1
|
Ji S, Kim K, Park SJ, Lee JY, Jung HW, Yoo HJ, Jang IY, Lee E, Baek JY, Kim BJ. Higher Plasma Stromal Cell-Derived Factor 1 Is Associated with Lower Risk for Sarcopenia in Older Asian Adults. Endocrinol Metab (Seoul) 2023; 38:701-708. [PMID: 37849050 PMCID: PMC10764998 DOI: 10.3803/enm.2023.1783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/22/2023] [Accepted: 09/13/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGRUOUND Despite the protective effects of stromal cell-derived factor 1 (SDF-1) in stimulating muscle regeneration shown in experimental research, there is a lack of clinical studies linking circulating SDF-1 concentrations with muscle phenotypes. In order to elucidate the role of SDF-1 as a potential biomarker reflecting human muscle health, we investigated the association of plasma SDF-1 levels with sarcopenia in older adults. METHODS This cross-sectional study included 97 community-dwelling participants who underwent a comprehensive geriatric assessment at a tertiary hospital in South Korea. Sarcopenia was defined by specific cutoff values applicable to the Asian population, whereas plasma SDF-1 levels were determined using an enzyme immunoassay. RESULTS After accounting for sex, age, and body mass index, participants with sarcopenia and low muscle mass exhibited plasma SDF-1 levels that were 21.8% and 18.3% lower than those without these conditions, respectively (P=0.008 and P=0.009, respectively). Consistently, higher plasma SDF-1 levels exhibited a significant correlation with higher skeletal muscle mass index (SMI) and gait speed (both P=0.043), and the risk of sarcopenia and low muscle mass decreased by 58% and 55% per standard deviation increase in plasma SDF-1 levels, respectively (P=0.045 and P=0.030, respectively). Furthermore, participants in the highest SDF-1 tertile exhibited significantly higher SMI compared to those in the lowest tertile (P=0.012). CONCLUSION These findings clinically corroborate earlier experimental discoveries highlighting the muscle anabolic effects of SDF- 1 and support the potential role of circulating SDF-1 as a biomarker reflecting human muscle health in older adults.
Collapse
Affiliation(s)
- Sunghwan Ji
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyunggon Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Department of Digital Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - So Jeong Park
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin Young Lee
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hee-Won Jung
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyun Ju Yoo
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Il-Young Jang
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eunju Lee
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji Yeon Baek
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Beom-Jun Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
2
|
Jiang S, Luo M, Bai X, Nie P, Zhu Y, Cai H, Li B, Luo P. Cellular crosstalk of glomerular endothelial cells and podocytes in diabetic kidney disease. J Cell Commun Signal 2022; 16:313-331. [PMID: 35041192 DOI: 10.1007/s12079-021-00664-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetic kidney disease (DKD) is a serious microvascular complication of diabetes and is the leading cause of end-stage renal disease (ESRD). Persistent proteinuria is an important feature of DKD, which is caused by the destruction of the glomerular filtration barrier (GFB). Glomerular endothelial cells (GECs) and podocytes are important components of the GFB, and their damage can be observed in the early stages of DKD. Recently, studies have found that crosstalk between cells directly affects DKD progression, which has prospective research significance. However, the pathways involved are complex and largely unexplored. Here, we review the literature on cellular crosstalk of GECs and podocytes in the context of DKD, and highlight specific gaps in the field to propose future research directions. Elucidating the intricates of such complex processes will help to further understand the pathogenesis of DKD and develop better prevention and treatment options.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Manyu Luo
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Xue Bai
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Ping Nie
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Yuexin Zhu
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Hangxi Cai
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China
| | - Bing Li
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China.
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Changchun, 130041, China.
| |
Collapse
|
3
|
Malik S, Westcott JM, Brekken RA, Burrows FJ. CXCL12 in Pancreatic Cancer: Its Function and Potential as a Therapeutic Drug Target. Cancers (Basel) 2021; 14:cancers14010086. [PMID: 35008248 PMCID: PMC8750050 DOI: 10.3390/cancers14010086] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Pancreatic cancer is a challenging disease to treat effectively. Fibroblasts associated with pancreatic cancer contribute to disease progression by secreting factors that enhance tumor cell survival and help tumor cells avoid detection by the immune system. This overview focuses on a chemokine, CXCL12, produced by cancer-associated fibroblasts and how CXCL12 signaling enhances pancreatic cancer progression by contributing to various hallmarks of cancer including, but not limited to, tumor growth and evasion of immune response. These pro-oncogenic functions of CXCL12 make it an attractive target in pancreatic cancer. We discuss the different approaches in development to therapeutically target CXCL12 and finally propose a novel approach, the use of the farnesyl transferase inhibitor tipifarnib to inhibit CXCL12 expression in pancreatic fibroblasts. Abstract Pancreatic ductal adenocarcinoma (PDAC) is a disease with limited therapeutic options and dismal long-term survival. The unique tumor environment of PDAC, consisting of desmoplastic stroma, immune suppressive cells, and activated fibroblasts, contributes to its resistance to therapy. Activated fibroblasts (cancer-associated fibroblasts and pancreatic stellate cells) secrete chemokines and growth factors that support PDAC growth, spread, chemoresistance, and immune evasion. In this review, we focus on one such chemokine, CXCL12, secreted by the cancer-associated fibroblasts and discuss its contribution to several of the classical hallmarks of PDAC and other tumors. We review the various therapeutic approaches in development to target CXCL12 signaling in PDAC. Finally, we propose an unconventional use of tipifarnib, a farnesyl transferase inhibitor, to inhibit CXCL12 production in PDAC.
Collapse
Affiliation(s)
| | - Jill M. Westcott
- Division of Surgical Oncology, Department of Surgery, and Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Rolf A. Brekken
- Division of Surgical Oncology, Department of Surgery, and Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Correspondence: (R.A.B.); (F.J.B.)
| | - Francis J. Burrows
- Kura Oncology, Inc., San Diego, CA 92130, USA;
- Correspondence: (R.A.B.); (F.J.B.)
| |
Collapse
|
4
|
Lai X, Chen J. C-X-C motif chemokine ligand 12: a potential therapeutic target in Duchenne muscular dystrophy. Bioengineered 2021; 12:5428-5439. [PMID: 34424816 PMCID: PMC8806931 DOI: 10.1080/21655979.2021.1967029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by a mutant dystrophin protein. DMD patients undergo gradual progressive paralysis until death. Chronic glucocorticoid therapy remains one of the main treatments for DMD, despite the significant side effects. However, its mechanisms of action remain largely unknown. We used bioinformatics tools to identify pathogenic genes involved in DMD and glucocorticoid target genes. Two gene expression profiles containing data from DMD patients and healthy controls (GSE38417 and GSE109178) were downloaded for further analysis. Differentially expressed genes (DEGs) between DMD patients and controls were identified using GEO2R, and glucocorticoid target genes were predicted from the Pharmacogenetics and Pharmacogenomics Knowledge Base. Surprisingly, only one gene, CXCL12 (C-X-C motif chemokine ligand 12), was both a glucocorticoid target and a DEG. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, Gene Ontology term enrichment analysis, and gene set enrichment analysis were performed. A protein-protein interaction network was constructed and hub genes identified using the Search Tool for the Retrieval of Interacting Genes (STRING) database and Cytoscape. Enriched pathways involving the DEGs, including CXCL12, were associated with the immune response and inflammation. Levels of CXCL12 and its receptor CXCR4 (C-X-C motif chemokine receptor 4) were increased in X-linked muscular dystrophy (mdx) mice (DMD models) but became significantly reduced after prednisone treatment. Metformin also reduced the expression of CXCL12 and CXCR4 in mdx mice. In conclusion, the CXCL12-CXCR4 pathway may be a potential target for DMD therapy.
