1
|
Aberkane FZ, Engler P, Boisard S, Benarbia MEA, Guilet D. A new perspective in avian bone health: dietary supplementation with a standardized dry grape extract improves pullets' bones' quality through metabolic modulation. Poult Sci 2025; 104:105270. [PMID: 40418877 DOI: 10.1016/j.psj.2025.105270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 05/05/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
In laying hens, maintaining optimal bone health and development from the early stages is crucial, as it directly affects their egg-laying efficiency, overall welfare and productivity. Studies have shown that grape polyphenols from Vitis vinifera enhance bone health in both humans and animals. Still, the mechanisms behind this effect remain unclear, given the diversity of grape polyphenols and their varying mechanisms of action. The objective of this study was to investigate the effect of a standardized dry grape extract (SDGE) on bone quality and metabolome of future laying hens reared in commercial conditions. Therefore, 36300 day-old pullets were randomly divided into two barns on the same farm site. Both received the same diet, with the addition of 30 mg/kg of SDGE (Nor-Grape®, Nor-Feed, France) from day 1 until week 17 in the supplemented group (SDGE). At the end of the supplementation period, several bone quality parameters were analyzed on 50 individuals per group. Additionally, non-targeted metabolomics on plasma and bones were performed to uncover the impact of SDGE supplementation on pullet's metabolome. Results demonstrated that keel bone deformity tended to improve following SDGE supplementation (P = 0.10). Moreover, SDGE intake significantly increased bone dry and mineral content, compared to the control group (P < 0.05) and tended to increase calcium (Ca) (P = 0.074) and phosphorus (P) content (P = 0.055). On the other hand, non-targeted metabolomics on plasma samples revealed an impact on fatty acids and glycerophospholipid metabolisms while bone samples analysis uncovered pathways related to sphingolipid and estrogen mediated signaling pathways. Overall, this study suggests novel mechanisms related to estrogen signaling through lipid metabolism in SDGE supplemented pullets and highlights that nutritional intervention with SDGE in early developmental stage of future laying hens improves their bone health prior to entry into the laying phase, thus proving a useful tool in bone health management.
Collapse
Affiliation(s)
- Fatima Zohra Aberkane
- Nor-Feed SAS, 3 Rue Amedeo Avogadro, 49070 Beaucouzé, France; Univ Angers, SONAS, SFR QUASAV, F-49000 Angers, France; Labcom FeedInTech, 42 Rue Georges Morel, 49070 Beaucouzé, France.
| | - Paul Engler
- Nor-Feed SAS, 3 Rue Amedeo Avogadro, 49070 Beaucouzé, France; Labcom FeedInTech, 42 Rue Georges Morel, 49070 Beaucouzé, France
| | - Severine Boisard
- Univ Angers, SONAS, SFR QUASAV, F-49000 Angers, France; Labcom FeedInTech, 42 Rue Georges Morel, 49070 Beaucouzé, France
| | - Mohamed El Amine Benarbia
- Nor-Feed SAS, 3 Rue Amedeo Avogadro, 49070 Beaucouzé, France; Labcom FeedInTech, 42 Rue Georges Morel, 49070 Beaucouzé, France
| | - David Guilet
- Univ Angers, SONAS, SFR QUASAV, F-49000 Angers, France; Labcom FeedInTech, 42 Rue Georges Morel, 49070 Beaucouzé, France
| |
Collapse
|
2
|
Liu Q, Tabrez S, Niekamp P, Kim CH. Circadian-clock-controlled endocrine and cytokine signals regulate multipotential innate lymphoid cell progenitors in the bone marrow. Cell Rep 2024; 43:114200. [PMID: 38717905 PMCID: PMC11264331 DOI: 10.1016/j.celrep.2024.114200] [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: 08/04/2023] [Revised: 12/12/2023] [Accepted: 04/22/2024] [Indexed: 06/01/2024] Open
Abstract
Innate lymphoid cells (ILCs), strategically positioned throughout the body, undergo population declines over time. A solution to counteract this problem is timely mobilization of multipotential progenitors from the bone marrow. It remains unknown what triggers the mobilization of bone marrow ILC progenitors (ILCPs). We report that ILCPs are regulated by the circadian clock to emigrate and generate mature ILCs in the periphery. We found that circadian-clock-defective ILCPs fail to normally emigrate and generate ILCs. We identified circadian-clock-controlled endocrine and cytokine cues that, respectively, regulate the retention and emigration of ILCPs at distinct times of each day. Activation of the stress-hormone-sensing glucocorticoid receptor upregulates CXCR4 on ILCPs for their retention in the bone marrow, while the interleukin-18 (IL-18) and RORα signals upregulate S1PR1 on ILCPs for their mobilization to the periphery. Our findings establish important roles of circadian signals for the homeostatic efflux of bone marrow ILCPs.
Collapse
Affiliation(s)
- Qingyang Liu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Immunology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shams Tabrez
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Patrick Niekamp
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Chang H Kim
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; Immunology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.
| |
Collapse
|
3
|
Xu X, Han Y, Zhu T, Fan F, Wang X, Liu Y, Luo D. The role of SphK/S1P/S1PR signaling pathway in bone metabolism. Biomed Pharmacother 2023; 169:115838. [PMID: 37944444 DOI: 10.1016/j.biopha.2023.115838] [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: 09/13/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
There are a large number of people worldwide who suffer from osteoporosis, which imposes a huge economic burden, so it is necessary to explore the underlying mechanisms to achieve better supportive and curative care outcomes. Sphingosine kinase (SphK) is an enzyme that plays a crucial role in the synthesis of sphingosine-1-phosphate (S1P). S1P with paracrine and autocrine activities that act through its cell surface S1P receptors (S1PRs) and intracellular signals. In osteoporosis, S1P is indispensable for both normal and disease conditions. S1P has complicated roles in regulating osteoblast and osteoclast, respectively, and there have been exciting developments in understanding how SphK/S1P/S1PR signaling regulates these processes in response to osteoporosis therapy. Here, we review the proliferation, differentiation, apoptosis, and functions of S1P, specifically detailing the roles of S1P and S1PRs in osteoblasts and osteoclasts. Finally, we focus on the S1P-based therapeutic approaches in bone metabolism, which may provide valuable insights into potential therapeutic strategies for osteoporosis.
Collapse
Affiliation(s)
- Xuefeng Xu
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Yi Han
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Tianxin Zhu
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Faxin Fan
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Xin Wang
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Yuqing Liu
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China
| | - Duosheng Luo
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, China.
| |
Collapse
|
4
|
González-Novo R, de Lope-Planelles A, Cruz Rodríguez MP, González-Murillo Á, Madrazo E, Acitores D, García de Lacoba M, Ramírez M, Redondo-Muñoz J. 3D environment controls H3K4 methylation and the mechanical response of the nucleus in acute lymphoblastic leukemia cells. Eur J Cell Biol 2023; 102:151343. [PMID: 37494871 DOI: 10.1016/j.ejcb.2023.151343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/30/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer, and the infiltration of leukemic cells is critical for disease progression and relapse. Nuclear deformability plays a critical role in cancer cell invasion through confined spaces; however, the direct impact of epigenetic changes on the nuclear deformability of leukemic cells remains unclear. Here, we characterized how 3D collagen matrix conditions induced H3K4 methylation in ALL cell lines and clinical samples. We used specific shRNA and chemical inhibitors to target WDR5 (a core subunit involved in H3K4 methylation) and determined that targeting WDR5 reduced the H3K4 methylation induced by the 3D environment and the invasiveness of ALL cells in vitro and in vivo. Intriguingly, targeting WDR5 did not reduce the adhesion or the chemotactic response of leukemia cells, suggesting a different mechanism by which H3K4 methylation might govern ALL cell invasiveness. Finally, we conducted biochemical, and biophysical experiments to determine that 3D environments promoted the alteration of the chromatin, the morphology, and the mechanical behavior of the nucleus in ALL cells. Collectively, our data suggest that 3D environments control an upregulation of H3K4 methylation in ALL cells, and targeting WDR5 might serve as a promising therapeutic target against ALL invasiveness in vivo.
