1
|
Kim S, Hong HS. Substance P alleviates liver fibrosis by modulating inflammation and mobilizing reparative stem cells. Int Immunopharmacol 2024; 142:113211. [PMID: 39321699 DOI: 10.1016/j.intimp.2024.113211] [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: 05/12/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
Repetitive hepatic damage resulting from viral hepatitis, toxins, and alcohol abuse induces chronic inflammation and excessive accumulation of the extracellular matrix, leading to the development of liver cirrhosis. Substance P (SP) promotes endogenous wound healing by mobilizing bone marrow stem cells and stimulating anti-inflammatory responses. This study aimed to investigate whether SP exerts a therapeutic effect on liver fibrosis by recruiting endogenous stem cells and modulating immune responses. A non-clinical model of liver cirrhosis was established through repeated injections of thioacetamide and recombinant leptin. After confirming liver fibrosis, SP was administered intravenously for 6 weeks. SP treatment decreased the formation of hepatic micronodules on the external surface of the liver and the infiltration of immune cells. Furthermore, SP treatment notably reduced the deposition of collagen and the activation of hepatic stellate cells, concomitant with decreased levels of transforming growth factor-β1 and matrix metalloproteinase activity. In the context of severe hepatic damage, SP increased the number of circulating stem cells, leading to the restoration of the reparative stem cell pool in the bone marrow. The findings of this study suggest that SP alleviates liver fibrosis by modulating the mobilization of functional stem cells and the immune response.
Collapse
Affiliation(s)
- Suna Kim
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Deokyoung dae-ro, 1732, Yong In 17104, Republic of Korea; Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee dae-ro 23, Hoegi-dong, Seoul 02447, Republic of Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Kyung Hee dae-ro, 24, Seoul 02461, Republic of Korea; East-West Medical Research Institute, Kyung Hee University, Kyung Hee dae-ro, 24, Hoegi-dong, Seoul 02461, Republic of Korea; Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee dae-ro 23, Hoegi-dong, Seoul 02447, Republic of Korea.
| |
Collapse
|
2
|
Beeraka NM, Basappa B, Nikolenko VN, Mahesh PA. Role of Neurotransmitters in Steady State Hematopoiesis, Aging, and Leukemia. Stem Cell Rev Rep 2024:10.1007/s12015-024-10761-z. [PMID: 38976142 DOI: 10.1007/s12015-024-10761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
Haematopoiesis within the bone marrow (BM) represents a complex and dynamic process intricately regulated by neural signaling pathways. This delicate orchestration is susceptible to disruption by factors such as aging, diabetes, and obesity, which can impair the BM niche and consequently affect haematopoiesis. Genetic mutations in Tet2, Dnmt3a, Asxl1, and Jak2 are known to give rise to clonal haematopoiesis of intermediate potential (CHIP), a condition linked to age-related haematological malignancies. Despite these insights, the exact roles of circadian rhythms, sphingosine-1-phosphate (S1P), stromal cell-derived factor-1 (SDF-1), sterile inflammation, and the complement cascade on various BM niche cells remain inadequately understood. Further research is needed to elucidate how BM niche cells contribute to these malignancies through neural regulation and their potential in the development of gene-corrected stem cells. This literature review describes the updated functional aspects of BM niche cells in haematopoiesis within the context of haematological malignancies, with a particular focus on neural signaling and the potential of radiomitigators in acute radiation syndrome. Additionally, it underscores the pressing need for technological advancements in stem cell-based therapies to alleviate the impacts of immunological stressors. Recent studies have illuminated the microheterogeneity and temporal stochasticity of niche cells within the BM during haematopoiesis, emphasizing the updated roles of neural signaling and immunosurveillance. The development of gene-corrected stem cells capable of producing blood, immune cells, and tissue-resident progeny is essential for combating age-related haematological malignancies and overcoming immunological challenges. This review aims to provide a comprehensive overview of these evolving insights and their implications for future therapeutic strategies.
Collapse
Affiliation(s)
- Narasimha M Beeraka
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN, 46202, USA.
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russia.
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, 515721, India.
| | - Basappa Basappa
- Department of Studies in Organic Chemistry, Laboratory of Chemical Biology, University of Mysore, Mysore, Karnataka, 570006, India
| | - Vladimir N Nikolenko
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russia
| | - P A Mahesh
- Department of Pulmonary Medicine, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| |
Collapse
|
3
|
Wang DC, Xu WD, Tang YY, Yang C, Li R, Wu GC, Huang AF. Neuropeptide Y, a potential marker for lupus, promotes lupus development. Int Immunopharmacol 2024; 126:111272. [PMID: 38006754 DOI: 10.1016/j.intimp.2023.111272] [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: 09/22/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 11/27/2023]
Abstract
OBJECTIVE Relationship between neuropeptide Y (NPY) serum levels, NPY genetic mutation with systemic lupus erythematosus (SLE) pathogenesis is yet to be clarified, and role of NPY in development of SLE needs elucidation. METHOD This study included 460 SLE patients, 472 non-SLE cases, 500 healthy volunteers. Serum NPY, matrix metalloproteinase-1 (MMP-1) and MMP-8 levels were tested by ELISA. Genotyping 7 NPY single nucleotides polymorphisms (SNPs) (rs5573, rs5574, rs16129, rs16138, rs16140, rs16147, rs16478) was obtained by Kompetitive Allele-Specific PCR (KASP) method. Pristane-induced lupus mice were treated with NPY-Y1 receptor antagonist, and histological analysis, serological changes of the mice were evaluated. RESULTS NPY serum concentrations were significantly increased in SLE patients when compared to that in healthy volunteers, non-SLE cases. Rs5573 G allele, rs16129 T allele, rs16147 G allele frequencies were significantly different between SLE cases and healthy controls. Rs5574 TT + TC genotypes were related to levels of IgG, C3, C4 and erythrocyte sedimentation rate, and rs16138 GG + GC genotypes correlated with SLE cases with anti-double-stranded deoxyribonucleic acid antibody (anti-dsDNA) (+). Serum MMP-1, MMP-8 concentrations were higher in SLE patients, and NPY levels were significantly related to MMP-1, MMP-8 levels. After treatment of lupus mice with NPY-Y1 receptor antagonist, damage of liver, spleen and kidney was alleviated, production of autoantibodies (anti-nuclear antibody (ANA), total IgG, anti-dsDNA) and MMP-1, MMP-8 was down-regulated, and differentiation of CD3+, CD8+ T cells, B cells, monocytes, macrophages, T helper 1 (Th1), Th2, Th17 cells was reversed. CONCLUSION NPY may be a biomarker for lupus, which may promote occurrence and development of lupus.