Collapse
Affiliation(s)
- Xinsheng Lai
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Jie Chen
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
5
|
Nitahara-Kasahara Y, Kuraoka M, Oda Y, Hayashita-Kinoh H, Takeda S, Okada T. Enhanced cell survival and therapeutic benefits of IL-10-expressing multipotent mesenchymal stromal cells for muscular dystrophy. Stem Cell Res Ther 2021; 12:105. [PMID: 33541428 PMCID: PMC7860619 DOI: 10.1186/s13287-021-02168-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/14/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Multipotent mesenchymal stromal cells (MSCs) are potentially therapeutic for muscle disease because they can accumulate at the sites of injury and act as immunosuppressants. MSCs are attractive candidates for cell-based strategies that target diseases with chronic inflammation, such as Duchenne muscular disease (DMD). We focused on the anti-inflammatory properties of IL-10 and hypothesized that IL-10 could increase the typically low survival of MSCs by exerting a paracrine effect after transplantation. METHODS We developed a continuous IL-10 expression system of MSCs using an adeno-associated virus (AAV) vector. To investigate the potential benefits of IL-10 expressing AAV vector-transduced MSCs (IL-10-MSCs), we examined the cell survival rates in the skeletal muscles after intramuscular injection into mice and dogs. Systemic treatment with IL-10-MSCs derived from dental pulp (DPSCs) was comprehensively analyzed using the canine X-linked muscular dystrophy model in Japan (CXMDJ), which has a severe phenotype similar to that of DMD patients. RESULTS In vivo bioluminescence imaging analysis revealed higher retention of IL-10-MSCs injected into the hindlimb muscle of mice. In the muscles of dogs, myofiber-like tissue was formed after the stable engraftment of IL-10-MSCs. Repeated systemic administration of IL-10-DPSCs into the CXMDJ model resulted in long-term engraftment of cells and slightly increased the serum levels of IL-10. IL-10-hDPSCs showed significantly reduced expression of pro-inflammatory MCP-1 and upregulation of stromal-derived factor-1 (SDF-1). MRI and histopathology of the hDPSC-treated CXMDJ indicated the regulation of inflammation in the muscles, but not myogenic differentiation from treated cells. hDPSC-treated CXMDJ showed improved running capability and recovery in tetanic force with concomitant increase in physical activity. Serum creatine kinase levels, which increased immediately after exercise, were suppressed in IL-10-hDPSC-treated CXMDJ. CONCLUSIONS In case of local injection, IL-10-MSCs could maintain the long-term engraftment status and facilitate associated tissue repair. In case of repeated systemic administration, IL-10-MSCs facilitated the long-term retention of the cells in the skeletal muscle and also protected muscles from physical damage-induced injury, which improved muscle dysfunction in DMD. We can conclude that the local and systemic administration of IL-10-producing MSCs offers potential benefits for DMD therapy through the beneficial paracrine effects of IL-10 involving SDF-1.
Collapse
Affiliation(s)
- Yuko Nitahara-Kasahara
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Bunkyo City, Tokyo, Japan. .,Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo City, Tokyo, Japan. .,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.
| | - Mutsuki Kuraoka
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Laboratory of Experimental Animal Science, Nippon Veterinary and Life Science University, Musashino, Tokyo, Japan
| | - Yuki Oda
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Bunkyo City, Tokyo, Japan
| | - Hiromi Hayashita-Kinoh
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Bunkyo City, Tokyo, Japan.,Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo City, Tokyo, Japan.,Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, 108-8639, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Takashi Okada
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Bunkyo City, Tokyo, Japan. .,Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo City, Tokyo, Japan. .,Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, 108-8639, Japan.
| |
Collapse
|
6
|
Puchert M, Koch C, Zieger K, Engele J. Identification of CXCL11 as part of chemokine network controlling skeletal muscle development. Cell Tissue Res 2021; 384:499-511. [PMID: 33502606 PMCID: PMC8141492 DOI: 10.1007/s00441-020-03398-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 12/11/2020] [Indexed: 11/09/2022]
Abstract
The chemokine, CXCL12, and its receptors, CXCR4 and CXCR7, play pivotal roles during development and maintenance of limb muscles. CXCR7 additionally binds CXCL11, which uses CXCR3 as its prime receptor. Based on this cross-talk, we investigate whether CXCL11 would likewise affect development and/or function of skeletal muscles. Western blotting and immunolabelling demonstrated the developmentally restricted expression of CXCL11 in rat limb muscles, which was contrasted by the continuous expression of its receptors in proliferating and differentiating C2C12 cells as well as in late embryonic to adult rat limb muscle fibres. Consistent with a prime role in muscle formation, functional studies identified CXCL11 as a potent chemoattractant for undifferentiated C2C12 cells and further showed that CXCL11 does neither affect myoblast proliferation and differentiation nor metabolic/catabolic pathways in formed myotubes. The use of selective receptor antagonists unravelled complementary effects of CXCL11 and CXCL12 on C2C12 cell migration, which either require CXCR3/CXCR7 or CXCR4, respectively. Our findings provide new insights into the chemokine network controlling skeletal muscle development and function and, thus, might provide a base for future therapies of muscular diseases.
Collapse
Affiliation(s)
- Malte Puchert
- Institute of Anatomy, Medical Faculty, University of Leipzig, Liebigstr.13, 04103, Leipzig, Germany
| | - Christian Koch
- Institute of Anatomy, Medical Faculty, University of Leipzig, Liebigstr.13, 04103, Leipzig, Germany
| | - Konstanze Zieger
- Institute of Anatomy, Medical Faculty, University of Leipzig, Liebigstr.13, 04103, Leipzig, Germany
| | - Jürgen Engele
- Institute of Anatomy, Medical Faculty, University of Leipzig, Liebigstr.13, 04103, Leipzig, Germany.
| |
Collapse
|
7
|
Taatjes DJ, Roth J. In focus in HCB. Histochem Cell Biol 2021; 155:1-8. [PMID: 33469707 DOI: 10.1007/s00418-020-01958-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2020] [Indexed: 10/22/2022]
Affiliation(s)
- Douglas J Taatjes
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA.
| | - Jürgen Roth
- University of Zurich, 8091, Zurich, Switzerland
| |
Collapse
|
8
|
Yahya I, Morosan-Puopolo G, Brand-Saberi B. The CXCR4/SDF-1 Axis in the Development of Facial Expression and Non-somitic Neck Muscles. Front Cell Dev Biol 2020; 8:615264. [PMID: 33415110 PMCID: PMC7783292 DOI: 10.3389/fcell.2020.615264] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/04/2020] [Indexed: 12/26/2022] Open
Abstract
Trunk and head muscles originate from distinct embryonic regions: while the trunk muscles derive from the paraxial mesoderm that becomes segmented into somites, the majority of head muscles develops from the unsegmented cranial paraxial mesoderm. Differences in the molecular control of trunk versus head and neck muscles have been discovered about 25 years ago; interestingly, differences in satellite cell subpopulations were also described more recently. Specifically, the satellite cells of the facial expression muscles share properties with heart muscle. In adult vertebrates, neck muscles span the transition zone between head and trunk. Mastication and facial expression muscles derive from the mesodermal progenitor cells that are located in the first and second branchial arches, respectively. The cucullaris muscle (non-somitic neck muscle) originates from the posterior-most branchial arches. Like other subclasses within the chemokines and chemokine receptors, CXCR4 and SDF-1 play essential roles in the migration of cells within a number of various tissues during development. CXCR4 as receptor together with its ligand SDF-1 have mainly been described to regulate the migration of the trunk muscle progenitor cells. This review first underlines our recent understanding of the development of the facial expression (second arch-derived) muscles, focusing on new insights into the migration event and how this embryonic process is different from the development of mastication (first arch-derived) muscles. Other muscles associated with the head, such as non-somitic neck muscles derived from muscle progenitor cells located in the posterior branchial arches, are also in the focus of this review. Implications on human muscle dystrophies affecting the muscles of face and neck are also discussed.