Collapse
Affiliation(s)
- Raquel González-Novo
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Ana de Lope-Planelles
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - María Pilar Cruz Rodríguez
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - África González-Murillo
- Oncolohematology Unit, Hospital Universitario Niño Jesús, Madrid, Spain; Health Research Institute La Princesa, Madrid, Spain
| | - Elena Madrazo
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - David Acitores
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Mario García de Lacoba
- Bioinformatics and Biostatistics Unit, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Manuel Ramírez
- Oncolohematology Unit, Hospital Universitario Niño Jesús, Madrid, Spain; Health Research Institute La Princesa, Madrid, Spain
| | - Javier Redondo-Muñoz
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain.
| |
Collapse
|
5
|
Frost K, Naylor AJ, McGettrick HM. The Ying and Yang of Sphingosine-1-Phosphate Signalling within the Bone. Int J Mol Sci 2023; 24:6935. [PMID: 37108099 PMCID: PMC10139073 DOI: 10.3390/ijms24086935] [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: 02/06/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Bone remodelling is a highly active and dynamic process that involves the tight regulation of osteoblasts, osteoclasts, and their progenitors to allow for a balance of bone resorption and formation to be maintained. Ageing and inflammation are risk factors for the dysregulation of bone remodelling. Once the balance between bone formation and resorption is lost, bone mass becomes compromised, resulting in disorders such as osteoporosis and Paget's disease. Key molecules in the sphingosine-1-phosphate signalling pathway have been identified for their role in regulating bone remodelling, in addition to its more recognised role in inflammatory responses. This review discusses the accumulating evidence for the different, and, in certain circumstances, opposing, roles of S1P in bone homeostasis and disease, including osteoporosis, Paget's disease, and inflammatory bone loss. Specifically, we describe the current, often conflicting, evidence surrounding S1P function in osteoblasts, osteoclasts, and their precursors in health and disease, concluding that S1P may be an effective biomarker of bone disease and also an attractive therapeutic target for disease.
Collapse
Affiliation(s)
| | - Amy J. Naylor
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | | |
Collapse
|
6
|
Yang C, Liu Y, Wang Z, Lin M, Liu C. Controlled mechanical loading improves bone regeneration by regulating type H vessels in a S1Pr1-dependent manner. FASEB J 2022; 36:e22530. [PMID: 36063128 DOI: 10.1096/fj.202200339rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/11/2022]
Abstract
Despite the best treatment, approximately 10% of fractures still face undesirable repair and result in delayed unions or non-unions. Dynamic mechanical stimulation promotes bone formation, when applied at the correct time frame, with optimal loading magnitude, frequency, and repetition. Controlled mechanical loading significantly increases osteogenic cells during the matrix deposition phase of bone repair. In the bone defect, the blood vessel network guides the initial bone formation activities. A unique blood vessel subtype (Type H) exists in bone, which expresses high levels of CD31 and endomucin, and functions to couple angiogenesis and osteogenesis. However, how this form of controlled mechanical loading regulates the Type H vessels and promotes bone formation is still not clear. Sphingosine 1-phosphate (S1P) participates in the bone anabolic process and is a key regulator of the blood vessel. Its receptor, sphingosine 1-phosphate receptor 1 (S1Pr1), is a mechanosensitive protein that regulates vascular integrity. Therefore, we hypothesis that controlled anabolic mechanical loading promotes bone repair by acting on Type H vessels. To study the effect of S1Pr1 on loading induced-bone repair, we utilized a stabilized tibial defect model, which allows for the application of anabolic mechanical loading. Mechanical loading upregulated S1Pr1 within the entire defect, with up to 80% expressed in blood vessels, as observed by deep tissue imaging. Additionally, S1Pr1 antagonism by W146 inhibited the anabolic effects of mechanical loading. We showed that mechanical loading or activating S1Pr1 could induce YAP nuclear translocation, a key regulator in the cell's mechanical response, in endothelial cells (ECs) in vitro. Inhibition of S1Pr1 in endothelial cells by siRNA reduced loading-induced YAP nuclear translocation and expressions of angiogenic genes. In vivo, YAP nuclear translocation in Type H vessels was up-regulated after mechanical loading but was inhibited by antagonizing S1Pr1. S1Pr1 agonist, FTY720, increased bone volume and Type H vessel volume, similar to that of mechanical stimulation. In conclusion, controlled anabolic mechanical loading enhanced bone formation mainly through Type H vessels in a S1Pr1-dependent manner.
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
7
|
Zheng M, Oh SH, Choi N, Choi YJ, Kim J, Sung JH. CXCL12 inhibits hair growth through CXCR4. Biomed Pharmacother 2022; 150:112996. [PMID: 35462338 DOI: 10.1016/j.biopha.2022.112996] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/12/2022] [Accepted: 04/17/2022] [Indexed: 11/26/2022] Open
Abstract
CXCL12 and its receptors, which are highly expressed in the skin, are associated with various cutaneous diseases, including androgenic alopecia. However, their expression and role during the hair cycle are unknown. This study aims to investigate the expression of CXCL12 and its receptor, CXCR4, in the vicinity of hair follicles and their effect on hair growth. CXCL12 was highly expressed in dermal fibroblasts (DFs) and its level was elevated throughout the catagen and telogen phases of the hair cycle. CXCR4 is expressed in the dermal papilla (DP) and outer root sheath (ORS). In hair organ culture, hair loss was induced by recombinant CXCL12 therapy, which delayed the telogen-to-anagen transition and decreased hair length. In contrast, the suppression of CXCL12 using a neutralizing antibody and siRNA triggered the telogen-to-anagen transition and increased hair length in hair organ culture. Neutralization of CXCR7, one of the two receptors for CXCL12, only slightly affected hair growth. However, inhibition of CXCR4, the other receptor for CXCL12, increased hair growth to a considerable extent. In addition, in hair organ culture, the conditioned medium from DFs with CXCL12 siRNA considerably increased the hair length and induced proliferation of DP and ORS cells. CXCL12, through CXCR4 activation, increased STAT3 and STAT5 phosphorylation in DP and ORS cells. In contrast, blocking CXCL12 and CXCR4 decreased the phosphorylation of STAT3 and STAT5. In summary, these findings suggest that CXCL12 inhibits hair growth via the CXCR4/STAT signaling pathway and that CXCL12/CXCR4 pathway inhibitors are a promising treatment option for hair growth.
Collapse
Affiliation(s)
- Mei Zheng
- Epi Biotech Co., Ltd., Incheon 21983, South Korea
| | - Sang Ho Oh
- Department of Dermatology and Cutaneous Biology Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Nahyun Choi
- Epi Biotech Co., Ltd., Incheon 21983, South Korea
| | | | - Jino Kim
- New Hair Institute, Seoul 06134, South Korea
| | - Jong-Hyuk Sung
- Epi Biotech Co., Ltd., Incheon 21983, South Korea; College of Pharmacy, Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, South Korea.
| |
Collapse
|
8
|
Grewe JM, Knapstein PR, Donat A, Jiang S, Smit DJ, Xie W, Keller J. The role of sphingosine-1-phosphate in bone remodeling and osteoporosis. Bone Res 2022; 10:34. [PMID: 35396384 PMCID: PMC8993882 DOI: 10.1038/s41413-022-00205-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/17/2021] [Accepted: 01/17/2022] [Indexed: 12/27/2022] Open
Abstract
Osteoporosis is a systemic bone disease that affects more than 200 million people worldwide and is caused by the disruption of the equilibrium between osteoclastic bone resorption and osteoblastic bone formation. Sphingosine-1-phosphate (S1P) is a natural, bioactive sphingolipid that has been shown to play a major role in cardiovascular and immunological pathologies by regulating biological and cellular processes, including migration, differentiation, proliferation and survival. Recent studies also suggest a central role for S1P in bone diseases, including osteoporosis; however, the effects of S1P, particularly in bone metabolism, remain to be further elucidated. In this review, we summarize the available literature on the role of S1P in bone metabolism with a focus on osteoporosis. On the cellular level, S1P acts as an osteoclast-osteoblast coupling factor to promote osteoblast proliferation and bone formation. Moreover, the recruitment of osteoclast precursors to resorption sites is regulated by the interplay of S1P gradients and S1P receptor expression. From a clinical perspective, increasing evidence suggests that systemically elevated S1P blood levels may serve as an independent risk factor for osteoporosis-related fractures. Taken together, S1P signaling is a potential therapeutic target and may serve as a novel biomarker in patients with systemic bone disease.