Collapse
Affiliation(s)
- Da-Cheng Wang
- Department of Evidence-Based Medicine, Southwest Medical University, 1 Xianglin Road, Luzhou, Sichuan 646000, China
| | - Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, 1 Xianglin Road, Luzhou, Sichuan 646000, China
| | - Yang-Yang Tang
- Department of Evidence-Based Medicine, Southwest Medical University, 1 Xianglin Road, Luzhou, Sichuan 646000, China
| | - Chan Yang
- Department of Evidence-Based Medicine, Southwest Medical University, 1 Xianglin Road, Luzhou, Sichuan 646000, China
| | - Rong Li
- Department of Evidence-Based Medicine, Southwest Medical University, 1 Xianglin Road, Luzhou, Sichuan 646000, China
| | - Guo-Cui Wu
- School of Nursing, Anhui Medical University, 15 Feicui Road, Hefei, Anhui 230601, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, Sichuan 646000, China.
| |
Collapse
|
4
|
Zheng XQ, Huang J, Lin JL, Song CL. Pathophysiological mechanism of acute bone loss after fracture. J Adv Res 2023; 49:63-80. [PMID: 36115662 PMCID: PMC10334135 DOI: 10.1016/j.jare.2022.08.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 07/29/2022] [Accepted: 08/31/2022] [Indexed: 10/14/2022] Open
Abstract
BACKGROUND Acute bone loss after fracture is associated with various effects on the complete recovery process and a risk of secondary fractures among patients. Studies have reported similarities in pathophysiological mechanisms involved in acute bone loss after fractures and osteoporosis. However, given the silence nature of bone loss and bone metabolism complexities, the actual underlying pathophysiological mechanisms have yet to be fully elucidated. AIM OF REVIEW To elaborate the latest findings in basic research with a focus on acute bone loss after fracture. To briefly highlight potential therapeutic targets and current representative drugs. To arouse researchers' attention and discussion on acute bone loss after fracture. KEY SCIENTIFIC CONCEPTS OF REVIEW Bone loss after fracture is associated with immobilization, mechanical unloading, blood supply damage, sympathetic nerve regulation, and crosstalk between musculoskeletals among other factors. Current treatment strategies rely on regulation of osteoblasts and osteoclasts, therefore, there is a need to elucidate on the underlying mechanisms of acute bone loss after fractures to inform the development of efficacious and safe drugs. In addition, attention should be paid towards ensuring long-term skeletal health.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jie Huang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jia-Liang Lin
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
| |
Collapse
|
5
|
Choi BJ, Park MH, Park KH, Han WH, Yoon HJ, Jung HY, Hong JY, Chowdhury MR, Kim KY, Lee J, Song IS, Pang M, Choi MK, Gulbins E, Reichel M, Kornhuber J, Hong CW, Kim C, Kim SH, Schuchman EH, Jin HK, Bae JS. Immunotherapy targeting plasma ASM is protective in a mouse model of Alzheimer's disease. Nat Commun 2023; 14:1631. [PMID: 36959217 PMCID: PMC10036484 DOI: 10.1038/s41467-023-37316-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 03/10/2023] [Indexed: 03/25/2023] Open
Abstract
Acid sphingomyelinase (ASM) has been implicated in neurodegenerative disease pathology, including Alzheimer's disease (AD). However, the specific role of plasma ASM in promoting these pathologies is poorly understood. Herein, we explore plasma ASM as a circulating factor that accelerates neuropathological features in AD by exposing young APP/PS1 mice to the blood of mice overexpressing ASM, through parabiotic surgery. Elevated plasma ASM was found to enhance several neuropathological features in the young APP/PS1 mice by mediating the differentiation of blood-derived, pathogenic Th17 cells. Antibody-based immunotherapy targeting plasma ASM showed efficient inhibition of ASM activity in the blood of APP/PS1 mice and, interestingly, led to prophylactic effects on neuropathological features by suppressing pathogenic Th17 cells. Our data reveals insights into the potential pathogenic mechanisms underlying AD and highlights ASM-targeting immunotherapy as a potential strategy for further investigation.
Collapse
Affiliation(s)
- Byung Jo Choi
- KNU Alzheimer's disease Research Institute, Kyungpook National University, Daegu, South Korea
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, South Korea
| | - Min Hee Park
- KNU Alzheimer's disease Research Institute, Kyungpook National University, Daegu, South Korea
- Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Kang Ho Park
- KNU Alzheimer's disease Research Institute, Kyungpook National University, Daegu, South Korea
- Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Wan Hui Han
- KNU Alzheimer's disease Research Institute, Kyungpook National University, Daegu, South Korea
- Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Hee Ji Yoon
- KNU Alzheimer's disease Research Institute, Kyungpook National University, Daegu, South Korea
- Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Hye Yoon Jung
- KNU Alzheimer's disease Research Institute, Kyungpook National University, Daegu, South Korea
- Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Ju Yeon Hong
- KNU Alzheimer's disease Research Institute, Kyungpook National University, Daegu, South Korea
- Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Md Riad Chowdhury
- KNU Alzheimer's disease Research Institute, Kyungpook National University, Daegu, South Korea
- Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Kyung Yeol Kim
- KNU Alzheimer's disease Research Institute, Kyungpook National University, Daegu, South Korea
- Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Jihoon Lee
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Im-Sook Song
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Minyeong Pang
- College of Pharmacy, Dankook University, Cheon-an, South Korea
| | - Min-Koo Choi
- College of Pharmacy, Dankook University, Cheon-an, South Korea
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Martin Reichel
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Chang-Won Hong
- Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Changho Kim
- Department of Emergency Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Seung Hyun Kim
- Department of Neurology, Hanyang University College of Medicine, Seoul, South Korea
| | - Edward H Schuchman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hee Kyung Jin
- KNU Alzheimer's disease Research Institute, Kyungpook National University, Daegu, South Korea.
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, South Korea.
| | - Jae-Sung Bae
- KNU Alzheimer's disease Research Institute, Kyungpook National University, Daegu, South Korea.
- Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
6
|
Royal Jelly Increases Hematopoietic Stem Cells in Peripheral Blood: A Double-Blind, Placebo-Controlled, Randomized Trial in Healthy Subjects. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:7665515. [PMID: 36686976 PMCID: PMC9859695 DOI: 10.1155/2023/7665515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023]
Abstract
Objectives Royal jelly (RJ), produced by honeybees, influences stem cell functions, such as pluripotency maintenance of mouse embryonic stem cells and prevention of aging-related muscle stem cell functional deterioration. Thus, we hypothesized that RJ administration has various health-promoting effects based on stem cells. However, its effects are unknown in humans. In this study, we have attempted for the first time to clarify whether the administration of RJ in humans affects stem cells. Materials and Methods This randomized, double-blind, placebo-controlled study was performed on healthy subjects (n = 90) who received protease-treated RJ at a dose of 1200 mg/day or placebo daily for four weeks. Also, the participants with a low number of hematopoietic stem cells (HSCs) in peripheral blood were preferentially selected. HSC counts, endothelial progenitor cell (EPC) counts, blood cell counts in peripheral blood, cytokines in serum, and physical conditions were evaluated. Results and Conclusion. Eligible data from 86 subjects (placebo: 42, RJ: 44) who completed the study were analyzed. There were no significant differences between the two groups regarding the changes in peripheral HSC count (p=0.103), while diastolic blood pressure showed a significant improvement in the RJ group compared to that in the placebo group (p=0.032). The subgroup analysis excluded 14 subjects who complained of cold symptoms at baseline or within five days of the four-week study. The changes in the HSC populations were significantly higher in the RJ group than those in the placebo group (p=0.042). No adverse effects were observed in any of the groups. These results suggest that RJ administration affected the peripheral HSC count and may influence stem cell functions. Further research is needed to reveal the various health-promoting benefits of RJ based on stem cells.
Collapse
|
7
|
Liu Y, Gu R, Gao M, Wei Y, Shi Y, Wang X, Gu Y, Gu X, Zhang H. Emerging role of substance and energy metabolism associated with neuroendocrine regulation in tumor cells. Front Endocrinol (Lausanne) 2023; 14:1126271. [PMID: 37051193 PMCID: PMC10084767 DOI: 10.3389/fendo.2023.1126271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/07/2023] [Indexed: 03/29/2023] Open
Abstract
Cancer is the second most common cause of mortality in the world. One of the unresolved difficult pathological mechanism issues in malignant tumors is the imbalance of substance and energy metabolism of tumor cells. Cells maintain life through energy metabolism, and normal cells provide energy through mitochondrial oxidative phosphorylation to generate ATP, while tumor cells demonstrate different energy metabolism. Neuroendocrine control is crucial for tumor cells' consumption of nutrients and energy. As a result, better combinatorial therapeutic approaches will be made possible by knowing the neuroendocrine regulating mechanism of how the neuroendocrine system can fuel cellular metabolism. Here, the basics of metabolic remodeling in tumor cells for nutrients and metabolites are presented, showing how the neuroendocrine system regulates substance and energy metabolic pathways to satisfy tumor cell proliferation and survival requirements. In this context, targeting neuroendocrine regulatory pathways in tumor cell metabolism can beneficially enhance or temper tumor cell metabolism and serve as promising alternatives to available treatments.
