1
|
Li M, Peng Y, Shi Y, Liu Y, Zhang J. Advancements in the study of DLK1 in the pathogenesis of diabetes. Life Sci 2025; 369:123535. [PMID: 40054732 DOI: 10.1016/j.lfs.2025.123535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/30/2025]
Abstract
DLK1, as a membrane-bound protein, has been extensively studied in the field of cancer research. As a ligand downstream of the Notch pathway, it broadly influences developmental and metabolic processes in the body. With deeper research, it has been found that DLK1 can induce the synthesis and secretion of insulin through the ERK and AKT pathways, playing a crucial role in the development of metabolic diseases. Diabetes mellitus (DM) is a chronic metabolic disorder characterized by insufficient insulin production by the pancreas or inadequate utilization of insulin by the body. This article aims to review the relationship between DLK1 and diabetes, recent research advancements, and to discuss future research directions and challenges.
Collapse
Affiliation(s)
- Min Li
- School of Bioengineering, Zunyi Medical University, Zhuhai 519041, China
| | - Yanqiu Peng
- School of Bioengineering, Zunyi Medical University, Zhuhai 519041, China
| | - Yuke Shi
- School of Bioengineering, Zunyi Medical University, Zhuhai 519041, China
| | - Yunfei Liu
- School of Bioengineering, Zunyi Medical University, Zhuhai 519041, China
| | - Jian Zhang
- School of Bioengineering, Zunyi Medical University, Zhuhai 519041, China.
| |
Collapse
|
2
|
Pomares O, Laborda J, Vales-Villamarín C, Pérez-Nadador I, Mejorado-Molano FJ, Parra-Rodríguez A, Mahillo-Fernández I, Soriano-Guillén L, Garcés C. Association of DLK1 SNPs with body mass index and plasma lipid levels in children. Pediatr Res 2025:10.1038/s41390-025-04005-0. [PMID: 40113998 DOI: 10.1038/s41390-025-04005-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/07/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND The DLK1 human gene encodes for the transmembrane EGF-like repeat-containing protein DLK1, which acts as a modulator of adipogenesis. A role for DLK1 in energy metabolism and lipid homeostasis has been suggested and DLK1 gene variants have been related to pubertal development. OBJECTIVE The aim of this study was to uncover DLK1 SNPs in a cohort of children and analyze their relationship with anthropometric and biochemical variables. METHODS Our population-based sample comprises 1237 healthy 6-to-8-year-old Caucasian children. The presence of five DLK1 SNPs (rs1802710, rs876374, rs7155375, rs57098752, and rs7149242) was analyzed by Real-Time PCR, using predesigned TaqMan™ Genotyping Assays. RESULTS We observed that the SNPs rs1802710 and rs876374 were associated with BMI, and the prevalence of these two SNPs was different in normal-weight children compared to children with obesity. Related to biochemical variables, we found a significant association of the SNPs rs1802710, rs876374, and rs57098752 and their combination with Apo-B plasma concentrations after adjusting by BMI and sex. The SNPs rs1802710 and rs57098752 were also significantly associated with plasma levels of LDL-C and HDL-C, respectively. CONCLUSION Our study reveals that DLK1 gene variants may influence both body weight and lipid homeostasis, affecting particularly to the Apo-B biology, in children. IMPACT DLK1 polymorphisms are associated with BMI and with lipid levels, independently of BMI, early in life. Our data add to the existing literature the evidence that DLK1 gene variants impact on lipid metabolism. The confirmation at the population level that DLK1 genetic variants are associated with anthropometric and lipid variables sustains the role of DLK1 in obesity and related disorders and should lead to further studies aimed at clarifying this effect.
Collapse
Affiliation(s)
- Olga Pomares
- Lipid Research Laboratory, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - Jorge Laborda
- Department of Inorganic and Organic Chemistry and Biochemistry, Pharmacy School, University of Castilla-La Mancha, Albacete, Spain
| | | | - Iris Pérez-Nadador
- Lipid Research Laboratory, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | | | | | | | | | - Carmen Garcés
- Lipid Research Laboratory, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain.
| |
Collapse
|
3
|
Hu X, Wang Z, Wang W, Cui P, Kong C, Chen X, Lu S. Irisin as an agent for protecting against osteoporosis: A review of the current mechanisms and pathways. J Adv Res 2024; 62:175-186. [PMID: 37669714 PMCID: PMC11331170 DOI: 10.1016/j.jare.2023.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Osteoporosis is recognized as a skeletal disorder characterized by diminished bone tissue quality and density. Regular physical exercise is widely acknowledged to preserve and enhance bone health, but the detailed molecular mechanisms involved remain unclear. Irisin, a factor derived from muscle during exercise, influences bone and muscle. Since its discovery in 2012, irisin has been found to promote bone growth and reduce bone resorption, establishing a tangible link between muscle exertion and bone health. Consequently, the mechanism by which irisin prevents osteoporosis have attracted significant scientific interest. AIM OF THE REVIEW This study aims to elucidate the multifaceted relationship between exercise, irisin, and bone health. Focusing on irisin, a muscle-derived factor released during exercise, we seek to understand its role in promoting bone growth and inhibiting resorption. Through a review of current research article on irisin in osteoporosis, Our review provides a deep dive into existing research on influence of irisin in osteoporosis, exploring its interaction with pivotal signaling pathways and its impact on various cell death mechanisms and inflammation. We aim to uncover the molecular underpinnings of how irisin, secreted during exercise, can serve as a therapeutic strategy for osteoporosis. KEY SCIENTIFIC CONCEPTS OF THE REVIEW Irisin, secreted during exercise, plays a vital role in bridging muscle function to bone health. It not only promotes bone growth but also inhibits bone resorption. Specifically, Irisin fosters osteoblast proliferation, differentiation, and mineralization predominantly through the ERK, p38, and AMPK signaling pathways. Concurrently, it regulates osteoclast differentiation and maturation via the JNK, Wnt/β-catenin and RANKL/RANK/OPG signaling pathways. This review further delves into the profound significance of irisin in osteoporosis and its involvement in diverse cellular death mechanisms, including apoptosis, autophagy, ferroptosis, and pyroptosis.