Collapse
Affiliation(s)
- Justus M Grewe
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.,Clinic and Polyclinic for Vascular Medicine, University Heart Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Paul-Richard Knapstein
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Antonia Donat
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Shan Jiang
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Daniel J Smit
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Weixin Xie
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Johannes Keller
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|
9
|
Qi T, Li L, Weidong T. The Role of Sphingolipid Metabolism in Bone Remodeling. Front Cell Dev Biol 2021; 9:752540. [PMID: 34912800 PMCID: PMC8666436 DOI: 10.3389/fcell.2021.752540] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/11/2021] [Indexed: 02/05/2023] Open
Abstract
Emerging studies of bioactive lipids have made many exciting discoveries in recent years. Sphingolipids and their metabolites perform a wide variety of cellular functions beyond energy metabolism. Emerging evidence based on genetically manipulated mouse models and molecular biology allows us to obtain new insights into the role sphingolipid played on skeletal remodeling. This review summarizes studies or understandings of the crosstalk between sphingomyelin, ceramide, and sphingosine-1-phosphate (S1P) of sphingolipids family and the cells, especially osteoblasts and osteoclasts of the bone through which bone is remodeled during life constantly. This review also shows agonists and antagonists of S1P as possible therapeutic options and opportunities on bone diseases.
Collapse
Affiliation(s)
- Tang Qi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Liao Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tian Weidong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Gambari L, Grassi F, Roseti L, Grigolo B, Desando G. Learning from Monocyte-Macrophage Fusion and Multinucleation: Potential Therapeutic Targets for Osteoporosis and Rheumatoid Arthritis. Int J Mol Sci 2020; 21:ijms21176001. [PMID: 32825443 PMCID: PMC7504439 DOI: 10.3390/ijms21176001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
Excessive bone resorption by osteoclasts (OCs) covers an essential role in developing bone diseases, such as osteoporosis (OP) and rheumatoid arthritis (RA). Monocytes or macrophages fusion and multinucleation (M-FM) are key processes for generating multinucleated mature cells with essential roles in bone remodelling. Depending on the phenotypic heterogeneity of monocyte/macrophage precursors and the extracellular milieu, two distinct morphological and functional cell types can arise mature OCs and giant cells (GCs). Despite their biological relevance in several physiological and pathological responses, many gaps exist in our understanding of their formation and role in bone, including the molecular determinants of cell fusion and multinucleation. Here, we outline fusogenic molecules during M-FM involved in OCs and GCs formation in healthy conditions and during OP and RA. Moreover, we discuss the impact of the inflammatory milieu on modulating macrophages phenotype and their differentiation towards mature cells. Methodological approach envisaged searches on Scopus, Web of Science Core Collection, and EMBASE databases to select relevant studies on M-FM, osteoclastogenesis, inflammation, OP, and RA. This review intends to give a state-of-the-art description of mechanisms beyond osteoclastogenesis and M-FM, with a focus on OP and RA, and to highlight potential biological therapeutic targets to prevent extreme bone loss.
Collapse
Affiliation(s)
| | | | - Livia Roseti
- Correspondence: (L.R.); (B.G.); Tel.: +39-051-6366090 (B.G.)
| | | | | |
Collapse
|
11
|
Rajendiran S, Smith-Berdan S, Kunz L, Risolino M, Selleri L, Schroeder T, Forsberg EC. Ubiquitous overexpression of CXCL12 confers radiation protection and enhances mobilization of hematopoietic stem and progenitor cells. Stem Cells 2020; 38:1159-1174. [PMID: 32442338 DOI: 10.1002/stem.3205] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022]
Abstract
C-X-C motif chemokine ligand 12 (CXCL12; aka SDF1α) is a major regulator of a number of cellular systems, including hematopoiesis, where it influences hematopoietic cell trafficking, proliferation, and survival during homeostasis and upon stress and disease. A variety of constitutive, temporal, ubiquitous, and cell-specific loss-of-function models have documented the functional consequences on hematopoiesis upon deletion of Cxcl12. Here, in contrast to loss-of-function experiments, we implemented a gain-of-function approach by generating a doxycycline-inducible transgenic mouse model that enables spatial and temporal overexpression of Cxcl12. We demonstrated that ubiquitous CXCL12 overexpression led to an increase in multipotent progenitors in the bone marrow and spleen. The CXCL12+ mice displayed reduced reconstitution potential as either donors or recipients in transplantation experiments. Additionally, we discovered that Cxcl12 overexpression improved hematopoietic stem and progenitor cell mobilization into the blood, and conferred radioprotection by promoting quiescence. Thus, this new CXCL12+ mouse model provided new insights into major facets of hematopoiesis and serves as a versatile resource for studying CXCL12 function in a variety of contexts.
Collapse
Affiliation(s)
- Smrithi Rajendiran
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, USA
| | - Stephanie Smith-Berdan
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, USA
| | - Leo Kunz
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Basel, Switzerland
| | - Maurizio Risolino
- Program in Craniofacial Biology, Institute of Human Genetics, Eli and Edyth Broad Center of Regeneration Medicine and Stem Cell Research, Departments of Orofacial Sciences and Anatomy, University of California, San Francisco, California, USA
| | - Licia Selleri
- Program in Craniofacial Biology, Institute of Human Genetics, Eli and Edyth Broad Center of Regeneration Medicine and Stem Cell Research, Departments of Orofacial Sciences and Anatomy, University of California, San Francisco, California, USA
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Basel, Switzerland
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, USA
| |
Collapse
|
12
|
Sadeghi L, Arvidsson G, Merrien M, Wasik AM, Görgens A, Smith CE, Sander B, P. Wright A. Differential B-Cell Receptor Signaling Requirement for Adhesion of Mantle Cell Lymphoma Cells to Stromal Cells. Cancers (Basel) 2020; 12:cancers12051143. [PMID: 32370190 PMCID: PMC7281289 DOI: 10.3390/cancers12051143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 01/01/2023] Open
Abstract
Interactions between lymphoma cells and stromal cells play a key role in promoting tumor survival and development of drug resistance. We identified differences in key signaling pathways between the JeKo-1 and REC-1 mantle cell lymphoma (MCL) cell lines, displaying different patterns of stromal cell adhesion and chemotaxis towards stroma-conditioned medium. The identified adhesion-regulated genes reciprocated important aspects of microenvironment-mediated gene modulation in MCL patients. Five-hundred and ninety genes were differently regulated between the cell lines upon adhesion to stromal cells, while 32 genes were similarly regulated in both cell lines. Regulation of B-cell Receptor (BCR) signature genes in adherent cells was specific for JeKo-1. Inhibition of BCR using siRNA or clinically approved inhibitors, Ibrutinib and Acalabrutinib, decreased adhesion of JeKo-1, but not REC-1 cells. Cell surface levels of chemokine receptor CXCR4 were higher in JeKo-1, facilitating migration and adhesion of JeKo-1 but not REC-1 cells. Surface levels of ICAM1 adhesion protein differ for REC-1 and JeKo-1. While ICAM1 played a positive role in adherence of both cell lines to stromal cells, S1PR1 had an inhibitory effect. Our results provide a model framework for further investigation of mechanistic differences in patient-response to new pathway-specific drugs.