Collapse
Affiliation(s)
- Yingying Liu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Renjun Gu
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Murong Gao
- Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yangwa Wei
- Department of Hepatobiliary Surgery, Hainan Provincial People’s Hospital, Haikou, China
| | - Yu Shi
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xu Wang
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yihuang Gu
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
- The Second Hospital of Nanjing, Nanjing, China
- *Correspondence: Hongru Zhang, ; Xin Gu, ; Yihuang Gu,
| | - Xin Gu
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Hongru Zhang, ; Xin Gu, ; Yihuang Gu,
| | - Hongru Zhang
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Hongru Zhang, ; Xin Gu, ; Yihuang Gu,
| |
Collapse
|
8
|
Li F, Xie W, Chen Z, Zhou Z, Wang Z, Xiao J, Li Z. Neuropeptide Y and receptors are associated with the pyroptosis of nucleus pulposus in aging and degenerative intervertebral discs of rats. Neuropeptides 2022; 96:102284. [PMID: 36027700 DOI: 10.1016/j.npep.2022.102284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 01/05/2023]
Abstract
The neuropeptide Y(NPY) mediates bone metabolism and the degradation of cartilage in the peripheral nervous system. However, its role in the intervertebral disc degeneration (IDD) is less clear and warrant further study. The process of IDD has always been accompanied by inflammatory response and pyroptosis of nucleus pulposus cells (NPCs). The aim of this study was to investigate the relationship between NPY, Y1R, Y2R and pyroptosis in aging and degenerative discs and the direct effect of NPY on NPCs. First, we have assessed NPY, Y1R, Y2R and the expression of pyroptosis related protein in the immature (6 weeks), mature (16 weeks), aged (54 weeks), and degenerated discs. As part of our studies, we also have evaluated pyroptotic changes in the NPCs, induced by exposure to NPY. Our results suggested that compared with natural aging discs, the degenerative discs showed the high expression of NPY, Y1R and Y2R. Correlation analysis showed that the level of NPY and Y1R in degenerative discs were positively correlated with GSDMD, whereas there was no significant correlation between Y2R and GSDMD. In vitro, NPY treatment stimulated the activation of caspase-1-dependent pyroptosis of NPCs. However, Y1R antagonist inhibited NPY-induced pyroptosis of NPCs. Western blot confirmed that Y1R antagonist decreased the level of cleaved.GSDMD and caspase-1 in NPCs. In conclusion, our results indicated that compared with natural aging discs, the degenerated discs showed the high expression of NPY, Y1R and Y2R. NPY-Y1R involve the IDD development by the regulation of pyroptosis in the NPCs. Regulating the function of NPY may be a promising strategy for IDD treatment.
Collapse
Affiliation(s)
- Fan Li
- Department of Orthopaedic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Weixin Xie
- Department of Orthopaedic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Zhijie Chen
- Department of Orthopaedic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Zhi Zhou
- Department of Orthopaedic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Zhenwei Wang
- Department of clinical laboratory, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Jie Xiao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China.
| | - Zhanchun Li
- Department of Orthopaedic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China.
| |
Collapse
|
9
|
Lv X, Gao F, Li TP, Xue P, Wang X, Wan M, Hu B, Chen H, Jain A, Shao Z, Cao X. Skeleton interoception regulates bone and fat metabolism through hypothalamic neuroendocrine NPY. eLife 2021; 10:e70324. [PMID: 34468315 PMCID: PMC8439655 DOI: 10.7554/elife.70324] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 05/21/2021] [Indexed: 01/04/2023] Open
Abstract
The central nervous system regulates activity of peripheral organs through interoception. In our previous study, we have demonstrated that PGE2/EP4 skeleton interception regulate bone homeostasis. Here, we show that ascending skeleton interoceptive signaling downregulates expression of hypothalamic neuropeptide Y (NPY) and induce lipolysis of adipose tissue for osteoblastic bone formation. Specifically, the ascending skeleton interoceptive signaling induces expression of small heterodimer partner-interacting leucine zipper protein (SMILE) in the hypothalamus. SMILE binds to pCREB as a transcriptional heterodimer on Npy promoters to inhibit NPY expression. Knockout of EP4 in sensory nerve increases expression of NPY causing bone catabolism and fat anabolism. Importantly, inhibition of NPY Y1 receptor (Y1R) accelerated oxidation of free fatty acids in osteoblasts and rescued bone loss in AvilCre:Ptger4fl/fl mice. Thus, downregulation of hypothalamic NPY expression lipolyzes free fatty acids for anabolic bone formation through a neuroendocrine descending interoceptive regulation.
Collapse
Affiliation(s)
- Xiao Lv
- Department of Orthopaedic Surgery, Institute of Cell Engineering, and Department of Biomedical Engineering, The Johns Hopkins UniversityBaltimoreUnited States
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Feng Gao
- Department of Orthopaedic Surgery, Institute of Cell Engineering, and Department of Biomedical Engineering, The Johns Hopkins UniversityBaltimoreUnited States
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Tuo Peter Li
- Department of Orthopaedic Surgery, Institute of Cell Engineering, and Department of Biomedical Engineering, The Johns Hopkins UniversityBaltimoreUnited States
| | - Peng Xue
- Department of Orthopaedic Surgery, Institute of Cell Engineering, and Department of Biomedical Engineering, The Johns Hopkins UniversityBaltimoreUnited States
| | - Xiao Wang
- Department of Orthopaedic Surgery, Institute of Cell Engineering, and Department of Biomedical Engineering, The Johns Hopkins UniversityBaltimoreUnited States
| | - Mei Wan
- Department of Orthopaedic Surgery, Institute of Cell Engineering, and Department of Biomedical Engineering, The Johns Hopkins UniversityBaltimoreUnited States
| | - Bo Hu
- Department of Orthopaedic Surgery, Institute of Cell Engineering, and Department of Biomedical Engineering, The Johns Hopkins UniversityBaltimoreUnited States
| | - Hao Chen
- Department of Orthopaedic Surgery, Institute of Cell Engineering, and Department of Biomedical Engineering, The Johns Hopkins UniversityBaltimoreUnited States
| | - Amit Jain
- Department of Orthopaedic Surgery, Institute of Cell Engineering, and Department of Biomedical Engineering, The Johns Hopkins UniversityBaltimoreUnited States
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Xu Cao
- Department of Orthopaedic Surgery, Institute of Cell Engineering, and Department of Biomedical Engineering, The Johns Hopkins UniversityBaltimoreUnited States
| |
Collapse
|
10
|
Zhang Y, Liu CY, Chen WC, Shi YC, Wang CM, Lin S, He HF. Regulation of neuropeptide Y in body microenvironments and its potential application in therapies: a review. Cell Biosci 2021; 11:151. [PMID: 34344469 PMCID: PMC8330085 DOI: 10.1186/s13578-021-00657-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/12/2021] [Indexed: 12/26/2022] Open
Abstract
Neuropeptide Y (NPY), one of the most abundant neuropeptides in the body, is widely expressed in the central and peripheral nervous systems and acts on the cardiovascular, digestive, endocrine, and nervous systems. NPY affects the nutritional and inflammatory microenvironments through its interaction with immune cells, brain-derived trophic factor (BDNF), and angiogenesis promotion to maintain body homeostasis. Additionally, NPY has great potential for therapeutic applications against various diseases, especially as an adjuvant therapy for stem cells. In this review, we discuss the research progress regarding NPY, as well as the current evidence for the regulation of NPY in each microenvironment, and provide prospects for further research on related diseases.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China
| | - Chu-Yun Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China
| | - Wei-Can Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China
| | - Yan-Chuan Shi
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Cong-Mei Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China
| | - Shu Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China. .,Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia. .,Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| | - He-Fan He
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China.