Collapse
Affiliation(s)
- Xinli Hu
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing 100053, China; National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Zheng Wang
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing 100053, China; National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Wei Wang
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing 100053, China; National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Peng Cui
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing 100053, China; National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Chao Kong
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing 100053, China; National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Xiaolong Chen
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing 100053, China; National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Shibao Lu
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing 100053, China; National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
4
|
Chen T, Peng Y, Hu W, Shi H, Li P, Que Y, Qiu J, Qiu X, Gao B, Zhou H, Chen Y, Zhu Y, Li S, Liang A, Gao W, Huang D. Irisin enhances chondrogenic differentiation of human mesenchymal stem cells via Rap1/PI3K/AKT axis. Stem Cell Res Ther 2022; 13:392. [PMID: 35922833 PMCID: PMC9351134 DOI: 10.1186/s13287-022-03092-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/17/2022] [Indexed: 12/11/2022] Open
Abstract
Background Human mesenchymal stem cells (hMSCs) have been proven to have inherent chondrogenic differentiation potential, which appears to be used in cartilage regeneration. Increasing evidence suggests that irisin enhances osteoblast differentiation of MSCs, but little is known about its potential on chondrogenic differentiation. Methods In the study, we investigated the effects of irisin on chondrogenic differentiation of hMSCs using a high-density pellet culture system. The cartilage pellets were evaluated by morphology, and the metabolism of cartilage matrix was detected by qPCR, western blot and immunohistochemistry. Next, RNA-seq was performed to explore the underlying mechanism. Furthermore, using the transduction of plasmid, miRNAs mimics and inhibitor, the activation of Rap1/PI3K/AKT axis, the expression level of SIPA1L2, and the functional verification of miR-125b-5p were detected on day 7 of chondrogenic differentiation of hMSCs. Results Compared with the controls, we found that irisin treatment could significantly enhance the chondrogenic differentiation of hMSCs, enlarge the induced-cartilage tissue and up-regulate the expression levels of cartilage markers. RNA-seq indicated that irisin activated the Rap1 and PI3K/AKT signaling pathway, and the lower expression level of SIPA1L2 and the higher expression level of miR-125b-5p were found in irisin-treated group. Further, we found that irisin treatment could up-regulate the expression level of miR-125b-5p, targeting SIPA1L2 and consequently activating the Rap1/PI3K/AKT axis on the process of chondrogenic differentiation of hMSCs. Conclusions Collectively, our study reveals that irisin can enhance chondrogenic differentiation of hMSCs via the Rap1/PI3K/AKT pathway, suggesting that irisin possesses prospects in cartilage regeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03092-8.
Collapse
Affiliation(s)
- Taiqiu Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Yan Peng
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Wenjun Hu
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Huihong Shi
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Pengfei Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Yichen Que
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Jincheng Qiu
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Xianjian Qiu
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Bo Gao
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Hang Zhou
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Yanbo Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Yuanxin Zhu
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Shaoguang Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Anjing Liang
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China
| | - Wenjie Gao
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China.
| | - Dongsheng Huang
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, #107 West Yan Jiang Road, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
Naranjo AI, González-Gómez MJ, Baladrón V, Laborda J, Nueda ML. Different Expression Levels of DLK2 Inhibit NOTCH Signaling and Inversely Modulate MDA-MB-231 Breast Cancer Tumor Growth In Vivo. Int J Mol Sci 2022; 23:1554. [PMID: 35163478 PMCID: PMC8836183 DOI: 10.3390/ijms23031554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 11/22/2022] Open
Abstract
NOTCH signaling is implicated in the development of breast cancer tumors. DLK2, a non-canonical inhibitor of NOTCH signaling, was previously shown to be involved in skin and breast cancer. In this work, we studied whether different levels of DLK2 expression influenced the breast cancer characteristics of MDA-MB-231 cells. We found that DLK2 overexpression inhibited NOTCH activation in a dose-dependent manner. Moreover, depending on the level of inhibition of NOTCH1 activation generated by different levels of DLK2 expression, cell proliferation, cell cycle dynamics, cell apoptosis, cell migration, and tumor growth in vivo were affected in opposite directions. Low levels of DLK2 expression produced a slight inhibition of NOTCH1 activation, and enhanced MDA-MB-231 cell invasion in vitro and cell proliferation both in vitro and in vivo. In contrast, MDA-MB-231 cells expressing elevated levels of DLK2 showed a strong inhibition of NOTCH1 activation, decreased cell proliferation, increased cell apoptosis, and were unable to generate tumors in vivo. In addition, DLK2 expression levels also affected some members of other cell signaling pathways implicated in cancer, such as ERK1/2 MAPK, AKT, and rpS6 kinases. Our data support an important role of DLK2 as a protein that can finely regulate NOTCH signaling and affect the tumor properties and growth dynamics of MDA-MB-231 breast cancer cells.