Collapse
Affiliation(s)
- Laia Sadeghi
- Department of Laboratory Medicine, Division of Biomedical and Cellular Medicine, Karolinska Institutet, 141 57 Stockholm, Sweden; (L.S.); (G.A.); (A.G.); (C.I.E.S.)
| | - Gustav Arvidsson
- Department of Laboratory Medicine, Division of Biomedical and Cellular Medicine, Karolinska Institutet, 141 57 Stockholm, Sweden; (L.S.); (G.A.); (A.G.); (C.I.E.S.)
| | - Magali Merrien
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, 141 52 Stockholm, Sweden; (M.M.); (A.M.W.); (B.S.)
| | - Agata M. Wasik
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, 141 52 Stockholm, Sweden; (M.M.); (A.M.W.); (B.S.)
| | - André Görgens
- Department of Laboratory Medicine, Division of Biomedical and Cellular Medicine, Karolinska Institutet, 141 57 Stockholm, Sweden; (L.S.); (G.A.); (A.G.); (C.I.E.S.)
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg, 45 147 Essen, Germany
| | - C.I. Edvard Smith
- Department of Laboratory Medicine, Division of Biomedical and Cellular Medicine, Karolinska Institutet, 141 57 Stockholm, Sweden; (L.S.); (G.A.); (A.G.); (C.I.E.S.)
| | - Birgitta Sander
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, 141 52 Stockholm, Sweden; (M.M.); (A.M.W.); (B.S.)
| | - Anthony P. Wright
- Department of Laboratory Medicine, Division of Biomedical and Cellular Medicine, Karolinska Institutet, 141 57 Stockholm, Sweden; (L.S.); (G.A.); (A.G.); (C.I.E.S.)
- Correspondence:
| |
Collapse
|
13
|
Wang JL, Han MZ. [The pathogenesis of poor graft function after allogeneic hematopoietic stem cell transplantation]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2020; 40:792-795. [PMID: 31648490 PMCID: PMC7342449 DOI: 10.3760/cma.j.issn.0253-2727.2019.09.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- J L Wang
- Institute of Hematology & Blood Diseases Hospital, CAMS & PUMC, Tianjin 300020, China
| | | |
Collapse
|
14
|
Exercise reduces inflammatory cell production and cardiovascular inflammation via instruction of hematopoietic progenitor cells. Nat Med 2019; 25:1761-1771. [PMID: 31700184 PMCID: PMC6858591 DOI: 10.1038/s41591-019-0633-x] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
Abstract
A sedentary lifestyle, chronic inflammation and leukocytosis increase atherosclerosis; however, it remains unclear whether regular physical activity influences leukocyte production. Here we show that voluntary running decreases hematopoietic activity in mice. Exercise protects mice and humans with atherosclerosis from chronic leukocytosis but does not compromise emergency hematopoiesis in mice. Mechanistically, exercise diminishes leptin production in adipose tissue, augmenting quiescence-promoting hematopoietic niche factors in leptin-receptor-positive stromal bone marrow cells. Induced deletion of the leptin receptor in Prrx1-creERT2; Leprfl/fl mice reveals that leptin's effect on bone marrow niche cells regulates hematopoietic stem and progenitor cell (HSPC) proliferation and leukocyte production, as well as cardiovascular inflammation and outcomes. Whereas running wheel withdrawal quickly reverses leptin levels, the impact of exercise on leukocyte production and on the HSPC epigenome and transcriptome persists for several weeks. Together, these data show that physical activity alters HSPCs via modulation of their niche, reducing hematopoietic output of inflammatory leukocytes.
Collapse
|
15
|
Xiang L, Gilkes DM. The Contribution of the Immune System in Bone Metastasis Pathogenesis. Int J Mol Sci 2019; 20:ijms20040999. [PMID: 30823602 PMCID: PMC6412551 DOI: 10.3390/ijms20040999] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/31/2022] Open
Abstract
Bone metastasis is associated with significant morbidity for cancer patients and results in a reduced quality of life. The bone marrow is a fertile soil containing a complex composition of immune cells that may actually provide an immune-privileged niche for disseminated tumor cells to colonize and proliferate. In this unique immune milieu, multiple immune cells including T cells, natural killer cells, macrophages, dendritic cells, myeloid-derived suppressor cells, and neutrophils are involved in the process of bone metastasis. In this review, we will discuss the crosstalk between immune cells in bone microenvironment and their involvement with cancer cell metastasis to the bone. Furthermore, we will highlight the anti-tumoral and pro-tumoral function of each immune cell type that contributes to bone metastasis. We will end with a discussion of current therapeutic strategies aimed at sensitizing immune cells.
Collapse
Affiliation(s)
- Lisha Xiang
- State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu 610041, China.
| | - Daniele M Gilkes
- Breast & Ovarian Cancer Program, Department of Oncology, The Johns Hopkins University School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA.
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
16
|
Golan K, Kumari A, Kollet O, Khatib-Massalha E, Subramaniam MD, Ferreira ZS, Avemaria F, Rzeszotek S, García-García A, Xie S, Flores-Figueroa E, Gur-Cohen S, Itkin T, Ludin-Tal A, Massalha H, Bernshtein B, Ciechanowicz AK, Brandis A, Mehlman T, Bhattacharya S, Bertagna M, Cheng H, Petrovich-Kopitman E, Janus T, Kaushansky N, Cheng T, Sagi I, Ratajczak MZ, Méndez-Ferrer S, Dick JE, Markus RP, Lapidot T. Daily Onset of Light and Darkness Differentially Controls Hematopoietic Stem Cell Differentiation and Maintenance. Cell Stem Cell 2018; 23:572-585.e7. [PMID: 30174297 DOI: 10.1016/j.stem.2018.08.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 04/10/2018] [Accepted: 08/06/2018] [Indexed: 12/31/2022]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) tightly couple maintenance of the bone marrow (BM) reservoir, including undifferentiated long-term repopulating hematopoietic stem cells (LT-HSCs), with intensive daily production of mature leukocytes and blood replenishment. We found two daily peaks of BM HSPC activity that are initiated by onset of light and darkness providing this coupling. Both peaks follow transient elevation of BM norepinephrine and TNF secretion, which temporarily increase HSPC reactive oxygen species (ROS) levels. Light-induced norepinephrine and TNF secretion augments HSPC differentiation and increases vascular permeability to replenish the blood. In contrast, darkness-induced TNF increases melatonin secretion to drive renewal of HSPCs and LT-HSC potential through modulating surface CD150 and c-Kit expression, increasing COX-2/αSMA+ macrophages, diminishing vascular permeability, and reducing HSPC ROS levels. These findings reveal that light- and darkness-induced daily bursts of norepinephrine, TNF, and melatonin within the BM are essential for synchronized mature blood cell production and HSPC pool repopulation.