| |
Collapse
|
11
|
Endocrine role of bone in the regulation of energy metabolism. Bone Res 2021; 9:25. [PMID: 34016950 PMCID: PMC8137703 DOI: 10.1038/s41413-021-00142-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 12/20/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Bone mainly functions as a supportive framework for the whole body and is the major regulator of calcium homeostasis and hematopoietic function. Recently, an increasing number of studies have characterized the significance of bone as an endocrine organ, suggesting that bone-derived factors regulate local bone metabolism and metabolic functions. In addition, these factors can regulate global energy homeostasis by altering insulin sensitivity, feeding behavior, and adipocyte commitment. These findings may provide a new pathological mechanism for related metabolic diseases or be used in the diagnosis, treatment, and prevention of metabolic diseases such as osteoporosis, obesity, and diabetes mellitus. In this review, we summarize the regulatory effect of bone and bone-derived factors on energy metabolism and discuss directions for future research.
Collapse
|
12
|
Dietrich P, Wormser L, Fritz V, Seitz T, De Maria M, Schambony A, Kremer AE, Günther C, Itzel T, Thasler WE, Teufel A, Trebicka J, Hartmann A, Neurath MF, von Hörsten S, Bosserhoff AK, Hellerbrand C. Molecular crosstalk between Y5 receptor and neuropeptide Y drives liver cancer. J Clin Invest 2021; 130:2509-2526. [PMID: 31999643 DOI: 10.1172/jci131919] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/23/2020] [Indexed: 12/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is clearly age-related and represents one of the deadliest cancer types worldwide. As a result of globally increasing risk factors including metabolic disorders, the incidence rates of HCC are still rising. However, the molecular hallmarks of HCC remain poorly understood. Neuropeptide Y (NPY) and NPY receptors represent a highly conserved, stress-activated system involved in diverse cancer-related hallmarks including aging and metabolic alterations, but its impact on liver cancer had been unclear. Here, we observed increased expression of NPY5 receptor (Y5R) in HCC, which correlated with tumor growth and survival. Furthermore, we found that its ligand NPY was secreted by peritumorous hepatocytes. Hepatocyte-derived NPY promoted HCC progression by Y5R activation. TGF-β1 was identified as a regulator of NPY in hepatocytes and induced Y5R in invasive cancer cells. Moreover, NPY conversion by dipeptidylpeptidase 4 (DPP4) augmented Y5R activation and function in liver cancer. The TGF-β/NPY/Y5R axis and DPP4 represent attractive therapeutic targets for controlling liver cancer progression.
Collapse
Affiliation(s)
- Peter Dietrich
- Institute of Biochemistry, Emil-Fischer-Zentrum.,Department of Medicine 1, University Hospital Erlangen, and
| | | | | | | | - Monica De Maria
- Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Alexandra Schambony
- Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | | | | - Timo Itzel
- Department of Internal Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Andreas Teufel
- Department of Internal Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jonel Trebicka
- Department of Medicine I, University Hospital Bonn, Bonn, Germany
| | - Arndt Hartmann
- Institute of Pathology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center Erlangen-European Metropolitan Region of Nuremberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University Hospital Erlangen, and.,Comprehensive Cancer Center Erlangen-European Metropolitan Region of Nuremberg, Erlangen, Germany
| | - Stephan von Hörsten
- Department of Experimental Therapy, Franz Penzoldt Center, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Anja K Bosserhoff
- Institute of Biochemistry, Emil-Fischer-Zentrum.,Comprehensive Cancer Center Erlangen-European Metropolitan Region of Nuremberg, Erlangen, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Emil-Fischer-Zentrum.,Comprehensive Cancer Center Erlangen-European Metropolitan Region of Nuremberg, Erlangen, Germany
| |
Collapse
|
13
|
Lin ST, Li YZ, Sun XQ, Chen QQ, Huang SF, Lin S, Cai SQ. Update on the Role of Neuropeptide Y and Other Related Factors in Breast Cancer and Osteoporosis. Front Endocrinol (Lausanne) 2021; 12:705499. [PMID: 34421823 PMCID: PMC8377469 DOI: 10.3389/fendo.2021.705499] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/19/2021] [Indexed: 01/19/2023] Open
Abstract
Breast cancer and osteoporosis are common diseases that affect the survival and quality of life in postmenopausal women. Women with breast cancer are more likely to develop osteoporosis than women without breast cancer due to certain factors that can affect both diseases simultaneously. For instance, estrogen and the receptor activator of nuclear factor-κB ligand (RANKL) play important roles in the occurrence and development of these two diseases. Moreover, chemotherapy and hormone therapy administered to breast cancer patients also increase the incidence of osteoporosis, and in recent years, neuropeptide Y (NPY) has also been found to impact breast cancer and osteoporosis.Y1 and Y5 receptors are highly expressed in breast cancer, and Y1 and Y2 receptors affect osteogenic response, thus potentially highlighting a potential new direction for treatment strategies. In this paper, the relationship between breast cancer and osteoporosis, the influence of NPY on both diseases, and the recent progress in the research and treatment of these diseases are reviewed.
Collapse
Affiliation(s)
- Shu-ting Lin
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yi-zhong Li
- Department of Bone, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xiao-qi Sun
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qian-qian Chen
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shun-fa Huang
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW, Australia
- *Correspondence: Si-qing Cai, ; Shu Lin,
| | - Si-qing Cai
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- *Correspondence: Si-qing Cai, ; Shu Lin,
| |
Collapse
|
14
|
Wu JQ, Jiang N, Yu B. Mechanisms of action of neuropeptide Y on stem cells and its potential applications in orthopaedic disorders. World J Stem Cells 2020; 12:986-1000. [PMID: 33033559 PMCID: PMC7524693 DOI: 10.4252/wjsc.v12.i9.986] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/25/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023] Open
Abstract
Musculoskeletal disorders are the leading causes of disability and result in reduced quality of life. The neuro-osteogenic network is one of the most promising fields in orthopaedic research. Neuropeptide Y (NPY) system has been reported to be involved in the regulations of bone metabolism and homeostasis, which also provide feedback to the central NPY system via NPY receptors. Currently, potential roles of peripheral NPY in bone metabolism remain unclear. Growing evidence suggests that NPY can regulate biological actions of bone marrow mesenchymal stem cells, hematopoietic stem cells, endothelial cells, and chondrocytes via a local autocrine or paracrine manner by different NPY receptors. The regulative activities of NPY may be achieved through the plasticity of NPY receptors, and interactions among the targeted cells as well. In general, NPY can influence proliferation, apoptosis, differentiation, migration, mobilization, and cytokine secretion of different types of cells, and play crucial roles in the development of bone delayed/non-union, osteoporosis, and osteoarthritis. Further basic research should clarify detailed mechanisms of action of NPY on stem cells, and clinical investigations are also necessary to comprehensively evaluate potential applications of NPY and its receptor-targeted drugs in management of musculoskeletal disorders.