Collapse
Affiliation(s)
- Ana-Isabel Naranjo
- Biochemistry and Molecular Biology Branch, Medical School/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha (UCLM)/CSIC, 02008 Albacete, Spain; (A.-I.N.); (V.B.)
| | - María-Julia González-Gómez
- Biochemistry and Molecular Biology Branch, Higher Technical School of Agricultural and Forestry Engineering/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha (UCLM)/CSIC, 02008 Albacete, Spain;
| | - Victoriano Baladrón
- Biochemistry and Molecular Biology Branch, Medical School/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha (UCLM)/CSIC, 02008 Albacete, Spain; (A.-I.N.); (V.B.)
| | - Jorge Laborda
- Biochemistry and Molecular Biology Branch, School of Pharmacy/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha (UCLM)/CSIC, 02008 Albacete, Spain
| | - María-Luisa Nueda
- Biochemistry and Molecular Biology Branch, School of Pharmacy/CRIB/Biomedicine Unit, Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha (UCLM)/CSIC, 02008 Albacete, Spain
| |
Collapse
|
6
|
Marchini M, Ashkin MR, Bellini M, Sun MMG, Workentine ML, Okuyan HM, Krawetz R, Beier F, Rolian C. A Na +/K + ATPase Pump Regulates Chondrocyte Differentiation and Bone Length Variation in Mice. Front Cell Dev Biol 2022; 9:708384. [PMID: 34970538 PMCID: PMC8712571 DOI: 10.3389/fcell.2021.708384] [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: 05/11/2021] [Accepted: 11/04/2021] [Indexed: 11/23/2022] Open
Abstract
The genetic and developmental mechanisms involved in limb formation are relatively well documented, but how these mechanisms are modulated by changes in chondrocyte physiology to produce differences in limb bone length remains unclear. Here, we used high throughput RNA sequencing (RNAseq) to probe the developmental genetic basis of variation in limb bone length in Longshanks, a mouse model of experimental evolution. We find that increased tibia length in Longshanks is associated with altered expression of a few key endochondral ossification genes such as Npr3, Dlk1, Sox9, and Sfrp1, as well reduced expression of Fxyd2, a facultative subunit of the cell membrane-bound Na+/K+ ATPase pump (NKA). Next, using murine tibia and cell cultures, we show a dynamic role for NKA in chondrocyte differentiation and in bone length regulation. Specifically, we show that pharmacological inhibition of NKA disrupts chondrocyte differentiation, by upregulating expression of mesenchymal stem cell markers (Prrx1, Serpina3n), downregulation of chondrogenesis marker Sox9, and altered expression of extracellular matrix genes (e.g., collagens) associated with proliferative and hypertrophic chondrocytes. Together, Longshanks and in vitro data suggest a broader developmental and evolutionary role of NKA in regulating limb length diversity.
Collapse
Affiliation(s)
- Marta Marchini
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Mitchell R Ashkin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Melina Bellini
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Margaret Man-Ger Sun
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Matthew Lloyd Workentine
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Hamza Malik Okuyan
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Roman Krawetz
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Campbell Rolian
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
7
|
Wuelling M, Neu C, Thiesen AM, Kitanovski S, Cao Y, Lange A, Westendorf AM, Hoffmann D, Vortkamp A. Epigenetic Mechanisms Mediating Cell State Transitions in Chondrocytes. J Bone Miner Res 2021; 36:968-985. [PMID: 33534175 DOI: 10.1002/jbmr.4263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 01/06/2023]
Abstract
Epigenetic modifications play critical roles in regulating cell lineage differentiation, but the epigenetic mechanisms guiding specific differentiation steps within a cell lineage have rarely been investigated. To decipher such mechanisms, we used the defined transition from proliferating (PC) into hypertrophic chondrocytes (HC) during endochondral ossification as a model. We established a map of activating and repressive histone modifications for each cell type. ChromHMM state transition analysis and Pareto-based integration of differential levels of mRNA and epigenetic marks revealed that differentiation-associated gene repression is initiated by the addition of H3K27me3 to promoters still carrying substantial levels of activating marks. Moreover, the integrative analysis identified genes specifically expressed in cells undergoing the transition into hypertrophy. Investigation of enhancer profiles detected surprising differences in enhancer number, location, and transcription factor binding sites between the two closely related cell types. Furthermore, cell type-specific upregulation of gene expression was associated with increased numbers of H3K27ac peaks. Pathway analysis identified PC-specific enhancers associated with chondrogenic genes, whereas HC-specific enhancers mainly control metabolic pathways linking epigenetic signature to biological functions. Since HC-specific enhancers show a higher conservation in postnatal tissues, the switch to metabolic pathways seems to be a hallmark of differentiated tissues. Surprisingly, the analysis of H3K27ac levels at super-enhancers revealed a rapid adaption of H3K27ac occupancy to changes in gene expression, supporting the importance of enhancer modulation for acute alterations in gene expression. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Manuela Wuelling
- Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Christoph Neu
- Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Andrea M Thiesen
- Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Simo Kitanovski
- Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Yingying Cao
- Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Anja Lange
- Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Daniel Hoffmann
- Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Andrea Vortkamp
- Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
8
|