Collapse
Affiliation(s)
- Karin Golan
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Anju Kumari
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Orit Kollet
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Zulma S Ferreira
- Physiology Department, University of São Paulo, São Paulo, Brazil
| | | | - Sylwia Rzeszotek
- Physiology Department, Pomeranian Medical University, Szczecin, Poland
| | | | - Stephanie Xie
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Eugenia Flores-Figueroa
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Oncology Research Unit, Oncology Hospital, National Medical Center Century XXI, IMSS, Mexico City, Mexico
| | - Shiri Gur-Cohen
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Tomer Itkin
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Aya Ludin-Tal
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Hassan Massalha
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Biana Bernshtein
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | | | - Alexander Brandis
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tevie Mehlman
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | | | - Mayla Bertagna
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Hui Cheng
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel; State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | | | - Tomasz Janus
- Forensic Medicine Department, Pomeranian Medical University, Szczecin, Poland
| | | | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Irit Sagi
- Biological Regulation Department, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Regina P Markus
- Physiology Department, University of São Paulo, São Paulo, Brazil
| | - Tsvee Lapidot
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
17
|
Mobilized Peripheral Blood versus Cord Blood: Insight into the Distinct Role of Proinflammatory Cytokines on Survival, Clonogenic Ability, and Migration of CD34 + Cells. Mediators Inflamm 2018; 2018:5974613. [PMID: 30116149 PMCID: PMC6079419 DOI: 10.1155/2018/5974613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/24/2018] [Accepted: 05/31/2018] [Indexed: 01/12/2023] Open
Abstract
Inflammation may play a role in cancer. However, the contribution of cytokine-mediated crosstalk between normal hemopoietic stem/progenitor cells (HSPCs) and their (inflammatory) microenvironment is largely elusive. Here we compared survival, phenotype, and function of neonatal (umbilical cord blood (CB)) and adult (normal G-CSF-mobilized peripheral blood (mPB)) CD34+ cells after in vitro exposure to combined crucial inflammatory factors such as interleukin- (IL-) 1β, IL-6, tumor necrosis factor- (TNF-) α, or tissue inhibitor of metalloproteinases-1 (TIMP-1). To mimic bone marrow (BM) niche, coculture experiments with normal BM stromal cells (BMSCs) were also performed. We found that combined inflammatory cytokines increased only the in vitro survival of CB-derived CD34+ cells by reducing apoptosis. Conversely, selected combinations of inflammatory cytokines (IL-1β + TNF-α, IL-6 + TNF-α, and IL-1β + TNF-α + TIMP-1) mainly enhanced the in vitro CXCR4-driven migration of mPB-derived CD34+ cells. TNF-α, alone or in combination, upregulated CD44 and CD13 expression in both sources. Finally, BMSCs alone increased survival/migration of CB- and mPB-derived CD34+ cells at the same extent of the combined inflammatory cytokines; importantly, their copresence did not show additive/synergistic effect. Taken together, these data indicate that combined proinflammatory stimuli promote distinct in vitro functional activation of neonatal or adult normal HSPCs.
Collapse
|
18
|
Romani R, Manni G, Donati C, Pirisinu I, Bernacchioni C, Gargaro M, Pirro M, Calvitti M, Bagaglia F, Sahebkar A, Clerici G, Matino D, Pomili G, Di Renzo GC, Talesa VN, Puccetti P, Fallarino F. S1P promotes migration, differentiation and immune regulatory activity in amniotic-fluid-derived stem cells. Eur J Pharmacol 2018; 833:173-182. [PMID: 29886240 PMCID: PMC6086338 DOI: 10.1016/j.ejphar.2018.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 06/01/2018] [Accepted: 06/06/2018] [Indexed: 12/22/2022]
Abstract
Stem cells have high potential for cell therapy in regenerative medicine. We previously isolated stem cell types from human amniotic fluid, derived from prenatal amniocentesis. One type, characterized by a fast doubling time, was designated as fast human amniotic stem cells (fHASCs). These cells exhibited high differentiation potential and immunoregulatory properties. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite that influences stem-cell pluripotency, differentiation, mobility, and regulates immune functions. In this study, we investigated the influence of S1P on fHASC migration, proliferation, differentiation and immune regulatory functions. We found that fHASC stimulation with S1P potentiated their migratory and proliferative activity in vitro. Notably, short fHASC exposure to S1P enhanced their differentiation towards multiple lineages, including adipocytes, osteocytes and endothelial cells, an effect that was associated with downregulation of the main transcription factors involved in the maintenance of a stem-cell undifferentiated state. A specific crosstalk between S1P and tumor growth factor β1 (TGF-β1) has recently been demonstrated. We found that fHASC exposure to S1P in combination with TGF-β1 promoted the expression of the immune regulatory pathway of indoleamine 2,3-dioxygenase 1 (IDO1). In addition, human peripheral blood mononuclear cells, co-cultured with fHASCs treated with S1P and TGF-β1, expanded regulatory T-cells, via a mechanism requiring IDO1. Overall, this study demonstrates that S1P potentiates several properties in fHASCs, an effect that may be critical for exploiting the therapeutic potential of fHASCs and might explain the specific effects of S1P on stem cells during pregnancy.
Collapse
Affiliation(s)
- Rita Romani
- Department of Experimental Medicine, University of Perugia, Italy
| | - Giorgia Manni
- Department of Experimental Medicine, University of Perugia, Italy
| | - Chiara Donati
- Department of Experimental Biomedical Sciences and Clinics University of Florence, Italy
| | - Irene Pirisinu
- Department of Experimental Medicine, University of Perugia, Italy
| | - Caterina Bernacchioni
- Department of Experimental Biomedical Sciences and Clinics University of Florence, Italy
| | - Marco Gargaro
- Department of Experimental Medicine, University of Perugia, Italy
| | - Matteo Pirro
- Department of Medicine, University of Perugia, Italy
| | - Mario Calvitti
- Department of Experimental Medicine, University of Perugia, Italy
| | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Graziano Clerici
- Department of Obstetrics and Gynaecology and Centre for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Davide Matino
- Department of Experimental Medicine, University of Perugia, Italy
| | - Giovanni Pomili
- Department of Obstetrics and Gynaecology and Centre for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Gian Carlo Di Renzo
- Department of Obstetrics and Gynaecology and Centre for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | | | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia, Italy
| | | |
Collapse
|
19
|
Graham N, Qian BZ. Mesenchymal Stromal Cells: Emerging Roles in Bone Metastasis. Int J Mol Sci 2018; 19:E1121. [PMID: 29642534 PMCID: PMC5979535 DOI: 10.3390/ijms19041121] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/25/2018] [Accepted: 03/29/2018] [Indexed: 12/13/2022] Open
Abstract
Bone metastasis is the most advanced stage of many cancers and indicates a poor prognosis for patients due to resistance to anti-tumor therapies. The establishment of metastasis within the bone is a multistep process. To ensure survival within the bone marrow, tumor cells must initially colonize a niche in which they can enter dormancy. Subsequently, reactivation permits the proliferation and growth of the tumor cells, giving rise to a macro-metastasis displayed clinically as a bone metastatic lesion. Here, we review the evidences that suggest mesenchymal stromal cells play an important role in each of these steps throughout the development of bone metastasis. Similarities between the molecular mechanisms implicated in these processes and those involved in the homeostasis of the bone indicate that the metastatic cells may exploit the homeostatic processes to their own advantage. Identifying the molecular interactions between the mesenchymal stromal cells and tumor cells that promote tumor development may offer insight into potential therapeutic targets that could be utilized to treat bone metastasis.
Collapse
Affiliation(s)
- Nicola Graham
- Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| | - Bin-Zhi Qian
- Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.