Collapse
Affiliation(s)
- Jian-Qun Wu
- Department of Orthopedics and Traumatology, Huadu District People’s Hospital, Guangzhou 510800, Guangdong Province, China
| | - Nan Jiang
- Division of Orthopaedics and Traumatology, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
15
|
Wu W, Peng S, Shi Y, Li L, Song Z, Lin S. NPY promotes macrophage migration by upregulating matrix metalloproteinase-8 expression. J Cell Physiol 2020; 236:1903-1912. [PMID: 32710469 DOI: 10.1002/jcp.29973] [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: 03/06/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 01/03/2023]
Abstract
Macrophage migration is thought to participate in obesity-related cardiovascular diseases. Matrix metalloproteinase-8 (MMP-8) possesses proteolytic activity on the extracellular matrix (ECM), which promotes macrophage migration to the site of vascular injury. Neuropeptide Y (NPY) is a bioactive peptide involved in MMP expression. However, it is uncertain whether NPY can regulate the expression of matrix metalloproteinase-8 (MMP-8) in macrophages. In this study, wild-type C57BL/6 and NPY-/- mice were fed a high-fat diet and subjected to subcutaneous carotid artery injury with ferric chloride, to observe the role of NPY and macrophages in neointima formation. In addition, Raw264.7 cells were treated with NPY and its antagonists to observe MMP-8 expression and macrophage migration. We found that NPY-/- mice exhibited significantly reduced neointima formation after carotid artery injury. The content of macrophages and MMP-8 in the neointima and media were also significantly reduced in NPY-/- mice compared with C57BL/6 mice. Moreover, the expression of MMP-8 in macrophages was also decreased in NPY-/- mice. NPY increased MMP-8 messenger RNA and protein expression in Raw264.7 cells in vitro, and this effect was abrogated by the Y1R antagonist. In addition, NPY increased the phosphorylation of ERK1/2, which was significantly attenuated by co-treatment with the Y1R antagonist. Moreover, NPY-induced MMP-8 expression could be decreased by the ERK1/2 inhibitor PD98059. Furthermore, NPY promoted macrophage migration across type I collagen in vitro. In conclusion, NPY promotes macrophage migration by upregulating MMP-8 expression, which we believe to be an underappreciated mechanism of the increased progression of neointima formation.
Collapse
Affiliation(s)
- Weiqiang Wu
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Song Peng
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yanchuan Shi
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia.,Faculty of Medicine, St Vincent's Clinical School, UNSW Australia, NSW, Australia
| | - Linyu Li
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhiyuan Song
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shu Lin
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia
| |
Collapse
|
16
|
N-AS-triggered SPMs are direct regulators of microglia in a model of Alzheimer's disease. Nat Commun 2020; 11:2358. [PMID: 32398649 PMCID: PMC7217877 DOI: 10.1038/s41467-020-16080-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 04/09/2020] [Indexed: 12/11/2022] Open
Abstract
Sphingosine kinase1 (SphK1) is an acetyl-CoA dependent acetyltransferase which acts on cyclooxygenase2 (COX2) in neurons in a model of Alzheimer’s disease (AD). However, the mechanism underlying this activity was unexplored. Here we show that N-acetyl sphingosine (N-AS) is first generated by acetyl-CoA and sphingosine through SphK1. N-AS then acetylates serine 565 (S565) of COX2, and the N-AS-acetylated COX2 induces the production of specialized pro-resolving mediators (SPMs). In a mouse model of AD, microglia show a reduction in N-AS generation, leading to decreased acetyl-S565 COX2 and SPM production. Treatment with N-AS increases acetylated COX2 and N-AS-triggered SPMs in microglia of AD mice, leading to resolution of neuroinflammation, an increase in microglial phagocytosis, and improved memory. Taken together, these results identify a role of N-AS in the dysfunction of microglia in AD. Neuronal sphingosine kinase 1 (SphK1) acetylates COX2 which is needed for microglial phagocytosis activity, and release of pro-resolving mediators (SPMs) from neurons. Here the authors examine how SphK1-mediates COX2 acetylation, and how this leads to increased secretion of SPMs from neurons in the context of Alzheimer’s disease models.
Collapse
|
17
|
Chen R, Hao Z, Chen X, Fu Q, Ma Y. Neuropeptide Y enhances proliferation and chondrogenic differentiation of ATDC5 cells. Neuropeptides 2020; 80:102022. [PMID: 31987472 DOI: 10.1016/j.npep.2020.102022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/19/2020] [Accepted: 01/19/2020] [Indexed: 12/19/2022]
Abstract
In recent years, emerging evidence has illustrated the indispensable role of sympathetic neurotransmitters and their receptors in cartilage mediation. The presence of neuropeptide Y (NPY)-positive sympathetic nerve fibres in cartilage and NPY-secretion function in chondrocytes raises the possibility of NPY directly regulating the function of chondrocytes. Therefore, this study intended to evaluate the effect of NPY and its receptors on the proliferation and chondrogenic differentiation of ATDC5 cells. Results showed NPY, especially at a concentration of 10-10 M, to significantly enhance proliferation of ATDC5 cells. Moreover, NPY effectively facilitated early chondrogenesis and late hypertrophy/mineralisation of ATDC5 cells via Y1 receptor signalling, rather than via Y2 receptor signalling. Taken together, the results help us to understand how NPY and its receptors affect the function of chondrocytes.
Collapse
Affiliation(s)
- Ruixin Chen
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Zhichao Hao
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xiaodan Chen
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Qiang Fu
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China.
| | - Yuanyuan Ma
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China.
| |
Collapse
|
18
|
Ulum B, Mammadova A, Özyüncü Ö, Uçkan-Çetinkaya D, Yanık T, Aerts-Kaya F. Neuropeptide Y is involved in the regulation of quiescence of hematopoietic stem cells. Neuropeptides 2020; 80:102029. [PMID: 32127176 DOI: 10.1016/j.npep.2020.102029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 12/18/2022]
Abstract
Differentiation, self-renewal and quiescence of Hematopoietic stem cells (HSCs) is tightly regulated in order to protect the HSCs from the strain of constant cell division and depletion of the stem cell pool. The neurotransmitter Neuropeptide Y (NPY) is released from sympathetic nerves in the bone marrow and has been shown to indirectly affect HSC function through effects on bone marrow (BM) multipotent Mesenchymal Stromal Cells (MSCs), osteoblasts (OBs) and macrophages. Although the absence of NPY has been shown to be accompanied by severe BM impairment and delayed engraftment of HSCs, the direct effects of NPY on HSCs have never been assessed. Here, we aimed to explore the effect of NPY on the regulation of HSCs. All NPY receptors Y1, Y2, Y4 and Y5 were found to be highly expressed on most HSCs and mature hematopoietic cell subsets. In culture, in particularly expression of the Y1 receptor was shown to decrease in time. Doses of 300 nM NPY suppressed HSC proliferation in cell cultures, as confirmed by an increase of HSCs in G0 phase and an increase in the gene expression levels of FOXO3, DICER1, SMARCA2 and PDK1, which all have been shown to play an important role in the regulation of cell quiescence. These data support the idea that NPY may have a direct effect on the regulation of HSC fate by modulating cell quiescence.