Ichinose M, Suzuki N, Wang T, Wright JA, Lannagan TRM, Vrbanac L, Kobayashi H, Gieniec KA, Ng JQ, Hayakawa Y, García-Gallastegui P, Monsalve EM, Bauer SR, Laborda J, García-Ramírez JJ, Ibarretxe G, Worthley DL, Woods SL. Stromal DLK1 promotes proliferation and inhibits differentiation of the intestinal epithelium during development. Am J Physiol Gastrointest Liver Physiol 2021; 320:G506-G520. [PMID: 33470182 DOI: 10.1152/ajpgi.00445.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/14/2021] [Indexed: 01/31/2023]
Abstract
The stem/progenitor cells of the developing intestine are biologically distinct from their adult counterparts. Here, we examine the microenvironmental cues that regulate the embryonic stem/progenitor population, focusing on the role of Notch pathway factor delta-like protein-1 (DLK1). mRNA-seq analyses of intestinal mesenchymal cells (IMCs) collected from embryonic day 14.5 (E14.5) or adult IMCs and a novel coculture system with E14.5 intestinal epithelial organoids were used. Following addition of recombinant DLK1 (rDLK) or Dlk1 siRNA (siDlk1), epithelial characteristics were compared using imaging, replating efficiency assays, qPCR, and immunocytochemistry. The intestinal phenotypes of littermate Dlk1+/+ and Dlk1-/- mice were compared using immunohistochemistry. Using transcriptomic analyses, we identified morphogens derived from the embryonic mesenchyme that potentially regulate the developing epithelial cells, to focus on Notch family candidate DLK1. Immunohistochemistry indicated that DLK1 was expressed exclusively in the intestinal stroma at E14.5 at the top of emerging villi, decreased after birth, and shifted to the intestinal epithelium in adulthood. In coculture experiments, addition of rDLK1 to adult IMCs inhibited organoid differentiation, whereas Dlk1 knockdown in embryonic IMCs increased epithelial differentiation to secretory lineage cells. Dlk1-/- mice had restricted Ki67+ cells in the villi base and increased secretory lineage cells compared with Dlk1+/+ embryos. Mesenchyme-derived DLK1 plays an important role in the promotion of epithelial stem/precursor expansion and prevention of differentiation to secretory lineages in the developing intestine.NEW & NOTEWORTHY Using a novel coculture system, transcriptomics, and transgenic mice, we investigated differential molecular signaling between the intestinal epithelium and mesenchyme during development and in the adult. We show that the Notch pathway factor delta-like protein-1 (DLK1) is stromally produced during development and uncover a new role for DLK1 in the regulation of intestinal epithelial stem/precursor expansion and differentiation to secretory lineages.
Collapse
Affiliation(s)
- Mari Ichinose
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Nobumi Suzuki
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tongtong Wang
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Josephine A Wright
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Tamsin R M Lannagan
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Laura Vrbanac
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Hiroki Kobayashi
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Krystyna A Gieniec
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Jia Q Ng
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Patricia García-Gallastegui
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country, Bizkaia, Spain
| | - Eva M Monsalve
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, Albacete, Spain
| | - Steven R Bauer
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland
| | - Jorge Laborda
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, Albacete, Spain
| | - J J García-Ramírez
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, Albacete, Spain
| | - Gaskon Ibarretxe
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country, Bizkaia, Spain
| | - Daniel L Worthley
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Susan L Woods
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
9
|
Rodriguez P, Sassi Y, Troncone L, Benard L, Ishikawa K, Gordon RE, Lamas S, Laborda J, Hajjar RJ, Lebeche D. Deletion of delta-like 1 homologue accelerates fibroblast-myofibroblast differentiation and induces myocardial fibrosis. Eur Heart J 2020; 40:967-978. [PMID: 29668883 DOI: 10.1093/eurheartj/ehy188] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/14/2018] [Accepted: 03/21/2018] [Indexed: 12/11/2022] Open
Abstract
AIMS Myocardial fibrosis is associated with profound changes in ventricular architecture and geometry, resulting in diminished cardiac function. There is currently no information on the role of the delta-like homologue 1 (Dlk1) in the regulation of the fibrotic response. Here, we investigated whether Dlk1 is involved in cardiac fibroblast-to-myofibroblast differentiation and regulates myocardial fibrosis and explored the molecular mechanism underpinning its effects in this process. METHODS AND RESULTS Using Dlk1-knockout mice and adenoviral gene delivery, we demonstrate that overexpression of Dlk1 in cardio-fibroblasts resulted in inhibition of fibroblast proliferation and differentiation into myofibroblasts. This process is mediated by TGF-β1 signalling, since isolated fibroblasts lacking Dlk1 exhibited a higher activation of the TGF-β1/Smad-3 pathway at baseline, leading to an earlier acquisition of a myofibroblast phenotype. Likewise, Dlk1-null mice displayed increased TGF-β1/Smad3 cardiac activity, resulting in infiltration/accumulation of myofibroblasts, induction and deposition of extra-domain A-fibronectin isoform and collagen, and activation of pro-fibrotic markers. Furthermore, these profibrotic events were associated with disrupted myofibril integrity, myocyte hypertrophy, and cardiac dysfunction. Interestingly, Dlk1 expression was down-regulated in ischaemic human and porcine heart tissues. Mechanistically, miR-370 mediated Dlk1's regulation of cardiac fibroblast-myofibroblast differentiation by directly targeting TGFβ-R2/Smad-3 signalling, while the Dlk1 canonical target, Notch pathway, does not seem to play a role in this process. CONCLUSION These findings are the first to demonstrate an inhibitory role of Dlk1 of cardiac fibroblast-to-myofibroblast differentiation by interfering with TGFβ/Smad-3 signalling in the myocardium. Given the deleterious effects of continuous activation of this pathway, we propose Dlk1 as a new potential candidate for therapy in cases where aberrant TGFβ signalling leads to chronic fibrosis.