- Edinburgh Cancer Research UK Centre, University of Edinburgh, Edinburgh EH4 2XR, UK.
| |
Collapse
|
20
|
Hintzsche H, Montag G, Stopper H. Induction of micronuclei by four cytostatic compounds in human hematopoietic stem cells and human lymphoblastoid TK6 cells. Sci Rep 2018; 8:3371. [PMID: 29463873 PMCID: PMC5820321 DOI: 10.1038/s41598-018-21680-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 02/08/2018] [Indexed: 12/21/2022] Open
Abstract
For mutagenicity testing, primary lymphocytes or mammalian cell lines are employed. However, the true target for carcinogenic action of mutagenic chemicals may be stem cells. Since hematopoietic cancers induced by chemical agents originate at the hematopoietic stem cell (HSC) stage and since one of the side effects of chemotherapeutic cancer treatment is the induction of secondary tumors, often leukemias, HSC may be a suitable cell system. We compared the sensitivity of HSC with the genotoxicity testing cell line TK6 for chromosomal mutations. HSC were less sensitive than TK6 cells for the genotoxic effects of the model genotoxins and chemotherapeutic agents doxorubicin, vinblastine, methyl methanesulfonate (MMS) and equally sensitive for mitomycin C (MMC). However, loss of viability after mitomycin C treatment was higher in HSC than in TK6 cells. Among the factors that may influence sensitivity for genomic damage, the generation or response to reactive oxygen species (ROS) and the effectiveness of DNA damage response can be discussed. Here we show that HSC can be used in a standard micronucleus test protocol for chromosomal mutations and that their sensitivity was not higher than that of a classical testing cell line.
Collapse
Affiliation(s)
- Henning Hintzsche
- Institute of Pharmacology and Toxicology, University of Wuerzburg, Versbacher Str. 9, 97078, Wuerzburg, Germany.,Bavarian Health and Food Safety Authority, Eggenreuther Weg 43, 91058, Erlangen, Germany
| | - Gracia Montag
- Institute of Pharmacology and Toxicology, University of Wuerzburg, Versbacher Str. 9, 97078, Wuerzburg, Germany
| | - Helga Stopper
- Institute of Pharmacology and Toxicology, University of Wuerzburg, Versbacher Str. 9, 97078, Wuerzburg, Germany.
| |
Collapse
|
21
|
Khatib-Massalha E, Lapid K, Golan K, Kollet O, Gur-Cohen S, Bitan M, Kumari A, Lapidot T. Interactions Between Hematopoietic Stem and Progenitor Cells and the Bone Marrow. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00014-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
22
|
Goichberg P. Current Understanding of the Pathways Involved in Adult Stem and Progenitor Cell Migration for Tissue Homeostasis and Repair. Stem Cell Rev Rep 2017; 12:421-37. [PMID: 27209167 DOI: 10.1007/s12015-016-9663-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the advancements in the field of adult stem and progenitor cells grows the recognition that the motility of primitive cells is a pivotal aspect of their functionality. There is accumulating evidence that the recruitment of tissue-resident and circulating cells is critical for organ homeostasis and effective injury responses, whereas the pathobiology of degenerative diseases, neoplasm and aging, might be rooted in the altered ability of immature cells to migrate. Furthermore, understanding the biological machinery determining the translocation patterns of tissue progenitors is of great relevance for the emerging methodologies for cell-based therapies and regenerative medicine. The present article provides an overview of studies addressing the physiological significance and diverse modes of stem and progenitor cell trafficking in adult mammalian organs, discusses the major microenvironmental cues regulating cell migration, and describes the implementation of live imaging approaches for the exploration of stem cell movement in tissues and the factors dictating the motility of endogenous and transplanted cells with regenerative potential.
Collapse
Affiliation(s)
- Polina Goichberg
- Department Anesthesiology, Perioperative and Pain Medicine, Harvard Medical School, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
23
|
Yang G, Chen S, Ma A, Lu J, Wang T. Identification of the difference in the pathogenesis in heart failure arising from different etiologies using a microarray dataset. Clinics (Sao Paulo) 2017; 72:600-608. [PMID: 29160422 PMCID: PMC5666440 DOI: 10.6061/clinics/2017(10)03] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/24/2017] [Accepted: 07/19/2017] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Clinically, patients with chronic heart failure arising from different etiologies receive the same treatment. However, the prognoses of these patients differ. The purpose of this study was to elucidate whether the pathogenesis of heart failure arising from different etiologies differs. METHODS Heart failure-related dataset GSE1145 was obtained from the Gene Expression Omnibus database. Differentially expressed genes were identified using R. A protein-protein interaction network of the differentially expressed genes was constructed using Search Tool for the Retrieval of Interacting Genes. The modules in each network were analyzed by Molecular Complex Detection of Cytoscape. The Database for Annotation, Visualization and Integrated Discovery was used to obtain the functions of the modules. RESULTS Samples contained in GSE1145 were myocardial tissues from patients with dilated cardiomyopathy, familial cardiomyopathy, hypertrophic cardiomyopathy, ischemic cardiomyopathy, and post-partum cardiomyopathy. The differentially expressed genes, modules, and functions of the modules associated with different etiologies varied. Abnormal formation of extracellular matrix was overlapping among five etiologies. The change in cytoskeleton organization was specifically detected in dilated cardiomyopathy. The activation of the Wnt receptor signaling pathway was limited to hypertrophic cardiomyopathy. The change in nucleosome and chromatin assembly was associated with only familial cardiomyopathy. Germ cell migration and disrupted cellular calcium ion homeostasis were solely detected in ischemic cardiomyopathy. The change in the metabolic process of glucose and triglyceride was detected in only post-partum cardiomyopathy. CONCLUSION These results indicate that the pathogenesis of heart failure arising from different etiologies varies, which may provide molecular evidence supporting etiology-based treatment for heart failure patients.
Collapse
Affiliation(s)
- Guodong Yang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, China
| | - Shuping Chen
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, China
| | - Aiqun Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, China
- Key Laboratory of Molecular Cardiology, Shaanxi Province, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, China
| | - Jun Lu
- Clinical Research Center, First Affiliated Hospital of Xi’an Jiaotong University, China
| | - Tingzhong Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, China
- Key Laboratory of Molecular Cardiology, Shaanxi Province, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, China
| |
Collapse
|
24
|
Sphingosine 1-phosphate (S1P) signalling: Role in bone biology and potential therapeutic target for bone repair. Pharmacol Res 2017; 125:232-245. [PMID: 28855094 DOI: 10.1016/j.phrs.2017.08.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/30/2022]
Abstract
The lipid mediator sphingosine 1-phosphate (S1P) affects cellular functions in most systems. Interest in its therapeutic potential has increased following the discovery of its G protein-coupled receptors and the recent availability of agents that can be safely administered in humans. Although the role of S1P in bone biology has been the focus of much less research than its role in the nervous, cardiovascular and immune systems, it is becoming clear that this lipid influences many of the functions, pathways and cell types that play a key role in bone maintenance and repair. Indeed, S1P is implicated in many osteogenesis-related processes including stem cell recruitment and subsequent differentiation, differentiation and survival of osteoblasts, and coupling of the latter cell type with osteoclasts. In addition, S1P's role in promoting angiogenesis is well-established. The pleiotropic effects of S1P on bone and blood vessels have significant potential therapeutic implications, as current therapeutic approaches for critical bone defects show significant limitations. Because of the complex effects of S1P on bone, the pharmacology of S1P-like agents and their physico-chemical properties, it is likely that therapeutic delivery of S1P agents will offer significant advantages compared to larger molecular weight factors. Hence, it is important to explore novel methods of utilizing S1P agents therapeutically, and improve our understanding of how S1P and its receptors modulate bone physiology and repair.
Collapse
|
25
|
Munoz AI, Tello JI. On a mathematical model of bone marrow metastatic niche. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2017; 14:289-304. [PMID: 27879134 DOI: 10.3934/mbe.2017019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
We propose a mathematical model to describe tumor cells movement towards a metastasis location into the bone marrow considering the influence of chemotaxis inhibition due to the action of a drug. The model considers the evolution of the signaling molecules CXCL-12 secreted by osteoblasts (bone cells responsible of the mineralization of the bone) and PTHrP (secreted by tumor cells) which activates osteoblast growth. The model consists of a coupled system of second order PDEs describing the evolution of CXCL-12 and PTHrP, an ODE of logistic type to model the Osteoblasts density and an extra equation for each cancer cell. We also simulate the system to illustrate the qualitative behavior of the solutions. The numerical method of resolution is also presented in detail.