Collapse
Affiliation(s)
- Imadeldin Yahya
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany.,Department of Anatomy, Faculty of Veterinary Medicine, University of Khartoum, Khartoum, Sudan
| | | | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
9
|
Yahya I, Böing M, Brand-Saberi B, Morosan-Puopolo G. How to distinguish between different cell lineages sharing common markers using combinations of double in-situ-hybridization and immunostaining in avian embryos: CXCR4-positive mesodermal and neural crest-derived cells. Histochem Cell Biol 2020; 155:145-155. [PMID: 33037504 PMCID: PMC7847855 DOI: 10.1007/s00418-020-01920-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2020] [Indexed: 11/25/2022]
Abstract
Cell migration plays a crucial role in early embryonic development. The chemokine receptor CXCR4 has been reported to guide migration of neural crest cells (NCCs) to form the dorsal root ganglia (DRG) and sympathetic ganglia (SG). CXCR4 also plays an important part during the formation of limb and cloacal muscles. NCCs migration and muscle formation during embryonic development are usually considered separately, although both cell lineages migrate in close neighbourhood and have markers in common. In this study, we present a new method for the simultaneous detection of CXCR4, mesodermal markers and NCCs markers during chicken embryo developmental stages HH18–HH25 by combining double whole-mount in situ hybridization (ISH) and immunostaining on floating vibratome sections. The simultaneous detection of CXCR4 and markers for the mesodermal and neural crest cells in multiple labelling allowed us to compare complex gene expression patterns and it could be easily used for a wide range of gene expression pattern analyses of other chicken embryonic tissues. All steps of the procedure, including the preparation of probes and embryos, prehybridization, hybridization, visualization of the double labelled transcripts and immunostaining, are described in detail.
Collapse
Affiliation(s)
- Imadeldin Yahya
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
- Department of Anatomy, Faculty of Veterinary Medicine, University of Khartoum, Khartoum, Sudan
| | - Marion Böing
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Gabriela Morosan-Puopolo
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
10
|
Exercise-Induced Myokines can Explain the Importance of Physical Activity in the Elderly: An Overview. Healthcare (Basel) 2020; 8:healthcare8040378. [PMID: 33019579 PMCID: PMC7712334 DOI: 10.3390/healthcare8040378] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/12/2022] Open
Abstract
Physical activity has been found to aid the maintenance of health in the elderly. Exercise-induced skeletal muscle contractions lead to the production and secretion of many small proteins and proteoglycan peptides called myokines. Thus, studies on myokines are necessary for ensuring the maintenance of skeletal muscle health in the elderly. This review summarizes 13 myokines regulated by physical activity that are affected by aging and aims to understand their potential roles in metabolic diseases. We categorized myokines into two groups based on regulation by aerobic and anaerobic exercise. With aging, the secretion of apelin, β-aminoisobutyric acid (BAIBA), bone morphogenetic protein 7 (BMP-7), decorin, insulin-like growth factor 1 (IGF-1), interleukin-15 (IL-15), irisin, stromal cell-derived factor 1 (SDF-1), sestrin, secreted protein acidic rich in cysteine (SPARC), and vascular endothelial growth factor A (VEGF-A) decreased, while that of IL-6 and myostatin increased. Aerobic exercise upregulates apelin, BAIBA, IL-15, IL-6, irisin, SDF-1, sestrin, SPARC, and VEGF-A expression, while anaerobic exercise upregulates BMP-7, decorin, IGF-1, IL-15, IL-6, irisin, and VEGF-A expression. Myostatin is downregulated by both aerobic and anaerobic exercise. This review provides a rationale for developing exercise programs or interventions that maintain a balance between aerobic and anaerobic exercise in the elderly.
Collapse
|
11
|
El-Saie A, Shivanna B. Novel Strategies to Reduce Pulmonary Hypertension in Infants With Bronchopulmonary Dysplasia. Front Pediatr 2020; 8:201. [PMID: 32457857 PMCID: PMC7225259 DOI: 10.3389/fped.2020.00201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/02/2020] [Indexed: 01/10/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a developmental lung disorder of preterm infants primarily caused by the failure of host defense mechanisms to prevent tissue injury and facilitate repair. This disorder is the most common complication of premature birth, and its incidence remains unchanged over the past few decades. Additionally, BPD increases long-term cardiopulmonary and neurodevelopmental morbidities of preterm infants. Pulmonary hypertension (PH) is a common morbidity of BPD. Importantly, the presence of PH increases both the short- and long-term morbidities and mortality in BPD infants. Further, there are no curative therapies for this complex disease. Besides providing an overview of the pathogenesis and diagnosis of PH associated with BPD, we have attempted to comprehensively review and summarize the current literature on the interventions to prevent and/or mitigate BPD and PH in preclinical studies. Our goal was to provide insight into the therapies that have a high translational potential to meaningfully manage BPD patients with PH.
Collapse
Affiliation(s)
- Ahmed El-Saie
- Department of Pediatrics, Section of Neonatology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Binoy Shivanna
- Department of Pediatrics, Section of Neonatology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
12
|
Kasprzycka P, Archacka K, Kowalski K, Mierzejewski B, Zimowska M, Grabowska I, Piotrowski M, Rafałko M, Ryżko A, Irhashava A, Senderowski K, Gołąbek M, Stremińska W, Jańczyk-Ilach K, Koblowska M, Iwanicka-Nowicka R, Fogtman A, Janowski M, Walczak P, Ciemerych MA, Brzoska E. The factors present in regenerating muscles impact bone marrow-derived mesenchymal stromal/stem cell fusion with myoblasts. Stem Cell Res Ther 2019; 10:343. [PMID: 31753006 PMCID: PMC6873517 DOI: 10.1186/s13287-019-1444-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/23/2019] [Accepted: 10/04/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Satellite cells, a population of unipotent stem cells attached to muscle fibers, determine the excellent regenerative capability of injured skeletal muscles. Myogenic potential is also exhibited by other cell populations, which exist in the skeletal muscles or come from other niches. Mesenchymal stromal/stem cells inhabiting the bone marrow do not spontaneously differentiate into muscle cells, but there is some evidence that they are capable to follow the myogenic program and/or fuse with myoblasts. METHODS In the present study we analyzed whether IGF-1, IL-4, IL-6, and SDF-1 could impact human and porcine bone marrow-derived mesenchymal stromal/stem cells (hBM-MSCs and pBM-MSCs) and induce expression of myogenic regulatory factors, skeletal muscle-specific structural, and adhesion proteins. Moreover, we investigated whether these factors could induce both types of BM-MSCs to fuse with myoblasts. IGF-1, IL-4, IL-6, and SDF-1 were selected on the basis of their role in embryonic myogenesis as well as skeletal muscle regeneration. RESULTS We found that hBM-MSCs and pBM-MSCs cultured in vitro in the presence of IGF-1, IL-4, IL-6, or SDF-1 did not upregulate myogenic regulatory factors. Consequently, we confirmed the lack of their naïve myogenic potential. However, we noticed that IL-4 and IL-6 impacted proliferation and IL-4, IL-6, and SDF-1 improved migration of hBM-MSCs. IL-4 treatment resulted in the significant increase in the level of mRNA encoding CD9, NCAM, VCAM, and m-cadherin, i.e., proteins engaged in cell fusion during myotube formation. Additionally, the CD9 expression level was also driven by IGF-1 treatment. Furthermore, the pre-treatment of hBM-MSCs either with IGF-1, IL-4, or SDF-1 and treatment of pBM-MSCs either with IGF-1 or IL-4 increased the efficacy of hybrid myotube formation between these cells and C2C12 myoblasts. CONCLUSIONS To conclude, our study revealed that treatment with IGF-1, IL-4, IL-6, or SDF-1 affects BM-MSC interaction with myoblasts; however, it does not directly promote myogenic differentiation of these cells.