Collapse
Affiliation(s)
| | - Yassine Sassi
- Department of Medicine, Cardiovascular Research Institute
| | - Luca Troncone
- Department of Medicine, Cardiovascular Research Institute
| | - Ludovic Benard
- Department of Medicine, Cardiovascular Research Institute
| | | | - Ronald E Gordon
- Department of Pathology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Santiago Lamas
- Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Nicolás Cabrera 1, Campus UAM, Madrid, Spain
| | - Jorge Laborda
- Department of Inorganic and Organic Chemistry and Biochemistry, Pharmacy School/Biomedical Unit/CRIB, University of Castilla-La Mancha/CSIC, Dr. José María Sánchez Ibáñez Street, s/n Albacete, Spain
| | - Roger J Hajjar
- Department of Medicine, Cardiovascular Research Institute
| | - Djamel Lebeche
- Department of Medicine, Cardiovascular Research Institute
| |
Collapse
|
10
|
Generation of Inducible CRISPRi and CRISPRa Human Stromal/Stem Cell Lines for Controlled Target Gene Transcription during Lineage Differentiation. Stem Cells Int 2020; 2020:8857344. [PMID: 32922451 PMCID: PMC7453244 DOI: 10.1155/2020/8857344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/20/2020] [Accepted: 08/04/2020] [Indexed: 01/12/2023] Open
Abstract
Background Human bone marrow stromal/stem cells (hMSCs, also known as the skeletal stem cells or mesenchymal stem cells) are being employed to study lineage fate determination to osteoblasts, adipocytes, and chondrocytes. However, mechanistic studies employing hMSC have been hampered by the difficulty of deriving genetically modified cell lines due to the low and unstable transfection efficiency. Methods We infected hMSC with a CRISPR/Cas9 lentivirus system, with specific inducible dCas9-coupled transcription activator or repressor: dCas9-KRAB or dCas9-VP64, respectively, and established two hMSC lines (hMSC-CRISPRi and hMSC-CRISPRa) that can inhibit or activate gene expression, respectively. The two cell lines showed similar cell morphology, cell growth kinetics, and similar lineage differentiation potentials as the parental hMSC line. The expression of KRAB-dCas9 or VP64-dCas9 was controlled by the presence or absence of doxycycline (Dox) in the cell culturing medium. To demonstrate the functionality of the dCas9-effector hMSC system, we tested controlled expression of alkaline phosphatase (ALP) gene through transfection with the same single ALP sgRNA. Results In the presence of Dox, the expression of ALP showed 60-90% inhibition in hMSC-CRISPRi while ALP showed more than 20-fold increased expression in hMSC-CRISPRa. As expected, the ALP was functionally active and the cells showed evidence for inhibition or enhancement of in vitro osteoblast differentiation, respectively. Conclusion hMSC-CRISPRi and hMSC-CRISPRa are useful resources to study genes and genetic pathways regulating lineage-specific differentiation of hMSC.
Collapse
|
11
|
Bian Q, Cheng YH, Wilson JP, Su EY, Kim DW, Wang H, Yoo S, Blackshaw S, Cahan P. A single cell transcriptional atlas of early synovial joint development. Development 2020; 147:dev185777. [PMID: 32580935 PMCID: PMC7390639 DOI: 10.1242/dev.185777] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 06/09/2020] [Indexed: 12/14/2022]
Abstract
Synovial joint development begins with the formation of the interzone, a region of condensed mesenchymal cells at the site of the prospective joint. Recently, lineage-tracing strategies have revealed that Gdf5-lineage cells native to and from outside the interzone contribute to most, if not all, of the major joint components. However, there is limited knowledge of the specific transcriptional and signaling programs that regulate interzone formation and fate diversification of synovial joint constituents. To address this, we have performed single cell RNA-Seq analysis of 7329 synovial joint progenitor cells from the developing murine knee joint from E12.5 to E15.5. By using a combination of computational analytics, in situ hybridization and in vitro characterization of prospectively isolated populations, we have identified the transcriptional profiles of the major developmental paths for joint progenitors. Our freely available single cell transcriptional atlas will serve as a resource for the community to uncover transcriptional programs and cell interactions that regulate synovial joint development.