Collapse
Affiliation(s)
- Baris Ulum
- Hacettepe University Center for Stem Cell Research, Ankara, Turkey; Middle East Technical University, Department of Biological Sciences, Ankara, Turkey
| | - Aynura Mammadova
- Hacettepe University Center for Stem Cell Research, Ankara, Turkey; Hacettepe University Graduate School of Health Sciences, Department of Stem Cell Sciences, Ankara, Turkey
| | - Özgür Özyüncü
- Hacettepe University Medical Faculty, Department of Obstetrics and Gynecology, Ankara, Turkey
| | - Duygu Uçkan-Çetinkaya
- Hacettepe University Center for Stem Cell Research, Ankara, Turkey; Hacettepe University Graduate School of Health Sciences, Department of Stem Cell Sciences, Ankara, Turkey
| | - Tülin Yanık
- Middle East Technical University, Department of Biological Sciences, Ankara, Turkey
| | - Fatima Aerts-Kaya
- Hacettepe University Center for Stem Cell Research, Ankara, Turkey; Hacettepe University Graduate School of Health Sciences, Department of Stem Cell Sciences, Ankara, Turkey.
| |
Collapse
|
19
|
Aerts-Kaya F, Ulum B, Mammadova A, Köse S, Aydin G, Korkusuz P, Uçkan-Çetinkaya D. Neurological Regulation of the Bone Marrow Niche. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1212:127-153. [PMID: 31342461 DOI: 10.1007/5584_2019_398] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The bone marrow (BM) hematopoietic niche is the microenvironment where in the adult hematopoietic stem and progenitor cells (HSPCs) are maintained and regulated. This regulation is tightly controlled through direct cell-cell interactions with mesenchymal stromal stem (MSCs) and reticular cells, adipocytes, osteoblasts and endothelial cells, through binding to extracellular matrix molecules and through signaling by cytokines and hematopoietic growth factors. These interactions provide a healthy environment and secure the maintenance of the HSPC pool, their proliferation, differentiation and migration. Recent studies have shown that innervation of the BM and interactions with the peripheral sympathetic neural system are important for maintenance of the hematopoietic niche, through direct interactions with HSCPs or via interactions with other cells of the HSPC microenvironment. Signaling through adrenergic receptors (ARs), opioid receptors (ORs), endocannabinoid receptors (CRs) on HSPCs and MSCs has been shown to play an important role in HSPC homeostasis and mobilization. In addition, a wide range of neuropeptides and neurotransmitters, such as Neuropeptide Y (NPY), Substance P (SP) and Tachykinins, as well as neurotrophins and neuropoietic growth factors have been shown to be involved in regulation of the hematopoietic niche. Here, a comprehensive overview is given of their role and interactions with important cells in the hematopoietic niche, including HSPCs and MSCs, and their effect on HSPC maintenance, regulation and mobilization.
Collapse
Affiliation(s)
- Fatima Aerts-Kaya
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Ankara, Turkey. .,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey.
| | - Baris Ulum
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey.,Faculty of Arts and Sciences, Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Aynura Mammadova
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Ankara, Turkey.,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Sevil Köse
- Faculty of Health Sciences, Department of Medical Biology, Atilim University, Ankara, Turkey
| | - Gözde Aydin
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Ankara, Turkey.,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Petek Korkusuz
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Ankara, Turkey.,Faculty of Medicine, Department of Histology and Embryology, Hacettepe University, Ankara, Turkey
| | - Duygu Uçkan-Çetinkaya
- Graduate School of Health Sciences, Department of Stem Cell Sciences, Hacettepe University, Ankara, Turkey.,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| |
Collapse
|
20
|
Abstract
Hypertension is an important risk factor for cardiovascular morbidity and mortality and for events such as myocardial infarction, stroke, heart failure and chronic kidney disease and is a major determinant of disability-adjusted life-years. Despite the importance of hypertension, the pathogenesis of essential hypertension, which involves the complex interaction of several mechanisms, is still poorly understood. Evidence suggests that interplay between bone marrow, microglia and immune mediators underlies the development of arterial hypertension, in particular through mechanisms involving cytokines and peptides, such as neuropeptide Y, substance P, angiotensin II and angiotensin-(1-7). Chronic psychological stress also seems to have a role in increasing the risk of hypertension, probably through the activation of neuroimmune pathways. In this Review, we summarize the available data on the possible role of neuroimmune crosstalk in the origin and maintenance of arterial hypertension and discuss the implications of this crosstalk for recovery and rehabilitation after cardiac and cerebral injuries.
Collapse
|
21
|
Leitão L, Alves CJ, Sousa DM, Neto E, Conceição F, Lamghari M. The alliance between nerve fibers and stem cell populations in bone marrow: life partners in sickness and health. FASEB J 2019; 33:8697-8710. [PMID: 31017803 DOI: 10.1096/fj.201900454r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The bone marrow (BM) is the central hematopoietic organ in adult mammals, with great potential to be used as a tool to improve the efficacy of the body's response to a number of malignancies and stressful conditions. The nervous system emerges as a critical regulatory player of the BM both under homeostatic and pathologic settings, with essential roles in cellular anchorage and egress, stem cell differentiation, and endothelial cell permeability. This review collects the current knowledge on the interplay between the nervous system and the BM cell populations, with a focus on how the nervous system modulates hematopoietic stem and progenitor cell, mesenchymal stromal cell, and endothelial progenitor cell activity in BM. We have also highlighted the pathologies that have been associated with disturbances in the neuronal signaling in BM and discussed if targeting the nervous system, either by modulating the activity of specific neuronal circuits or by pharmacologically leveling the activity of sympathetic and sensorial signaling-responsive cells in BM, is a promising therapeutic approach to tackling pathologies from BM origin.-Leitão, L., Alves, C. J., Sousa, D. M., Neto, E., Conceição, F., Lamghari, M. The alliance between nerve fibers and stem cell populations in bone marrow: life partners in sickness and health.
Collapse
Affiliation(s)
- Luís Leitão
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Cecília J Alves
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Daniela M Sousa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Estrela Neto
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Francisco Conceição
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Meriem Lamghari
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
22
|
de Kruijf EJFM, Fibbe WE, van Pel M. Cytokine-induced hematopoietic stem and progenitor cell mobilization: unraveling interactions between stem cells and their niche. Ann N Y Acad Sci 2019; 1466:24-38. [PMID: 31006885 PMCID: PMC7217176 DOI: 10.1111/nyas.14059] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/15/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023]
Abstract
Peripheral blood hematopoietic stem and progenitor cells (HSPCs), mobilized by granulocyte colony‐stimulating factor, are widely used as a source for both autologous and allogeneic stem cell transplantation. The use of mobilized HSPCs has several advantages over traditional bone marrow–derived HSPCs, including a less invasive harvesting process for the donor, higher HSPC yields, and faster hematopoietic reconstitution in the recipient. For years, the mechanisms by which cytokines and other agents mobilize HSPCs from the bone marrow were not fully understood. The field of stem cell mobilization research has advanced significantly over the past decade, with major breakthroughs in the elucidation of the complex mechanisms that underlie stem cell mobilization. In this review, we provide an overview of the events that underlie HSPC mobilization and address the relevant cellular and molecular components of the bone marrow niche. Furthermore, current and future mobilizing agents will be discussed.