Collapse
Affiliation(s)
- Paulina Kasprzycka
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Karolina Archacka
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Kamil Kowalski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Bartosz Mierzejewski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Małgorzata Zimowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Mariusz Piotrowski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Milena Rafałko
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Agata Ryżko
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Aliksandra Irhashava
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Kamil Senderowski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Magdalena Gołąbek
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Władysława Stremińska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Katarzyna Jańczyk-Ilach
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Marta Koblowska
- Laboratory of Systems Biology, Faculty of Biology, University of Warsaw, Pawinskiego 5a St, 02-106 Warsaw, Poland
| | - Roksana Iwanicka-Nowicka
- Laboratory of Systems Biology, Faculty of Biology, University of Warsaw, Pawinskiego 5a St, 02-106 Warsaw, Poland
- Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a St, 02-106 Warsaw, Poland
| | - Anna Fogtman
- Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a St, 02-106 Warsaw, Poland
| | - Mirosław Janowski
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5 St, 02-106 Warsaw, Poland
| | - Piotr Walczak
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-719 Olsztyn, Poland
- Institute for Cell Engineering, Cellular Imaging Section, The Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Maria A. Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| |
Collapse
|
13
|
Guerra K, Bryan C, Dapaah-Siakwan F, Sammour I, Drummond S, Zambrano R, Chen P, Huang J, Sharma M, Shrager S, Benny M, Wu S, Young KC. Intra-tracheal administration of a naked plasmid expressing stromal derived factor-1 improves lung structure in rodents with experimental bronchopulmonary dysplasia. Respir Res 2019; 20:255. [PMID: 31718614 PMCID: PMC6852969 DOI: 10.1186/s12931-019-1224-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 10/29/2019] [Indexed: 12/15/2022] Open
Abstract
Background Bronchopulmonary dysplasia (BPD) is characterized by alveolar simplification and disordered angiogenesis. Stromal derived factor-1 (SDF-1) is a chemokine which modulates cell migration, proliferation, and angiogenesis. Here we tested the hypothesis that intra-tracheal (IT) administration of a naked plasmid DNA expressing SDF-1 would attenuate neonatal hyperoxia-induced lung injury in an experimental model of BPD, by promoting angiogenesis. Design/methods Newborn Sprague-Dawley rat pups (n = 18–20/group) exposed to room air (RA) or hyperoxia (85% O2) from postnatal day (P) 1 to 14 were randomly assigned to receive IT a naked plasmid expressing SDF-1, JVS-100 (Juventas Therapeutics, Cleveland, Ohio) or placebo (PL) on P3. Lung alveolarization, angiogenesis, inflammation, vascular remodeling and pulmonary hypertension (PH) were assessed on P14. PH was determined by measuring right ventricular systolic pressure (RVSP) and the weight ratio of the right to left ventricle + septum (RV/LV + S). Capillary tube formation in SDF-1 treated hyperoxia-exposed human pulmonary microvascular endothelial cells (HPMEC) was determined by matrigel assay. Data is expressed as mean ± SD and analyzed by two-way ANOVA. Results Exposure of neonatal pups to 14 days of hyperoxia decreased lung SDF-1 gene expression. Moreover, whilst hyperoxia exposure inhibited capillary tube formation in HPMEC, SDF-1 treatment increased tube length and branching in HPMEC. PL-treated hyperoxia-exposed pups had decreased alveolarization and lung vascular density. This was accompanied by an increase in RVSP, RV/LV + S, pulmonary vascular remodeling and inflammation. In contrast, IT JVS-100 improved lung structure, reduced inflammation, PH and vascular remodeling. Conclusions Intratracheal administration of a naked plasmid expressing SDF-1 improves alveolar and vascular structure in an experimental model of BPD. These findings suggest that therapies which modulate lung SDF-1 expression may have beneficial effects in preterm infants with BPD.
Collapse
Affiliation(s)
- Kasonya Guerra
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Carleene Bryan
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Frederick Dapaah-Siakwan
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Ibrahim Sammour
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Shelly Drummond
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Ronald Zambrano
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Pingping Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Jian Huang
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Mayank Sharma
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Sebastian Shrager
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Merline Benny
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Shu Wu
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Karen C Young
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA. .,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA. .,The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.
| |
Collapse
|
14
|
Nord H, Dennhag N, Tydinger H, von Hofsten J. The zebrafish HGF receptor met controls migration of myogenic progenitor cells in appendicular development. PLoS One 2019; 14:e0219259. [PMID: 31287821 PMCID: PMC6615617 DOI: 10.1371/journal.pone.0219259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022] Open
Abstract
The hepatocyte growth factor receptor C-met plays an important role in cellular migration, which is crucial for many developmental processes as well as for cancer cell metastasis. C-met has been linked to the development of mammalian appendicular muscle, which are derived from migrating muscle progenitor cells (MMPs) from within the somite. Mammalian limbs are homologous to the teleost pectoral and pelvic fins. In this study we used Crispr/Cas9 to mutate the zebrafish met gene and found that the MMP derived musculature of the paired appendages was severely affected. The mutation resulted in a reduced muscle fibre number, in particular in the pectoral abductor, and in a disturbed pectoral fin function. Other MMP derived muscles, such as the sternohyoid muscle and posterior hypaxial muscle were also affected in met mutants. This indicates that the role of met in MMP function and appendicular myogenesis is conserved within vertebrates.
Collapse
Affiliation(s)
- Hanna Nord
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Nils Dennhag
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Hanna Tydinger
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Jonas von Hofsten
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- Umeå Centre for Molecular Medicine, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
15
|
Dissecting cell diversity and connectivity in skeletal muscle for myogenesis. Cell Death Dis 2019; 10:427. [PMID: 31160550 PMCID: PMC6546706 DOI: 10.1038/s41419-019-1647-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 03/25/2019] [Accepted: 05/06/2019] [Indexed: 12/28/2022]
Abstract
Characterized by their slow adhering property, skeletal muscle myogenic progenitor cells (MPCs) have been widely utilized in skeletal muscle tissue engineering for muscle regeneration, but with limited efficacy. Skeletal muscle regeneration is regulated by various cell types, including a large number of rapidly adhering cells (RACs) where their functions and mechanisms are still unclear. In this study, we explored the function of RACs by co-culturing them with MPCs in a biomimetic skeletal muscle organoid system. Results showed that RACs promoted the myogenic potential of MPCs in the organoid. Single-cell RNA-Seq was also performed, classifying RACs into 7 cell subtypes, including one newly described cell subtype: teno-muscular cells (TMCs). Connectivity map of RACs and MPCs subpopulations revealed potential growth factors (VEGFA and HBEGF) and extracellular matrix (ECM) proteins involvement in the promotion of myogenesis of MPCs during muscle organoid formation. Finally, trans-well experiments and small molecular inhibitors blocking experiments confirmed the role of RACs in the promotion of myogenic differentiation of MPCs. The RACs reported here revealed complex cell diversity and connectivity with MPCs in the biomimetic skeletal muscle organoid system, which not only offers an attractive alternative for disease modeling and in vitro drug screening but also provides clues for in vivo muscle regeneration.
Collapse
|
16
|
Liu L, Fu Y, Zhu F, Mu C, Li R, Song W, Shi C, Ye Y, Wang C. Transcriptomic analysis of Portunus trituberculatus reveals a critical role for WNT4 and WNT signalling in limb regeneration. Gene 2018. [PMID: 29524579 DOI: 10.1016/j.gene.2018.03.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The swimming crab (Portunus trituberculatus) is among the most economically important seawater crustacean species in Asia. Despite its commercial importance and being well-studied status, genomic and transcriptomic data are scarce for this crab species. In the present study, limb bud tissue was collected at different developmental stages post amputation for transcriptomic analysis. Illumina RNA-sequencing was applied to characterise the limb regeneration transcriptome and identify the most characteristic genes. A total of 289,018 transcripts were obtained by clustering and assembly of clean reads, producing 150,869 unigenes with an average length of 956 bp. Subsequent analysis revealed WNT signalling as the key pathway involved in limb regeneration, with WNT4 a key mediator. Overall, limb regeneration appears to be regulated by multiple signalling pathways, with numerous cell differentiation, muscle growth, moult, metabolism, and immune-related genes upregulated, including WNT4, LAMA, FIP2, FSTL5, TNC, HUS1, SWI5, NCGL, SLC22, PLA2, Tdc2, SMOX, GDH, and SMPD4. This is the first experimental study done on regenerating claws of P. trituberculatus. These findings expand existing sequence resources for crab species, and will likely accelerate research into regeneration and development in crustaceans, particularly functional studies on genes involved in limb regeneration.