Collapse
Affiliation(s)
- Qin Bian
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Yu-Hao Cheng
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
- Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Jordan P Wilson
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Emily Y Su
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Hong Wang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Seth Blackshaw
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Patrick Cahan
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
- Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| |
Collapse
|
12
|
Abnormally localized DLK1 interacts with NCOR1 in non-small cell lung cancer cell nuclear. Biosci Rep 2019; 39:220954. [PMID: 31661545 PMCID: PMC6911156 DOI: 10.1042/bsr20192362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/18/2019] [Accepted: 10/28/2019] [Indexed: 02/07/2023] Open
Abstract
Delta-like homolog 1 (DLK1) regulates noncanonical Notch signaling pathway as ligand. DLK1 was abnormally expressed in a variety of tumors, affecting tumorigenesis and developments. The biological function of DLK1 toward cell proliferation and signaling activation was controversial across different cell types. Two currently known isoforms of DLK1, which are membrane-tethered isoform and soluble isoform, are believed to be the key of DLK1 dual behaviors. While these isoforms are not enough to explain the phenomena, our observations offer the possibility of a third isoform of DLK1. In the present study, we verified the nuclear localization of DLK1 in lung cancer cells. The nuclear localized DLK1 was observed in 107 of 351 non-small cell lung cancer (NSCLC) samples and was associated with tissue differentiation and tumor size. Through co-immunoprecipitation (co-IP) combined mass spectrometry (MS), we identified nuclear receptor corepressor 1 (NCOR1) as DLK1's novel interaction protein and confirmed their interaction in nuclear. We analyzed the expression of NCOR1 in two independent cohorts and demonstrated that NCOR1 is a tumor suppressor and has prognosis potential in lung squamous carcinomas. At last, we analyzed the colocalization of DLK1 and NCOR1 in 147 NSCLC samples by immunohistochemistry (IHC). The result indicated NCOR1 might participate with nuclear localized DLK1 in regulating cell differentiation.
Collapse
|
13
|
Traustadóttir GÁ, Lagoni LV, Ankerstjerne LBS, Bisgaard HC, Jensen CH, Andersen DC. The imprinted gene Delta like non-canonical Notch ligand 1 (Dlk1) is conserved in mammals, and serves a growth modulatory role during tissue development and regeneration through Notch dependent and independent mechanisms. Cytokine Growth Factor Rev 2019; 46:17-27. [DOI: 10.1016/j.cytogfr.2019.03.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/21/2019] [Accepted: 03/21/2019] [Indexed: 12/22/2022]
|
14
|
Garcia-Gallastegi P, Ruiz-García A, Ibarretxe G, Rivero-Hinojosa S, González-Siccha AD, Laborda J, Crende O, Unda F, García-Ramírez JJ. Similarities and differences in tissue distribution of DLK1 and DLK2 during E16.5 mouse embryogenesis. Histochem Cell Biol 2019; 152:47-60. [DOI: 10.1007/s00418-019-01778-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2019] [Indexed: 10/27/2022]
|
15
|
Chen L, Shi K, Andersen TL, Qiu W, Kassem M. KIAA1199 is a secreted molecule that enhances osteoblastic stem cell migration and recruitment. Cell Death Dis 2019; 10:126. [PMID: 30755597 PMCID: PMC6372631 DOI: 10.1038/s41419-018-1202-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022]
Abstract
Factors mediating mobilization of osteoblastic stem and progenitor cells from their bone marrow niche to be recruited to bone formation sites during bone remodeling are poorly known. We have studied secreted factors present in the bone marrow microenvironment and identified KIAA1199 (also known as CEMIP, cell migration inducing hyaluronan binding protein) in human bone biopsies as highly expressed in osteoprogenitor reversal cells (Rv.C) recruited to the eroded surfaces (ES), which are the future bone formation sites. In vitro, KIAA1199 did not affect the proliferation of human osteoblastic stem cells (also known as human bone marrow skeletal or stromal stem cells, hMSCs); but it enhanced cell migration as determined by scratch assay and trans-well migration assay. KIAA1199 deficient hMSCs (KIAA1199down) exhibited significant changes in cell size, cell length, ratio of cell width to length and cell roundness, together with reduction of polymerization actin (F-actin) and changes in phos-CFL1 (cofflin1), phos-LIMK1 (LIM domain kinase 1) and DSTN (destrin), key factors regulating actin cytoskeletal dynamics and cell motility. Moreover, KIAA1199down hMSC exhibited impaired Wnt signaling in TCF-reporter assay and decreased expression of Wnt target genes and these effects were rescued by KIAA1199 treatment. Finally, KIAA1199 regulated the activation of P38 kinase and its associated changes in Wnt-signaling. Thus, KIAA1199 is a mobilizing factor that interacts with P38 and Wnt signaling, and induces changes in actin cytoskeleton, as a mechanism mediating recruitment of hMSC to bone formation sites.
Collapse
Affiliation(s)
- Li Chen
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark.
| | - Kaikai Shi
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark
| | - Thomas Levin Andersen
- Department of Clinical Cell Biology, Vejle Hospital-Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, 7100, Vejle, Denmark
| | - Weimin Qiu
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark.