Collapse
Affiliation(s)
- Evert-Jan F M de Kruijf
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Willem E Fibbe
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Melissa van Pel
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
23
|
Wee NKY, Sinder BP, Novak S, Wang X, Stoddard C, Matthews BG, Kalajzic I. Skeletal phenotype of the neuropeptide Y knockout mouse. Neuropeptides 2019; 73:78-88. [PMID: 30522780 PMCID: PMC6326877 DOI: 10.1016/j.npep.2018.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/04/2018] [Accepted: 11/28/2018] [Indexed: 10/27/2022]
Abstract
Neuropeptide Y (NPY) is involved in multiple processes such as behavior, energy and bone metabolism. Previous studies have relied on global NPY depletion to examine its effects on bone. However, this approach is unable to distinguish the central or local source of NPY influencing bone. Our aim was to identify which cells within the skeleton express Npy and establish a model that will enable us to differentiate effects of NPY derived from different cell types. We have generated the NPY floxed (NPYflox) mice using CRISPR technology. By crossing the NPYflox mice with Hypoxanthine Phosphoribosyltransferase 1 (Hprt)-cre to generate a global knockout, we were able to validate and confirm loss of Npy transcript and protein in our global NPYKO. Global deletion of NPY results in a smaller femoral cortical cross-sectional area (-12%) and reduced bone strength (-18%) in male mice. In vitro, NPY-deficient bone marrow stromal cells (BMSCs) showed increase in osteogenic differentiation detected by increases in alkaline phosphatase staining and bone sialoprotein and osteocalcin expression. Despite both sexes presenting with increased adiposity, female mice had no alterations in bone mass, suggesting that NPY may have sex-specific effects on bone. In this study we identified Npy expression in the skeleton and examined the effect of global NPY depletion to bone mass. The differential impact of NPY deletion in cortical and cancellous compartments along with differences in phenotypes between in vitro and in vivo, highlights the complex nature of NPY signaling, indicative of distinct sources that can be dissected in the future using this NPYflox model.
Collapse
Affiliation(s)
- Natalie K Y Wee
- Department of Reconstructive Sciences, Farmington, CT 06030, USA
| | | | - Sanja Novak
- Department of Reconstructive Sciences, Farmington, CT 06030, USA
| | - Xi Wang
- Department of Reconstructive Sciences, Farmington, CT 06030, USA
| | - Chris Stoddard
- Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Brya G Matthews
- Department of Reconstructive Sciences, Farmington, CT 06030, USA; Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, Farmington, CT 06030, USA.
| |
Collapse
|
24
|
Abstract
The interplay between the immune and nervous systems has been acknowledged in the past, but only more recent studies have started to unravel the cellular and molecular players of such interactions. Mounting evidence indicates that environmental signals are sensed by discrete neuro-immune cell units (NICUs), which represent defined anatomical locations in which immune and neuronal cells colocalize and functionally interact to steer tissue physiology and protection. These units have now been described in multiple tissues throughout the body, including lymphoid organs, adipose tissue, and mucosal barriers. As such, NICUs are emerging as important orchestrators of multiple physiological processes, including hematopoiesis, organogenesis, inflammation, tissue repair, and thermogenesis. In this review we focus on the impact of NICUs in tissue physiology and how this fast-evolving field is driving a paradigm shift in our understanding of immunoregulation and organismal physiology.
Collapse
Affiliation(s)
- Cristina Godinho-Silva
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal; , ,
| | - Filipa Cardoso
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal; , ,
| | | |
Collapse
|
25
|
Park MH, Baek B, Jin HK, Bae JS. Novel peptides derived from neuropeptide Y prevent chemotherapy-induced bone marrow damage by regulating hematopoietic stem cell microenvironment. Anim Cells Syst (Seoul) 2018; 22:281-288. [PMID: 30460109 PMCID: PMC6171453 DOI: 10.1080/19768354.2018.1517826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/13/2018] [Accepted: 08/19/2018] [Indexed: 10/30/2022] Open
Abstract
Chemotherapy-induced bone marrow damage is accompanied by acute nerve injury in the bone marrow (BM), resulting in sensory and autonomic neuropathy. Cisplatin, a popular chemotherapy drugs, induces the impairment of hematopoietic stem cells (HSCs) and bone marrow regeneration, leading to chronic bone marrow abnormalities. Previously, we reported the protective roles of neuropeptide Y (NPY) against cisplatin-induced bone marrow impairment. In this study, we identified novel peptides, generated from full-length NPY that rescued cisplatin-induced sensory neuropathy and HSC suppression by regulating cell survival in the BM microenvironment. One of these peptides, especially, showed a better protective property against these impairments compared to that seen in full-length NPY. Therefore, we suggest the NPY sequences most effective against the chemotherapy-induced bone marrow dysfunction that could be potentially useful as therapeutic agents for patients receiving chemotherapy.
Collapse
Affiliation(s)
- Min Hee Park
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu, South Korea.,Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea.,Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, South Korea
| | - Bosung Baek
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu, South Korea.,Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, South Korea
| | - Hee Kyung Jin
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu, South Korea.,Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, South Korea
| | - Jae-Sung Bae
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu, South Korea.,Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea.,Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
26
|
Abstract
Bones provide both skeletal scaffolding and space for hematopoiesis in its marrow. Previous work has shown that these functions were tightly regulated by the nervous system. The central and peripheral nervous systems tightly regulate compact bone remodeling, its metabolism, and hematopoietic homeostasis in the bone marrow (BM). Accumulating evidence indicates that the nervous system, which fine-tunes inflammatory responses and alterations in neural functions, may regulate autoimmune diseases. Neural signals also influence the progression of hematological malignancies such as acute and chronic myeloid leukemias. Here, we review the interplay of the nervous system with bone, BM, and immunity, and discuss future challenges to target hematological diseases through modulation of activity of the nervous system.
Collapse
Affiliation(s)
- Maria Maryanovich
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Shoichiro Takeishi
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
27
|
Park MH, Jung IK, Min WK, Choi JH, Kim GM, Jin HK, Bae JS. Neuropeptide Y improves cisplatin-induced bone marrow dysfunction without blocking chemotherapeutic efficacy in a cancer mouse model. BMB Rep 2018; 50:417-422. [PMID: 28712386 PMCID: PMC5595171 DOI: 10.5483/bmbrep.2017.50.8.099] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Indexed: 11/25/2022] Open
Abstract
Cisplatin is the most effective and widely used chemo-therapeutic agent for many types of cancer. Unfortunately, its clinical use is limited by its adverse effects, notably bone marrow suppression leading to abnormal hematopoiesis. We previously revealed that neuropeptide Y (NPY) is responsible for the maintenance of hematopoietic stem cell (HSC) function by protecting the sympathetic nervous system (SNS) fibers survival from chemotherapy-induced bone marrow impairment. Here, we show the NPY-mediated protective effect against bone marrow dysfunction due to cisplatin in an ovarian cancer mouse model. During chemotherapy, NPY mitigates reduction in HSC abundance and destruction of SNS fibers in the bone marrow without blocking the anticancer efficacy of cisplatin, and it results in the restoration of blood cells and amelioration of sensory neuropathy. Therefore, these results suggest that NPY can be used as a potentially effective agent to improve bone marrow dysfunction during cisplatin-based cancer therapy.
Collapse
Affiliation(s)
- Min Hee Park
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu 41566; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| | - In Kyung Jung
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu 41566; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| | - Woo-Kie Min
- Department of Orthopaedic Surgery, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Jin Ho Choi
- Department of Mechanical Engineering, Gumi University, Gumi 39213, Korea
| | - Gyu Man Kim
- School of Mechanical Engineering, Kyungpook National University, Daegu 41566, Korea
| | - Hee Kyung Jin
- Stem Cell Neuroplasticity Research Group and Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Koreaa
| | - Jae-Sung Bae
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu 41566; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
28
|
Do Neuroendocrine Peptides and Their Receptors Qualify as Novel Therapeutic Targets in Osteoarthritis? Int J Mol Sci 2018; 19:ijms19020367. [PMID: 29373492 PMCID: PMC5855589 DOI: 10.3390/ijms19020367] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 01/15/2023] Open
Abstract
Joint tissues like synovium, articular cartilage, meniscus and subchondral bone, are targets for neuropeptides. Resident cells of these tissues express receptors for various neuroendocrine-derived peptides including proopiomelanocortin (POMC)-derived peptides, i.e., α-melanocyte-stimulating hormone (α-MSH), adrenocorticotropin (ACTH) and β-endorphin (β-ED), and sympathetic neuropeptides like vasoactive intestinal peptide (VIP) and neuropeptide y (NPY). Melanocortins attained particular attention due to their immunomodulatory and anti-inflammatory effects in several tissues and organs. In particular, α-MSH, ACTH and specific melanocortin-receptor (MCR) agonists appear to have promising anti-inflammatory actions demonstrated in animal models of experimentally induced arthritis and osteoarthritis (OA). Sympathetic neuropeptides have obtained increasing attention as they have crucial trophic effects that are critical for joint tissue and bone homeostasis. VIP and NPY are implicated in direct and indirect activation of several anabolic signaling pathways in bone and synovial cells. Additionally, pituitary adenylate cyclase-activating polypeptide (PACAP) proved to be chondroprotective and, thus, might be a novel target in OA. Taken together, it appears more and more likely that the anabolic effects of these neuroendocrine peptides or their respective receptor agonists/antagonists may be exploited for the treatment of patients with inflammatory and degenerative joint diseases in the future.