Collapse
Affiliation(s)
- Ana Isabel Munoz
- Departamento de Matematica Aplicada, Ciencia e Ingeniera de Materiales y Tecnologia Electronica, ESCET, Universidad Rey Juan Carlos, E28933, Mostoles, Madrid, Spain
| | | |
Collapse
|
26
|
Kan C, Vargas G, Pape FL, Clézardin P. Cancer Cell Colonisation in the Bone Microenvironment. Int J Mol Sci 2016; 17:ijms17101674. [PMID: 27782035 PMCID: PMC5085707 DOI: 10.3390/ijms17101674] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 02/06/2023] Open
Abstract
Bone metastases are a common complication of epithelial cancers, of which breast, prostate and lung carcinomas are the most common. The establishment of cancer cells to distant sites such as the bone microenvironment requires multiple steps. Tumour cells can acquire properties to allow epithelial-to-mesenchymal transition, extravasation and migration. Within the bone metastatic niche, disseminated tumour cells may enter a dormancy stage or proliferate to adapt and survive, interacting with bone cells such as hematopoietic stem cells, osteoblasts and osteoclasts. Cross-talk with the bone may alter tumour cell properties and, conversely, tumour cells may also acquire characteristics of the surrounding microenvironment, in a process known as osteomimicry. Alternatively, these cells may also express osteomimetic genes that allow cell survival or favour seeding to the bone marrow. The seeding of tumour cells in the bone disrupts bone-forming and bone-resorbing activities, which can lead to macrometastasis in bone. At present, bone macrometastases are incurable with only palliative treatment available. A better understanding of how these processes influence the early onset of bone metastasis may give insight into potential therapies. This review will focus on the early steps of bone colonisation, once disseminated tumour cells enter the bone marrow.
Collapse
Affiliation(s)
- Casina Kan
- National Institute of Health and Medical Research (INSERM), UMR 1033, Lyon 69372, France.
- Faculty of Medicine RTH Laennec, University of Lyon, Villeurbanne 69372, France.
| | - Geoffrey Vargas
- National Institute of Health and Medical Research (INSERM), UMR 1033, Lyon 69372, France.
- Faculty of Medicine RTH Laennec, University of Lyon, Villeurbanne 69372, France.
| | - François Le Pape
- National Institute of Health and Medical Research (INSERM), UMR 1033, Lyon 69372, France.
- Faculty of Medicine RTH Laennec, University of Lyon, Villeurbanne 69372, France.
| | - Philippe Clézardin
- National Institute of Health and Medical Research (INSERM), UMR 1033, Lyon 69372, France.
- Faculty of Medicine RTH Laennec, University of Lyon, Villeurbanne 69372, France.
| |
Collapse
|
27
|
ATF4 plays a pivotal role in the development of functional hematopoietic stem cells in mouse fetal liver. Blood 2015; 126:2383-91. [PMID: 26384355 DOI: 10.1182/blood-2015-03-633354] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 09/08/2015] [Indexed: 01/23/2023] Open
Abstract
The fetal liver (FL) serves as a predominant site for expansion of functional hematopoietic stem cells (HSCs) during mouse embryogenesis. However, the mechanisms for HSC development in FL remain poorly understood. In this study, we demonstrate that deletion of activating transcription factor 4 (ATF4) significantly impaired hematopoietic development and reduced HSC self-renewal in FL. In contrast, generation of the first HSC population in the aorta-gonad-mesonephros region was not affected. The migration activity of ATF4(-/-) HSCs was moderately reduced. Interestingly, the HSC-supporting ability of both endothelial and stromal cells in FL was significantly compromised in the absence of ATF4. Gene profiling using RNA-seq revealed downregulated expression of a panel of cytokines in ATF4(-/-) stromal cells, including angiopoietin-like protein 3 (Angptl3) and vascular endothelial growth factor A (VEGFA). Addition of Angptl3, but not VEGFA, partially rescued the repopulating defect of ATF4(-/-) HSCs in the culture. Furthermore, chromatin immunoprecipitation assay in conjunction with silencing RNA-mediated silencing and complementary DNA overexpression showed transcriptional control of Angptl3 by ATF4. To summarize, ATF4 plays a pivotal role in functional expansion and repopulating efficiency of HSCs in developing FL, and it acts through upregulating transcription of cytokines such as Angptl3 in the microenvironment.
Collapse
|
28
|
Klamer S, Voermans C. The role of novel and known extracellular matrix and adhesion molecules in the homeostatic and regenerative bone marrow microenvironment. Cell Adh Migr 2015; 8:563-77. [PMID: 25482635 PMCID: PMC4594522 DOI: 10.4161/19336918.2014.968501] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Maintenance of haematopoietic stem cells and differentiation of committed progenitors occurs in highly specialized niches. The interactions of haematopoietic stem and progenitor cells (HSPCs) with cells, growth factors and extracellular matrix (ECM) components of the bone marrow (BM) microenvironment control homeostasis of HSPCs. We only start to understand the complexity of the haematopoietic niche(s) that comprises endosteal, arterial, sinusoidal, mesenchymal and neuronal components. These distinct niches produce a broad range of soluble factors and adhesion molecules that modulate HSPC fate during normal hematopoiesis and BM regeneration. Adhesive interactions between HSPCs and the microenvironment will influence their localization and differentiation potential. In this review we highlight the current understanding of the functional role of ECM- and adhesion (regulating) molecules in the haematopoietic niche during homeostatic and regenerative hematopoiesis. This knowledge may lead to the improvement of current cellular therapies and more efficient development of future cellular products.
Collapse
Affiliation(s)
- Sofieke Klamer
- a Department of Hematopoiesis; Sanquin Research; Landsteiner Laboratory; Academic Medical Centre ; University of Amsterdam ; Amsterdam , The Netherlands
| | | |
Collapse
|
29
|
Agas D, Marchetti L, Douni E, Sabbieti MG. The unbearable lightness of bone marrow homeostasis. Cytokine Growth Factor Rev 2015; 26:347-59. [DOI: 10.1016/j.cytogfr.2014.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/22/2014] [Accepted: 12/17/2014] [Indexed: 01/10/2023]
|
30
|
Kumar A, Saba JD. Regulation of Immune Cell Migration by Sphingosine-1-Phosphate. CELLULAR AND MOLECULAR BIOLOGY (OMICS) 2015; 61:121. [PMID: 30294722 PMCID: PMC6169313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Sphingosine-1-phosphate [S1P] is a potent bioactive sphingolipid molecule. In response to a stimulus, S1P is produced intracellularly by the action of two sphingosine kinases, and then it is exported to the extracellular environment or acts as an intracellular second messenger. S1P binds to its cognate G-protein coupled receptors, which are known as S1P receptors. There are five S1P receptors that have been identified in vertebrates. By activating S1P receptors, S1P controls a variety of physiological and pathological processes including cell migration, angiogenesis, vascular maturation, inflammation, and invasion, metastasis, and chemoresistance in cancer. S1P has emerged as a critical regulator of leukocyte migration and plays a central role in lymphocyte egress from the thymus and secondary lymphoid organs. In the current review article, we summarize the current understanding of the emigration of lymphocytes and other leukocytes from bone marrow, thymus and secondary lymphoid organs to the circulation, as well as the clinical implications of modulating the activity of the major S1P receptor, S1PR1.