- The Danish Stem Cell Center (DanStem), University of Copenhagen, 2200, Copenhagen, Denmark.
- Stem Cell Unit, Department of Anatomy, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
16
|
Figeac F, Andersen DC, Nipper Nielsen CA, Ditzel N, Sheikh SP, Skjødt K, Kassem M, Jensen CH, Abdallah BM. Antibody-based inhibition of circulating DLK1 protects from estrogen deficiency-induced bone loss in mice. Bone 2018; 110:312-320. [PMID: 29499415 DOI: 10.1016/j.bone.2018.02.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 02/26/2018] [Accepted: 02/26/2018] [Indexed: 11/23/2022]
Abstract
Soluble delta-like 1 homolog (DLK1) is a circulating protein that belongs to the Notch/Serrate/delta family, which regulates many differentiation processes including osteogenesis and adipogenesis. We have previously demonstrated an inhibitory effect of DLK1 on bone mass via stimulation of bone resorption and inhibition of bone formation. Further, serum DLK1 levels are elevated and positively correlated to bone turnover markers in estrogen (E)-deficient rodents and women. In this report, we examined whether inhibition of serum DLK1 activity using a neutralizing monoclonal antibody protects from E deficiency-associated bone loss in mice. Thus, we generated mouse monoclonal anti-mouse DLK1 antibodies (MAb DLK1) that enabled us to reduce and also quantitate the levels of bioavailable serum DLK1 in vivo. Ovariectomized (ovx) mice were injected intraperitoneally twice weekly with MAb DLK1 over a period of one month. DEXA-, microCT scanning, and bone histomorphometric analyses were performed. Compared to controls, MAb DLK1 treated ovx mice were protected against ovx-induced bone loss, as revealed by significantly increased total bone mass (BMD) due to increased trabecular bone volume fraction (BV/TV) and inhibition of bone resorption. No significant changes were observed in total fat mass or in the number of bone marrow adipocytes. These results support the potential use of anti-DLK1 antibody therapy as a novel intervention to protect from E deficiency associated bone loss.
Collapse
Affiliation(s)
- Florence Figeac
- Molecular Endocrinology Lab. (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Ditte C Andersen
- Laboratory of Molecular and Cellular Cardiology, Dep. of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Danish Center for Regenerative Medicine, Odense University Hospital, Denmark; Clinical Institute (University of Southern Denmark), Winsløwparken 213rd, Odense, Denmark
| | - Casper A Nipper Nielsen
- Department of Cancer and Inflammation, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Nicholas Ditzel
- Molecular Endocrinology Lab. (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Søren P Sheikh
- Laboratory of Molecular and Cellular Cardiology, Dep. of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Danish Center for Regenerative Medicine, Odense University Hospital, Denmark; Department of Cardiac and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Karsten Skjødt
- Department of Cancer and Inflammation, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Moustapha Kassem
- Molecular Endocrinology Lab. (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; Department of Cellular and Molecular Medicine, DanStem (Danish Stem Cell Center), Panum Institute, University of Copenhagen, Copenhagen, Denmark; Stem Cell Unit, Department of Anatomy, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Charlotte H Jensen
- Laboratory of Molecular and Cellular Cardiology, Dep. of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Danish Center for Regenerative Medicine, Odense University Hospital, Denmark; Department of Cardiac and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| | - Basem M Abdallah
- Molecular Endocrinology Lab. (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; Department of Biological Sciences, College of Science, King Faisal University, Hofuf, Saudi Arabia.
| |
Collapse
|
17
|
Qi S, Zhu X, Wang X, Chen F, Yan Y, Shang G, Chen W. Role of protein delta homolog 1 in the proliferation and differentiation of ameloblasts. Mol Med Rep 2017; 17:3537-3544. [PMID: 29257328 PMCID: PMC5802151 DOI: 10.3892/mmr.2017.8290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/12/2017] [Indexed: 12/25/2022] Open
Abstract
Protein delta homolog 1 (DLK1) regulates the odontoblastic differentiation of human dental pulp stem cells. It was hypothesized that DLK1 may exert regulatory effects on epithelial‑mesenchymal interactions in tooth development. The present study investigated the expression of DLK1 during the development of mouse enamel and its role in the proliferation and differentiation of ameloblast‑lineage cells (ALCs). DLK1 expression was upregulated in ameloblasts in the first mandibular molar during the entire process of enamel development. The mRNA and protein levels of DLK1 were significantly upregulated following ameloblastic induction in ALCs. In addition, overexpression of DLK1 promoted the proliferation of ALCs, inhibited ameloblastic differentiation, upregulated the expression of amelogenin and enamelin, and downregulated the expression of odontogenic ameloblast‑associated protein and kallikrein 4. The results of the present study suggested that DLK1 may be a potent regulator of ameloblast proliferation and differentiation, and may regulate enamel formation during tooth development.