Collapse
|
29
|
Singh P, Hoggatt J, Kamocka MM, Mohammad KS, Saunders MR, Li H, Speth J, Carlesso N, Guise TA, Pelus LM. Neuropeptide Y regulates a vascular gateway for hematopoietic stem and progenitor cells. J Clin Invest 2017; 127:4527-4540. [PMID: 29130940 DOI: 10.1172/jci94687] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/12/2017] [Indexed: 12/11/2022] Open
Abstract
Endothelial cells (ECs) are components of the hematopoietic microenvironment and regulate hematopoietic stem and progenitor cell (HSPC) homeostasis. Cytokine treatments that cause HSPC trafficking to peripheral blood are associated with an increase in dipeptidylpeptidase 4/CD26 (DPP4/CD26), an enzyme that truncates the neurotransmitter neuropeptide Y (NPY). Here, we show that enzymatically altered NPY signaling in ECs caused reduced VE-cadherin and CD31 expression along EC junctions, resulting in increased vascular permeability and HSPC egress. Moreover, selective NPY2 and NPY5 receptor antagonists restored vascular integrity and limited HSPC mobilization, demonstrating that the enzymatically controlled vascular gateway specifically opens by cleavage of NPY by CD26 signaling via NPY2 and NPY5 receptors. Mice lacking CD26 or NPY exhibited impaired HSPC trafficking that was restored by treatment with truncated NPY. Thus, our results point to ECs as gatekeepers of HSPC trafficking and identify a CD26-mediated NPY axis that has potential as a pharmacologic target to regulate hematopoietic trafficking in homeostatic and stress conditions.
Collapse
Affiliation(s)
- Pratibha Singh
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jonathan Hoggatt
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Cancer Center and Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Mary R Saunders
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hongge Li
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jennifer Speth
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nadia Carlesso
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Hematologic Malignancies Translational Science, Beckman Research Institute of City of Hope, Duarte, California, USA
| | | | - Louis M Pelus
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
30
|
Effects of Neuropeptide Y on Stem Cells and Their Potential Applications in Disease Therapy. Stem Cells Int 2017; 2017:6823917. [PMID: 29109742 PMCID: PMC5646323 DOI: 10.1155/2017/6823917] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/01/2017] [Accepted: 08/08/2017] [Indexed: 01/04/2023] Open
Abstract
Neuropeptide Y (NPY), a 36-amino acid peptide, is widely distributed in the central and peripheral nervous systems and other peripheral tissues. It takes part in regulating various biological processes including food intake, circadian rhythm, energy metabolism, and neuroendocrine secretion. Increasing evidence indicates that NPY exerts multiple regulatory effects on stem cells. As a kind of primitive and undifferentiated cells, stem cells have the therapeutic potential to replace damaged cells, secret paracrine molecules, promote angiogenesis, and modulate immunity. Stem cell-based therapy has been demonstrated effective and considered as one of the most promising treatments for specific diseases. However, several limitations still hamper its application, such as poor survival and low differentiation and integration rates of transplanted stem cells. The regulatory effects of NPY on stem cell survival, proliferation, and differentiation may be helpful to overcome these limitations and facilitate the application of stem cell-based therapy. In this review, we summarized the regulatory effects of NPY on stem cells and discussed their potential applications in disease therapy.
Collapse
|
31
|
Park MH, Kim N, Jin HK, Bae JS. Neuropeptide Y-based recombinant peptides ameliorate bone loss in mice by regulating hematopoietic stem/progenitor cell mobilization. BMB Rep 2017; 50:138-143. [PMID: 27998395 PMCID: PMC5422026 DOI: 10.5483/bmbrep.2017.50.3.191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Indexed: 12/30/2022] Open
Abstract
Ovariectomy-induced bone loss is related to an increased deposition of osteoclasts on bone surfaces. We reported that the 36-amino-acid-long neuropeptide Y (NPY) could mobilize hematopoietic stem/progenitor cells (HSPCs) from the bone marrow to the peripheral blood by regulating HSPC maintenance factors and that mobilization of HSPCs ameliorated low bone density in an ovariectomy-induced osteoporosis mouse model by reducing the number of osteoclasts. Here, we demonstrated that new NPY peptides, recombined from the cleavage of the full-length NPY, showed better functionality for HSPC mobilization than the full-length peptide. These recombinant peptides mediated HSPC mobilization with greater efficiency by decreasing HSPC maintenance factors. Furthermore, treatment with these peptides reduced the number of osteoclasts and relieved ovariectomy-induced bone loss in mice more effectively than treatment with full-length NPY. Therefore, these results suggest that peptides recombined from full-length NPY can be used to treat osteoporosis.
Collapse
Affiliation(s)
- Min Hee Park
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu 41566; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| | - Namoh Kim
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu 41566; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| | - Hee Kyung Jin
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu 41566; Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Jae-Sung Bae
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu 41566; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
32
|
Jung WC, Levesque JP, Ruitenberg MJ. It takes nerve to fight back: The significance of neural innervation of the bone marrow and spleen for immune function. Semin Cell Dev Biol 2017; 61:60-70. [DOI: 10.1016/j.semcdb.2016.08.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 01/17/2023]
|
33
|
Cellular players of hematopoietic stem cell mobilization in the bone marrow niche. Int J Hematol 2016; 105:129-140. [PMID: 27943116 DOI: 10.1007/s12185-016-2162-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 11/21/2016] [Accepted: 11/29/2016] [Indexed: 12/23/2022]
Abstract
Hematopoietic stem cells (HSC) reside in perivascular regions of the bone marrow (BM) embedded within a complex regulatory unit called the niche. Cellular components of HSC niches include vascular endothelial cells, mesenchymal stromal progenitor cells and a variety of mature hematopoietic cells such as macrophages, neutrophils, and megakaryocytes-further regulated by sympathetic nerves and complement components as described in this review. Three decades ago the discovery that cytokines induce a large number of HSC to mobilize from the BM into the blood where they are easily harvested, revolutionised the field of HSC transplantation-curative for immune-deficiencies and some malignancies. However, despite now routine use of granulocyte-colony stimulating factor (G-CSF) to mobilise HSC for transplant, only in last 15 years has research on the mechanisms behind why and how HSC can be induced to move into the blood began. These studies have revealed the complexity of the niche that retains HSC in the BM. This review describes how BM niches and HSC themselves change during administration of G-CSF-or in the recovery phase of chemotherapy-to facilitate movement of HSC into the blood, and research now leading to development of novel therapeutics to further boost HSC mobilization and transplant success.
Collapse
|