Collapse
Affiliation(s)
- A. Kumar
- Department of Biochemistry, All India Institute of Medical Sciences [AIIMS], Saket Nagar, Bhopal 462 020, India
| | - JD. Saba
- Children’s Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, California 94609, USA
| |
Collapse
|
31
|
Zabel BA, Rott A, Butcher EC. Leukocyte chemoattractant receptors in human disease pathogenesis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2014; 10:51-81. [PMID: 25387059 DOI: 10.1146/annurev-pathol-012513-104640] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Combinations of leukocyte attractant ligands and cognate heptahelical receptors specify the systemic recruitment of circulating cells by triggering integrin-dependent adhesion to endothelial cells, supporting extravasation, and directing specific intratissue localization via gradient-driven chemotaxis. Chemoattractant receptors also control leukocyte egress from lymphoid organs and peripheral tissues. In this article, we summarize the fundamental mechanics of leukocyte trafficking, from the evolution of multistep models of leukocyte recruitment and navigation to the regulation of chemoattractant availability and function by atypical heptahelical receptors. To provide a more complete picture of the migratory circuits involved in leukocyte trafficking, we integrate a number of nonchemokine chemoattractant receptors into our discussion. Leukocyte chemoattractant receptors play key roles in the pathogenesis of autoimmune diseases, allergy, inflammatory disorders, and cancer. We review recent advances in our understanding of chemoattractant receptors in disease pathogenesis, with a focus on genome-wide association studies in humans and the translational implications of mechanistic studies in animal disease models.
Collapse
Affiliation(s)
- Brian A Zabel
- Palo Alto Veterans Institute for Research and Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304;
| | | | | |
Collapse
|
32
|
Blin-Wakkach C, Rouleau M, Wakkach A. Roles of osteoclasts in the control of medullary hematopoietic niches. Arch Biochem Biophys 2014; 561:29-37. [PMID: 24998177 DOI: 10.1016/j.abb.2014.06.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 06/23/2014] [Accepted: 06/25/2014] [Indexed: 12/23/2022]
Abstract
Bone marrow is the major site of hematopoiesis in mammals. The bone marrow environment plays an essential role in the regulation of hematopoietic stem and progenitor cells by providing specialized niches in which these cells are maintained. Many cell types participate to the composition and regulation of hematopoietic stem cell (HSC) niches, integrating complex signals from the bone, immune and nervous systems. Among these cells, the bone-resorbing osteoclasts (OCLs) have been described as main regulators of HSC niches. They are not limited to carving space for HSCs, but they also provide signals that affect the molecular and cellular niche components. However, their exact role in HSC niches remains unclear because of the variety of models, signals and conditions used to address the question. The present review will discuss the importance of the implication of OCLs focusing on the formation of HSC niches, the maintenance of HSCs in these niches and the mobilization of HSCs from the bone marrow. It will underline the importance of OCLs in HSC niches.
Collapse
Affiliation(s)
- Claudine Blin-Wakkach
- CNRS UMR7370, LP2M, Faculty of Medicine, 28 Av de Valombrose, 06107 Nice, France; University Nice Sophia Antipolis, Faculty of Sciences, Parc Valrose, 06100 Nice, France.
| | - Matthieu Rouleau
- CNRS UMR7370, LP2M, Faculty of Medicine, 28 Av de Valombrose, 06107 Nice, France; University Nice Sophia Antipolis, Faculty of Sciences, Parc Valrose, 06100 Nice, France
| | - Abdelilah Wakkach
- CNRS UMR7370, LP2M, Faculty of Medicine, 28 Av de Valombrose, 06107 Nice, France; University Nice Sophia Antipolis, Faculty of Sciences, Parc Valrose, 06100 Nice, France
| |
Collapse
|
33
|
Noels H, Zhou B, Tilstam PV, Theelen W, Li X, Pawig L, Schmitz C, Akhtar S, Simsekyilmaz S, Shagdarsuren E, Schober A, Adams RH, Bernhagen J, Liehn EA, Döring Y, Weber C. Deficiency of endothelial CXCR4 reduces reendothelialization and enhances neointimal hyperplasia after vascular injury in atherosclerosis-prone mice. Arterioscler Thromb Vasc Biol 2014; 34:1209-20. [PMID: 24723559 DOI: 10.1161/atvbaha.113.302878] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The Cxcl12/Cxcr4 chemokine ligand/receptor axis mediates the mobilization of smooth muscle cell progenitors, driving injury-induced neointimal hyperplasia. This study aimed to investigate the role of endothelial Cxcr4 in neointima formation. APPROACH AND RESULTS β-Galactosidase staining using bone marrow x kinase (Bmx)-CreER(T2) reporter mice and double immunofluorescence revealed an efficient and endothelial-specific deletion of Cxcr4 in Bmx-CreER(T2+) compared with Bmx-CreER(T2-) Cxcr4-floxed apolipoprotein E-deficient (Apoe(-/-)) mice (referred to as Cxcr4(EC-KO)ApoE(-/-) and Cxcr4(EC-WT) ApoE(-/-), respectively). Endothelial Cxcr4 deficiency significantly increased wire injury-induced neointima formation in carotid arteries from Cxcr4(EC-KO)ApoE(-/-) mice. The lesions displayed a higher number of macrophages, whereas the smooth muscle cell and collagen content were reduced. This was associated with a significant reduction in reendothelialization and endothelial cell proliferation in injured Cxcr4(EC-KO)ApoE(-/-) carotids compared with Cxcr4(EC-WT)ApoE(-/-) controls. Furthermore, stimulation of human aortic endothelial cells with chemokine (C-X-C motif) ligand 12 (CXCL12) significantly enhanced their wound-healing capacity in an in vitro scratch assay, an effect that could be reversed with the CXCR4 antagonist AMD3100. Also, flow cytometric analysis showed a reduced mobilization of Sca1(+)Flk1(+)Cd31(+) and of Lin(-)Sca1(+) progenitors in Cxcr4(EC-KO) ApoE(-/-) mice after vascular injury, although Cxcr4 surface expression was unaltered. No differences could be detected in plasma concentrations of Cxcl12, vascular endothelial growth factor, sphingosine 1-phosphate, or Flt3 (fms-related tyrosine kinase 3) ligand, all cytokines with an established role in progenitor cell mobilization. Nonetheless, double immunofluorescence revealed a significant reduction in local endothelial Cxcl12 staining in injured carotids from Cxcr4(EC-KO)ApoE(-/-) mice. CONCLUSIONS Endothelial Cxcr4 is crucial for efficient reendothelialization after vascular injury through endothelial wound healing and proliferation, and through the mobilization of Sca1(+)Flk1(+)Cd31(+) cells, often referred to as circulating endothelial progenitor cells.
Collapse
Affiliation(s)
- Heidi Noels
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.).
| | - Baixue Zhou
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Pathricia V Tilstam
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Wendy Theelen
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Xiaofeng Li
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Lukas Pawig
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Corinna Schmitz
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Shamima Akhtar
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Sakine Simsekyilmaz
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Erdenechimeg Shagdarsuren
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Andreas Schober
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Ralf H Adams
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Jürgen Bernhagen
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Elisa A Liehn
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Yvonne Döring
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Christian Weber
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.).
| |
Collapse
|
34
|
Targeting the molecular and cellular interactions of the bone marrow niche in immunologic disease. Curr Allergy Asthma Rep 2014; 14:402. [PMID: 24408534 DOI: 10.1007/s11882-013-0402-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recent investigations have expanded our knowledge of the regulatory bone marrow (BM) niche, which is critical in maintaining and directing hematopoietic stem cell (HSC) self-renewal and differentiation. Osteoblasts, mesenchymal stem cells (MSCs), and CXCL12-abundant reticular (CAR) cells are niche components in close association with HSCs and have been more clearly defined in immune cell function and homeostasis. Importantly, cellular inhabitants of the BM niche signal through G protein-coupled surface receptors (GPCRs) for various appropriate immune functions. In this article, recent literature on BM niche inhabitants (HSCs, osteoblasts, MSCs, CAR cells) and their GPCR mechanistic interactions are reviewed for better understanding of the BM cells involved in immune development, immunologic disease, and current immune reconstitution therapies.
Collapse
|