Collapse
Affiliation(s)
- Lei Liu
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo 315211, China
| | - Yuanyuan Fu
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo 315211, China
| | - Fang Zhu
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo 315211, China
| | - Changkao Mu
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo 315211, China
| | - Ronghua Li
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo 315211, China
| | - Weiwei Song
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo 315211, China
| | - Ce Shi
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo 315211, China
| | - Yangfang Ye
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo 315211, China
| | - Chunlin Wang
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
17
|
Heinze B, Fuss CT, Mulatero P, Beuschlein F, Reincke M, Mustafa M, Schirbel A, Deutschbein T, Williams TA, Rhayem Y, Quinkler M, Rayes N, Monticone S, Wild V, Gomez-Sanchez CE, Reis AC, Petersenn S, Wester HJ, Kropf S, Fassnacht M, Lang K, Herrmann K, Buck AK, Bluemel C, Hahner S. Targeting CXCR4 (CXC Chemokine Receptor Type 4) for Molecular Imaging of Aldosterone-Producing Adenoma. Hypertension 2017; 71:317-325. [PMID: 29279316 DOI: 10.1161/hypertensionaha.117.09975] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 07/25/2017] [Accepted: 11/28/2017] [Indexed: 12/20/2022]
Abstract
Primary aldosteronism is the most frequent cause of secondary hypertension and is associated with increased morbidity and mortality compared with hypertensive controls. The central diagnostic challenge is the differentiation between bilateral and unilateral disease, which determines treatment options. Bilateral adrenal venous sampling, currently recommended for differential diagnosis, is an invasive procedure with several drawbacks, making it desirable to develop novel noninvasive diagnostic tools. When investigating the expression pattern of chemokine receptors by quantitative real-time polymerase chain reaction and immunohistochemistry, we observed high expression of CXCR4 (CXC chemokine receptor type 4) in aldosterone-producing tissue in normal adrenals, adjacent adrenal cortex from adrenocortical adenomas, and in aldosterone-producing adenomas (APA), correlating strongly with the expression of CYP11B2 (aldosterone synthase). In contrast, CXCR4 was not detected in the majority of nonfunctioning adenomas that are frequently found coincidently. The specific CXCR4 ligand 68Ga-pentixafor has recently been established as radiotracer for molecular imaging of CXCR4 expression and showed strong and specific binding to cryosections of APAs in our study. We further investigated 9 patients with primary aldosteronism because of APA by 68Ga-pentixafor-positron emission tomography. The tracer uptake was significantly higher on the side of increased adrenocortical aldosterone secretion in patients with APAs compared with patients investigated by 68Ga-pentixafor-positron emission tomography for other causes. Molecular imaging of aldosterone-producing tissue by a CXCR4-specific ligand may, therefore, be a highly promising tool for noninvasive characterization of patients with APAs.
Collapse
Affiliation(s)
- Britta Heinze
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Carmina T Fuss
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Paolo Mulatero
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Felix Beuschlein
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Martin Reincke
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Mona Mustafa
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Andreas Schirbel
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Timo Deutschbein
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Tracy Ann Williams
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Yara Rhayem
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Marcus Quinkler
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Nada Rayes
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Silvia Monticone
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Vanessa Wild
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Celso E Gomez-Sanchez
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Anna-Carinna Reis
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Stephan Petersenn
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Hans-Juergen Wester
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Saskia Kropf
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Martin Fassnacht
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Katharina Lang
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Ken Herrmann
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Andreas K Buck
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Christina Bluemel
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.)
| | - Stefanie Hahner
- From the Department of Internal Medicine I, Endocrinology and Diabetes Unit (B.H., C.T.F., M.F., K.L., S.H.), Department of Nuclear Medicine (A.S., K.H., A.K.B., C.B.), and Comprehensive Cancer Center Wuerzburg (T.D., M.F.), University Hospital of Wuerzburg, University of Wuerzburg, Germany; Division of Internal Medicine and Hypertension, Department of Medical Sciences, University of Torino, Italy (P.M., T.A.W., S.M.); Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Germany (F.B., M.R., T.A.W., Y.R.); Department of Nuclear Medicine, Klinikum rechts der Isar der Technischen Universität München, Germany (M.M.); Endocrinology in Charlottenburg, Berlin, Germany (M.Q.); Department of General, Visceral, and Transplant Surgery, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Germany (N.R.); Department of Pathology, University of Würzburg, Germany (V.W.); Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, MS (C.E.G.-S.); Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany (A.-C.R.); ENDOC, Center for Endocrine Tumors, Hamburg, Germany (S.P.); Pharmaceutical Radiochemistry, Technische Universität München, Garching bei München, Germany (H.-J.W.); and Scintomics GmbH, Fürstenfeldbruck, Germany (S.K.).
| |
Collapse
|
18
|
Odd skipped-related 1 identifies a population of embryonic fibro-adipogenic progenitors regulating myogenesis during limb development. Nat Commun 2017; 8:1218. [PMID: 29084951 PMCID: PMC5662571 DOI: 10.1038/s41467-017-01120-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 08/17/2017] [Indexed: 12/31/2022] Open
Abstract
Fibro-adipogenic progenitors (FAPs) are an interstitial cell population in adult skeletal muscle that support muscle regeneration. During development, interstitial muscle connective tissue (MCT) cells support proper muscle patterning, however the underlying molecular mechanisms are not well understood and it remains unclear whether adult FAPs and embryonic MCT cells share a common lineage. We show here that mouse embryonic limb MCT cells expressing the transcription factor Osr1, differentiate into fibrogenic and adipogenic cells in vivo and in vitro defining an embryonic FAP-like population. Genetic lineage tracing shows that developmental Osr1+ cells give rise to a subset of adult FAPs. Loss of Osr1 function leads to a reduction of myogenic progenitor proliferation and survival resulting in limb muscle patterning defects. Transcriptome and functional analyses reveal that Osr1+ cells provide a critical pro-myogenic niche via the production of MCT specific extracellular matrix components and secreted signaling factors. Fibro-adipogenic progenitors (FAPs) form part of interstitial muscle connective tissue (MCT) in adults but the origin of this non-myogenic lineage is unclear. Here, the authors show that Odd skipped related 1 (Osr1) in mice marks embryonic MCT, giving rise to FAPs, and loss of Osr1 in the limb causes muscle defects.
Collapse
|
19
|
Thakar D, Dalonneau F, Migliorini E, Lortat-Jacob H, Boturyn D, Albiges-Rizo C, Coche-Guerente L, Picart C, Richter RP. Binding of the chemokine CXCL12α to its natural extracellular matrix ligand heparan sulfate enables myoblast adhesion and facilitates cell motility. Biomaterials 2017; 123:24-38. [PMID: 28152381 PMCID: PMC5405871 DOI: 10.1016/j.biomaterials.2017.01.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/04/2017] [Accepted: 01/17/2017] [Indexed: 01/24/2023]
Abstract
The chemokine CXCL12α is a potent chemoattractant that guides the migration of muscle precursor cells (myoblasts) during myogenesis and muscle regeneration. To study how the molecular presentation of chemokines influences myoblast adhesion and motility, we designed multifunctional biomimetic surfaces as a tuneable signalling platform that enabled the response of myoblasts to selected extracellular cues to be studied in a well-defined environment. Using this platform, we demonstrate that CXCL12α, when presented by its natural extracellular matrix ligand heparan sulfate (HS), enables the adhesion and spreading of myoblasts and facilitates their active migration. In contrast, myoblasts also adhered and spread on CXCL12α that was quasi-irreversibly surface-bound in the absence of HS, but were essentially immotile. Moreover, co-presentation of the cyclic RGD peptide as integrin ligand along with HS-bound CXCL12α led to enhanced spreading and motility, in a way that indicates cooperation between CXCR4 (the CXCL12α receptor) and integrins (the RGD receptors). Our findings reveal the critical role of HS in CXCL12α induced myoblast adhesion and migration. The biomimetic surfaces developed here hold promise for mechanistic studies of cellular responses to different presentations of biomolecules. They may be broadly applicable for dissecting the signalling pathways underlying receptor cross-talks, and thus may guide the development of novel biomaterials that promote highly specific cellular responses.
