clinical Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, 207 Ju Ye Road, Pudong New Area, Shanghai, 200135, People's Republic of China
| | - Denghai Zhang
- Sino-French Cooperative Central Lab, Shanghai Gongli Hospital, The Second Military Medical University, 207 Ju Ye Road, Pudong New District, Shanghai, 200135, People's Republic of China
| | - Ying Wang
- Sino-French Cooperative Central Lab, Shanghai Gongli Hospital, The Second Military Medical University, 207 Ju Ye Road, Pudong New District, Shanghai, 200135, People's Republic of China
| | - Xiaoxia Hu
- Department of Clinical Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, 207 Ju Ye Road, Pudong New Area, Shanghai, 200135, People's Republic of China
| | - Fengxia Xu
- Department of Clinical Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, 207 Ju Ye Road, Pudong New Area, Shanghai, 200135, People's Republic of China
| | - Mingming Jin
- Department of Clinical Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, 207 Ju Ye Road, Pudong New Area, Shanghai, 200135, People's Republic of China.
| | - Fanfan Cao
- Sino-French Cooperative Central Lab, Shanghai Gongli Hospital, The Second Military Medical University, 207 Ju Ye Road, Pudong New District, Shanghai, 200135, People's Republic of China.
| | - Limin Xu
- Department of Clinical Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, 207 Ju Ye Road, Pudong New Area, Shanghai, 200135, People's Republic of China.
| |
Collapse
|