Collapse
Affiliation(s)
- Dhruv Thakar
- Université Grenoble Alpes, Département de Chimie Moléculaire (DCM), Grenoble, France; CNRS, DCM, Grenoble, France
| | - Fabien Dalonneau
- CNRS UMR 5628 (LMGP), Grenoble, France; Grenoble Institute of Technology, Université Grenoble Alpes, LMGP, Grenoble, France
| | - Elisa Migliorini
- Université Grenoble Alpes, Département de Chimie Moléculaire (DCM), Grenoble, France; CNRS, DCM, Grenoble, France
| | - Hugues Lortat-Jacob
- Institut de Biologie Structurale, UMR 5075, Université Grenoble Alpes, CNRS, CEA, Grenoble, France
| | - Didier Boturyn
- Université Grenoble Alpes, Département de Chimie Moléculaire (DCM), Grenoble, France; CNRS, DCM, Grenoble, France
| | - Corinne Albiges-Rizo
- Institut Albert Bonniot, Université Grenoble Alpes, INSERM, CNRS, Grenoble, France
| | - Liliane Coche-Guerente
- Université Grenoble Alpes, Département de Chimie Moléculaire (DCM), Grenoble, France; CNRS, DCM, Grenoble, France
| | - Catherine Picart
- CNRS UMR 5628 (LMGP), Grenoble, France; Grenoble Institute of Technology, Université Grenoble Alpes, LMGP, Grenoble, France.
| | - Ralf P Richter
- Université Grenoble Alpes, Département de Chimie Moléculaire (DCM), Grenoble, France; CNRS, DCM, Grenoble, France; University of Leeds, School of Biomedical Sciences and School of Physics and Astronomy, Leeds, United Kingdom; CIC biomaGUNE, San Sebastian, Spain.
| |
Collapse
|
20
|
Nassari S, Duprez D, Fournier-Thibault C. Non-myogenic Contribution to Muscle Development and Homeostasis: The Role of Connective Tissues. Front Cell Dev Biol 2017; 5:22. [PMID: 28386539 PMCID: PMC5362625 DOI: 10.3389/fcell.2017.00022] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/07/2017] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscles belong to the musculoskeletal system, which is composed of bone, tendon, ligament and irregular connective tissue, and closely associated with motor nerves and blood vessels. The intrinsic molecular signals regulating myogenesis have been extensively investigated. However, muscle development, homeostasis and regeneration require interactions with surrounding tissues and the cellular and molecular aspects of this dialogue have not been completely elucidated. During development and adult life, myogenic cells are closely associated with the different types of connective tissue. Connective tissues are defined as specialized (bone and cartilage), dense regular (tendon and ligament) and dense irregular connective tissue. The role of connective tissue in muscle morphogenesis has been investigated, thanks to the identification of transcription factors that characterize the different types of connective tissues. Here, we review the development of the various connective tissues in the context of the musculoskeletal system and highlight their important role in delivering information necessary for correct muscle morphogenesis, from the early step of myoblast differentiation to the late stage of muscle maturation. Interactions between muscle and connective tissue are also critical in the adult during muscle regeneration, as impairment of the regenerative potential after injury or in neuromuscular diseases results in the progressive replacement of the muscle mass by fibrotic tissue. We conclude that bi-directional communication between muscle and connective tissue is critical for a correct assembly of the musculoskeletal system during development as well as to maintain its homeostasis in the adult.
Collapse
Affiliation(s)
- Sonya Nassari
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| | - Delphine Duprez
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| | - Claire Fournier-Thibault
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| |
Collapse
|
21
|
Puchert M, Adams V, Linke A, Engele J. Evidence for the involvement of the CXCL12 system in the adaptation of skeletal muscles to physical exercise. Cell Signal 2016; 28:1205-1215. [DOI: 10.1016/j.cellsig.2016.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/19/2016] [Accepted: 05/24/2016] [Indexed: 12/23/2022]
|
22
|
Lu J, Zhou WH, Ren L, Zhang YZ. CXCR4, CXCR7, and CXCL12 are associated with trophoblastic cells apoptosis and linked to pathophysiology of severe preeclampsia. Exp Mol Pathol 2015; 100:184-91. [PMID: 26721717 DOI: 10.1016/j.yexmp.2015.12.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/20/2015] [Accepted: 12/20/2015] [Indexed: 11/26/2022]
Abstract
Preeclampsia is a pregnancy disorder with sudden onset of maternal hypertension and proteinuria, which is characterized by defective cytotrophoblast invasion, increased apoptosis in cytotrophoblast, and diminished syncytial differentiation. In this study, samples from 11 mild preeclamptic patients, 18 severe preeclamptic patients, and 21 normal pregnant women were collected. The expression level of CXCL12 and its two receptors (CXCR4 and CXCR7) in these samples and their relationship with apoptosis were investigated. Morphological change of trophoblast cells that was observed by scanning electron microscope (SEM) indicated a significant tendency of apoptosis in the preeclamptic placenta. Immunohistochemical staining showed that expression level of three proteins was significantly lower in severe preeclamptic placentas compared with normal placentas (P<0.05), whereas no significant difference was found between mild preeclamptic and normal placentas (P>0.05). Real time quantitative PCR (RT-qPCR) and Western blot showed that both mRNA and protein expression level of CXCR4, CXCR7, and CXCL12 of trophoblasts were lower in the severe preeclampsia group than that in the normal group (P<0.05 for mRNA, P<0.01 for protein). In conclusion, our data revealed that the roles of CXCR4, CXCR7, and CXCL12 are associated with trophoblastic cells apoptosis and may be linked to the occurrence and development of severe preeclampsia.
Collapse
Affiliation(s)
- Jing Lu
- Department of Obstetrics and Gynecology, Medicine Center for Human Reproduction, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, People's Republic of China
| | - Wen-Hui Zhou
- Medicine Center for Human Reproduction, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Liang Ren
- Department of Obstetrics and Gynecology, Medicine Center for Human Reproduction, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, People's Republic of China
| | - Yuan-Zhen Zhang
- Department of Obstetrics and Gynecology, Medicine Center for Human Reproduction, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, People's Republic of China.
| |
Collapse
|
23
|
Ponsuksili S, Siengdee P, Du Y, Trakooljul N, Murani E, Schwerin M, Wimmers K. Identification of common regulators of genes in co-expression networks affecting muscle and meat properties. PLoS One 2015; 10:e0123678. [PMID: 25875247 PMCID: PMC4397042 DOI: 10.1371/journal.pone.0123678] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/21/2015] [Indexed: 12/21/2022] Open
Abstract
Understanding the genetic contributions behind skeletal muscle composition and metabolism is of great interest in medicine and agriculture. Attempts to dissect these complex traits combine genome-wide genotyping, expression data analyses and network analyses. Weighted gene co-expression network analysis (WGCNA) groups genes into modules based on patterns of co-expression, which can be linked to phenotypes by correlation analysis of trait values and the module eigengenes, i.e. the first principal component of a given module. Network hub genes and regulators of the genes in the modules are likely to play an important role in the emergence of respective traits. In order to detect common regulators of genes in modules showing association with meat quality traits, we identified eQTL for each of these genes, including the highly connected hub genes. Additionally, the module eigengene values were used for association analyses in order to derive a joint eQTL for the respective module. Thereby major sites of orchestrated regulation of genes within trait-associated modules were detected as hotspots of eQTL of many genes of a module and of its eigengene. These sites harbor likely common regulators of genes in the modules. We exemplarily showed the consistent impact of candidate common regulators on the expression of members of respective modules by RNAi knockdown experiments. In fact, Cxcr7 was identified and validated as a regulator of genes in a module, which is involved in the function of defense response in muscle cells. Zfp36l2 was confirmed as a regulator of genes of a module related to cell death or apoptosis pathways. The integration of eQTL in module networks enabled to interpret the differentially-regulated genes from a systems perspective. By integrating genome-wide genomic and transcriptomic data, employing co-expression and eQTL analyses, the study revealed likely regulators that are involved in the fine-tuning and synchronization of genes with trait-associated expression.
Collapse
Affiliation(s)
- Siriluck Ponsuksili
- Institute for ‘Genome Biology’, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
| | - Puntita Siengdee
- Institute for ‘Genome Biology’, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
| | - Yang Du
- Institute for ‘Genome Biology’, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
| | - Nares Trakooljul
- Institute for ‘Genome Biology’, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
| | - Eduard Murani
- Institute for ‘Genome Biology’, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
| | - Manfred Schwerin
- Institute for ‘Genome Biology’, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
| | - Klaus Wimmers
- Institute for ‘Genome Biology’, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
- * E-mail:
| |
Collapse
|
24
|
Kozakowska M, Kotlinowski J, Grochot-Przeczek A, Ciesla M, Pilecki B, Derlacz R, Dulak J, Jozkowicz A. Myoblast-conditioned media improve regeneration and revascularization of ischemic muscles in diabetic mice. Stem Cell Res Ther 2015; 6:61. [PMID: 25889676 PMCID: PMC4431532 DOI: 10.1186/s13287-015-0063-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 02/21/2015] [Accepted: 03/24/2015] [Indexed: 12/17/2022] Open
Abstract
Introduction Diabetes is associated with reduced expression of heme oxygenase-1 (HO-1), a heme-degrading enzyme with cytoprotective and proangiogenic properties. In myoblasts and muscle satellite cells HO-1 improves survival, proliferation and production of proangiogenic growth factors. Induction of HO-1 in injured tissues facilitates neovascularization, the process impaired in diabetes. We aimed to examine whether conditioned media from the HO-1 overexpressing myoblast cell line can improve a blood-flow recovery in ischemic muscles of diabetic mice. Methods Analysis of myogenic markers was performed at the mRNA level in primary muscle satellite cells, isolated by a pre-plate technique from diabetic db/db and normoglycemic wild-type mice, and then cultured under growth or differentiation conditions. Hind limb ischemia was performed by femoral artery ligation in db/db mice and blood recovery was monitored by laser Doppler measurements. Mice were treated with a single intramuscular injection of conditioned media harvested from wild-type C2C12 myoblast cell line, C2C12 cells stably transduced with HO-1 cDNA, or with unconditioned media. Results Expression of HO-1 was lower in muscle satellite cells isolated from muscles of diabetic db/db mice when compared to their wild-type counterparts, what was accompanied by increased levels of Myf5 or CXCR4, and decreased Mef2 or Pax7. Such cells also displayed diminished differentiation potential when cultured in vitro, as shown by less effective formation of myotubes and reduced expression of myogenic markers (myogenic differentiation antigen - myoD, myogenin and myosin). Blood flow recovery after induction of severe hind limb ischemia was delayed in db/db mice compared to that in normoglycemic individuals. To improve muscle regeneration after ischemia, conditioned media collected from differentiating C2C12 cells (control and HO-1 overexpressing) were injected into hind limbs of diabetic mice. Analysis of blood flow revealed that media from HO-1 overexpressing cells accelerated blood-flow recovery, while immunohistochemical staining assessment of vessel density in injected muscle confirmed increased angiogenesis. The effect might be mediated by stromal-cell derived factor-1α proangiogenic factor, as its secretion is elevated in HO-1 overexpressing cells. Conclusions In conclusion, paracrine stimulation of angiogenesis in ischemic skeletal muscle using conditioned media may be a safe approach exploiting protective and proangiogenic properties of HO-1 in diabetes.
Collapse
Affiliation(s)
- Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland.
| | - Jerzy Kotlinowski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland.
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland.
| | - Maciej Ciesla
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland.
| | - Bartosz Pilecki
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland.
| | - Rafal Derlacz
- R&D Department, Adamed Ltd, Pienkow 149, Czosnow, 05-152, Poland. .,Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Miecznikowa 1, Warsaw, 02-096, Poland.
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland. .,Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland.
| |
Collapse
|
25
|
Geng X, Xu T, Niu Z, Zhou X, Zhao L, Xie Z, Xue D, Zhang F, Xu C. Differential proteome analysis of the cell differentiation regulated by BCC, CRH, CXCR4, GnRH, GPCR, IL1 signaling pathways in Chinese fire-bellied newt limb regeneration. Differentiation 2014; 88:85-96. [DOI: 10.1016/j.diff.2014.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 10/07/2014] [Accepted: 10/29/2014] [Indexed: 12/11/2022]
|
26
|
Lipfert J, Ödemis V, Wagner DC, Boltze J, Engele J. CXCR4 and CXCR7 form a functional receptor unit for SDF-1/CXCL12 in primary rodent microglia. Neuropathol Appl Neurobiol 2014; 39:667-80. [PMID: 23289420 DOI: 10.1111/nan.12015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 11/30/2012] [Indexed: 12/25/2022]
Abstract
AIMS Microglial cells have been originally identified as a target for the CXC chemokine, SDF-1, by their expression of CXCR4. More recently, it has been recognized that SDF-1 additionally binds to CXCR7, which depending on the cell type acts as either a nonclassical, a classical or a scavenger chemokine receptor. Here, we asked whether primary microglial cells additionally express CXCR7 and if so how this chemokine receptor functions in this cell type. METHODS CXCR4 and CXCR7 expression was analysed in cultured rat microglia and in the brain of animals with permanent occlusion of the middle cerebral artery (MCAO) by either Western blotting, RT-PCR, flow cytometry and/or immunocytochemistry. The function of CXCR4 and CXCR7 was assessed in the presence of selective antagonists. RESULTS Cultured primary rat microglia expressed CXCR4 and CXCR7 to similar levels. Treatment with SDF-1 resulted in the activation of Erk1/2 and Akt signalling. Erk1/2 and Akt signalling were required for subsequent SDF-1-dependent promotion of microglial proliferation. In contrast, Erk1/2 signalling was sufficient for SDF-1-induced migration of microglial cells. Both SDF-1-dependent signalling and the resulting effects on microglial proliferation and migration were abrogated following pharmacological inactivation of either CXCR4 or CXCR7. Moreover, treatment of cultured microglia with lipopolysaccharide resulted in the co-ordinated up-regulation of CXCR4 and CXCR7 expression. Likewise, reactive microglia accumulating in the area adjacent to the lesion core in MCAO rats expressed both CXCR4 and CXCR7. CONCLUSIONS CXCR4 and CXCR7 form a functional receptor unit in microglial cells, which is up-regulated during activation of microglia both in vitro and in vivo.
Collapse
Affiliation(s)
- J Lipfert
- Institute of Anatomy, Medical Faculty, University of Leipzig, Leipzig, Germany
| | | | | | | | | |
Collapse
|
27
|
Bobadilla M, Sainz N, Abizanda G, Orbe J, Rodriguez JA, Páramo JA, Prósper F, Pérez-Ruiz A. The CXCR4/SDF1 axis improves muscle regeneration through MMP-10 activity. Stem Cells Dev 2014; 23:1417-27. [PMID: 24548137 DOI: 10.1089/scd.2013.0491] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The CXCR4/SDF1 axis participates in various cellular processes, including cell migration, which is essential for skeletal muscle repair. Although increasing evidence has confirmed the role of CXCR4/SDF1 in embryonic muscle development, the function of this pathway during adult myogenesis remains to be fully elucidated. In addition, a role for CXCR4 signaling in muscle maintenance and repair has only recently emerged. Here, we have demonstrated that CXCR4 and stromal cell-derived factor-1 (SDF1) are up-regulated in injured muscle, suggesting their involvement in the repair process. In addition, we found that notexin-damaged muscles showed delayed muscle regeneration on treatment with CXCR4 agonist (AMD3100). Accordingly, small-interfering RNA-mediated silencing of SDF1 or CXCR4 in injured muscles impaired muscle regeneration, whereas the addition of SDF1 ligand accelerated repair. Furthermore, we identified that CXCR4/SDF1-regulated muscle repair was dependent on matrix metalloproteinase-10 (MMP-10) activity. Thus, our findings support a model in which MMP-10 activity modulates CXCR4/SDF1 signaling, which is essential for efficient skeletal muscle regeneration.
Collapse
Affiliation(s)
- Miriam Bobadilla
- 1 Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra , Pamplona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
28
|
C/EBPβ mediates osteoclast recruitment by regulating endothelial progenitor cell expression of SDF-1α. PLoS One 2014; 9:e91217. [PMID: 24618682 PMCID: PMC3949754 DOI: 10.1371/journal.pone.0091217] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 02/10/2014] [Indexed: 12/12/2022] Open
Abstract
Integration of tissue-engineered bone grafts with the host bone is vital for the healing of critical-size bone defects. An important aspect of this process is bone resorption, which must be carried out by osteoclasts derived from the host. However, the mechanism underlying recruitment of host osteoclast precursors to graft sites remains unclear. Endothelial progenitor cells (EPCs) mobilize from the bone marrow into the circulation and home to sites of angiogenesis such as tissue remodeling. Since EPCs express SDF-1, and C/EBPβ is known to regulate SDF-1α expression, we hypothesized that EPCs may recruit CXCR4-expressing host osteoclast precursors to the repair area and that this recruitment may be mediated through C/EBPβ signaling. Using an inflammatory EPC model we showed that EPCs upregulate protein levels of both SDF-1α and C/EBPβ. A luciferase assay confirmed that C/EBPβ acts on the SDF-1α promoter in these cells, and that binding is increased under conditions of inflammation, while silencing of C/EBPβ reduces expression of SDF-1 α and C/EBPβ. Using RAW264.7 cells as a model of osteoclastic monocyte precursors, we investigated their responses to migratory factors in EPC conditioned medium. We showed that RAW264.7 cells migrate towards conditioned medium from EPCs treated with IL-1β, an effect which could be abolished by silencing C/EBPβ in EPCs, and was almost completely blocked by silencing CXCR4 in RAW264.7 cells. These findings show that EPCs respond to inflammatory stimuli by signaling to osteoclast precursors via SDF-1, and that C/EBPβ mediates this response.
Collapse
|
29
|
Dalonneau F, Liu XQ, Sadir R, Almodovar J, Mertani HC, Bruckert F, Albiges-Rizo C, Weidenhaupt M, Lortat-Jacob H, Picart C. The effect of delivering the chemokine SDF-1α in a matrix-bound manner on myogenesis. Biomaterials 2014; 35:4525-4535. [PMID: 24612919 DOI: 10.1016/j.biomaterials.2014.02.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/05/2014] [Indexed: 02/07/2023]
Abstract
Several chemokines are important in muscle myogenesis and in the recruitment of muscle precursors during muscle regeneration. Among these, the SDF-1α chemokine (CXCL12) is a potent chemoattractant known to be involved in muscle repair. SDF-1α was loaded in polyelectrolyte multilayer films made of poly(L-lysine) and hyaluronan to be delivered locally to myoblast cells in a matrix-bound manner. The adsorbed amounts of SDF-1α were tuned over a large range from 100 ng/cm(2) to 5 μg/cm(2), depending on the initial concentration of SDF-1α in solution, its pH, and on the film crosslinking extent. Matrix-bound SDF-1α induced a striking increase in myoblast spreading, which was revealed when it was delivered from weakly crosslinked films. It also significantly enhanced cell migration in a dose-dependent manner, which again depended on its presentation by the biopolymeric film. The low-crosslinked film was the most efficient in boosting cell migration. Furthermore, matrix-bound SDF-1α also increased the expression of myogenic markers but the fusion index decreased in a dose-dependent manner with the adsorbed amount of SDF-1α. At high adsorbed amounts of SDF-1α, a large number of Troponin T-positive cells had only one nucleus. Overall, this work reveals the importance of the presentation mode of SDF-1α to emphasize its effect on myogenic processes. These films may be further used to provide insight into the role of SDF-1α presented by a biomaterial in physiological or pathological processes.
Collapse
Affiliation(s)
- Fabien Dalonneau
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016 Grenoble, France.,Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Xi Qiu Liu
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016 Grenoble, France.,Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France.,FONDATION ARC, 9 rue Guy Môquet 94803 Villejuif, France
| | - Rabia Sadir
- Univ. Grenoble Alpes, Institut de Biologie Structurale (IBS), F-38027 Grenoble, France.,CNRS, IBS, F-38027 Grenoble, France.,CEA, DSV, IBS, F-38027 Grenoble, France
| | - Jorge Almodovar
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016 Grenoble, France.,Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Hichem C Mertani
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM 1052 - CNRS 5286, 28, rue Laennec, 69373 LYON cedex 08, France
| | - Franz Bruckert
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016 Grenoble, France.,Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Corinne Albiges-Rizo
- Inserm U823, ERL CNRS5284, Université Joseph Fourier, Institut Albert Bonniot, Site Santé, BP170, 38042 Grenoble cedex 9, France
| | - Marianne Weidenhaupt
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016 Grenoble, France.,Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Hugues Lortat-Jacob
- Univ. Grenoble Alpes, Institut de Biologie Structurale (IBS), F-38027 Grenoble, France.,CNRS, IBS, F-38027 Grenoble, France.,CEA, DSV, IBS, F-38027 Grenoble, France
| | - Catherine Picart
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016 Grenoble, France.,Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
| |
Collapse
|
30
|
The peculiarities of the SDF-1/CXCL12 system: in some cells, CXCR4 and CXCR7 sing solos, in others, they sing duets. Cell Tissue Res 2013; 355:239-53. [DOI: 10.1007/s00441-013-1747-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 10/17/2013] [Indexed: 12/26/2022]
|
31
|
Leucht P, Temiyasathit S, Russell A, Arguello JF, Jacobs CR, Helms JA, Castillo AB. CXCR4 antagonism attenuates load-induced periosteal bone formation in mice. J Orthop Res 2013; 31:1828-38. [PMID: 23881789 DOI: 10.1002/jor.22440] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 06/25/2013] [Indexed: 02/04/2023]
Abstract
Mechanical loading is a key anabolic regulator of bone mass. Stromal cell-derived factor-1 (SDF-1) is a stem cell homing factor that is important in hematopoiesis, angiogenesis, and fracture healing, though its involvement in skeletal mechanoadaptation is virtually unknown. The objective of this study was to characterize skeletal expression patterns of SDF-1 and CXCR4, the receptor for SDF-1, and to determine the role of SDF-1 signaling in load-induced periosteal bone formation. Sixteen-week-old C57BL/6 mice were treated with PBS or AMD3100, an antagonist against CXCR4, and exposed to in vivo ulnar loading (2.8 N peak-to-peak, 2 Hz, 120 cycles). SDF-1 was expressed in cortical and trabecular osteocytes and marrow cells, and CXCR4 was primarily expressed in marrow cells. SDF-1 and CXCR4 expression was enhanced in response to mechanical stimulation. The CXCR4 receptor antagonist AMD3100 significantly attenuated load-induced bone formation and led to smaller adaptive changes in cortical geometric properties as determined by histomorphometric analysis. Our data suggest that SDF-1/CXCR4 signaling plays a critical role in skeletal mechanoadaptation, and may represent a unique therapeutic target for prevention and treatment of age-related and disuse bone loss.
Collapse
Affiliation(s)
- Philipp Leucht
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | | | | | | | | | | | | |
Collapse
|
32
|
Bajoghli B. Evolution and function of chemokine receptors in the immune system of lower vertebrates. Eur J Immunol 2013; 43:1686-92. [DOI: 10.1002/eji.201343557] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 05/20/2013] [Accepted: 05/27/2013] [Indexed: 01/09/2023]
Affiliation(s)
- Baubak Bajoghli
- European Molecular Biology Laboratory (EMBL); Heidelberg; Germany
| |
Collapse
|
33
|
Expression of CXCL12 and CXCL14 during eye development in chick and mouse. Gene Expr Patterns 2013; 13:303-10. [PMID: 23727298 DOI: 10.1016/j.gep.2013.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 05/13/2013] [Indexed: 02/05/2023]
Abstract
Vertebrate eye development is a complex multistep process coordinated by signals from the lens, optic cup and periocular mesenchyme. Although chemokines are increasingly being recognized as key players in cell migration, proliferation, and differentiation during embryonic development, their potential role during eye development has not been examined. In this study, we demonstrate by section in situ hybridization that CXCL12 and CXCL14 are expressed during ocular development. CXCL12 is expressed in the periocular mesenchyme, ocular blood vessels, retina, and eyelid mesenchyme, and its expression pattern is conserved between chick and mouse in most tissues. Expression of CXCL14 is localized in the ocular ectoderm, limbal epithelium, scleral papillae, eyelid mesenchyme, corneal keratocytes, hair follicles, and retina, and it was only conserved in the upper eyelid ectoderm of chick and mouse. The unique and non-overlapping patterns of CXCL12 and CXCL14 expression in ocular tissues suggest that these two chemokines may interact and have important functions in cell proliferation, differentiation and migration during eye development.
Collapse
|
34
|
Ramsey DM, McAlpine SR. Halting metastasis through CXCR4 inhibition. Bioorg Med Chem Lett 2012; 23:20-5. [PMID: 23211868 DOI: 10.1016/j.bmcl.2012.10.138] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 10/31/2012] [Indexed: 12/14/2022]
Abstract
Metastasis occurs when cancer cells leave the primary tumor site and migrate to distant parts of the body. The CXCR4-SDF-1 pathway facilitates this migration, and its expression has become the hallmark of several metastatic cancers. Targeted approaches are currently being developed to inhibit this pathway, and several candidates are now in clinical trials. Continued exploration of CXCR4 inhibitors will generate compounds that have improved activity over current candidates.
Collapse
Affiliation(s)
- Deborah M Ramsey
- Department of Chemistry, University of New South Wales, Sydney NSW 2052, Australia.
| | | |
Collapse
|