Collapse
Affiliation(s)
- Shengcai Qi
- Department of Oral and Maxillofacial‑Head and Neck Oncology, and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Xueqin Zhu
- Department of Oral and Maxillofacial‑Head and Neck Oncology, and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Xiaoning Wang
- Department of Oral and Maxillofacial‑Head and Neck Oncology, and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Fubo Chen
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Yanhong Yan
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, P.R. China
| | - Guangwei Shang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Wantao Chen
- Department of Oral and Maxillofacial‑Head and Neck Oncology, and Faculty of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
18
|
Xu W, Wang Y, Zhao H, Fan B, Guo K, Cai M, Zhang S. Delta-like 2 negatively regulates chondrogenic differentiation. J Cell Physiol 2017; 233:6574-6582. [PMID: 29057471 DOI: 10.1002/jcp.26244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 10/13/2017] [Indexed: 01/21/2023]
Abstract
Delta-like 2 (Dlk2), a glycoprotein highly homologous to Dlk1, belongs to the Notch/Delta/Serrata family. Dlk2 has been shown to be an important regulator of adipogenesis; however, its role in other cellular differentiation processes is still unknown. Therefore, in this study, we aimed to determine the role of Dlk2 in chondrogenic differentiation. We found that Dlk2 overexpression promoted the growth of ATDC5 cells but inhibited insulin-induced ATDC5 chondrogenic differentiation, as supported by the reduction in cartilage matrix formation and gene expression of aggrecan (acan), collagentype II (col2a1) and X (col10a1). In contrast, Dlk2 silencing inhibited the proliferation of ATDC5 cells but enhanced their chondrogenic differentiation. We then evaluated the roles of mitogen-activated protein kinases (MAPKs), which are activated by insulin during the chondrogenesis of ATDC5 cells. Overexpression of Dlk2 protein strongly promoted the activation of p38, but not extracellular signal-regulated kinase (ERK) 1/2 and c-Jun N-terminal kinase (JNK). Moreover, as expected, Dlk2 silencing inhibited the activation of p38, but had no effect on the ERK1/2 and JNK pathways. Finally, we also detected the expression of Dlk2 in mouse epiphyseal cartilage during embryo development. The expression of the Dlk2 protein in the limb bud could be detected at embryonic day 11.5; additionally, it was found to decrease in the superficial zones, but remained unchanged in the deep/hypertrophic zones. In conclusion, our results suggested that Dlk2 acted as an important regulator of chondrogenesis through the p38 pathway. These findings may lead to strategies for the treatment of cartilage-related diseases such as osteoarthritis.
Collapse
Affiliation(s)
- Weifeng Xu
- Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yexin Wang
- Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Haoming Zhao
- Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Baotin Fan
- Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Ke Guo
- Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Ming Cai
- Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Department of Craniomaxillofacial Surgery, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Shanyong Zhang
- Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Department of Oral Surgery, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
19
|
Nueda ML, Naranjo AI, Baladrón V, Laborda J. Different expression levels of DLK1 inversely modulate the oncogenic potential of human MDA-MB-231 breast cancer cells through inhibition of NOTCH1 signaling. FASEB J 2017; 31:3484-3496. [PMID: 28461338 DOI: 10.1096/fj.201601341rrr] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/11/2017] [Indexed: 01/01/2023]
Abstract
NOTCH receptors participate in cancer cell proliferation and survival. Accumulated evidence indicates that, depending on the cellular context, these receptors can function as oncogenes or as tumor-suppressor genes. The epidermal growth factor-like protein delta-like homolog (DLK)1 acts as a NOTCH inhibitor and is involved in the regulation of normal and tumoral growth. In this work, we focused on the role of DLK1 in the control of breast cancer cell growth, a tumor type in which NOTCH receptors have been shown to play both opposite roles. We found that human DLK1 inhibits NOTCH signaling in MDA-MB-231 breast cancer cells. The proliferation rate and invasion capabilities of these cells depended on the level of NOTCH activation and signaling, as regulated by DLK1. High levels of DLK1 expression led to a significant decrease in NOTCH signaling, which was associated with a decrease in breast cancer cell proliferation and invasion. On the contrary, lower levels of NOTCH inhibition, caused by lower levels of DLK1 overexpression, led to enhanced in vitro MDA-MB-231 cell invasion, and to both in vitro and in vivo increased cell proliferation. The data presented in this work suggest that a fine regulation of NOTCH signaling plays an important role in the control of breast cancer cell proliferation and invasion.-Nueda, M.-L., Naranjo, A.-I., Baladrón V., Laborda, J. Different expression levels of DLK1 inversely modulate the oncogenic potential of human MDA-MB-231 breast cancer cells through inhibition of NOTCH1 signaling.
Collapse
Affiliation(s)
- María-Luisa Nueda
- Biochemistry and Molecular Biology Branch-Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha, Spanish National Research Council (CSIC), Albacete, Spain .,School of Pharmacy, University of Castilla-La Mancha, Spanish National Research Council (CSIC), Albacete, Spain
| | - Ana-Isabel Naranjo
- Biochemistry and Molecular Biology Branch-Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha, Spanish National Research Council (CSIC), Albacete, Spain.,School of Medicine, University of Castilla-La Mancha, Spanish National Research Council (CSIC), Albacete, Spain and.,Regional Center for Biomedical Research (CRIB)-Biomedicine Unit, University of Castilla-La Mancha, Spanish National Research Council (CSIC), Albacete, Spain
| | - Victoriano Baladrón
- Biochemistry and Molecular Biology Branch-Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha, Spanish National Research Council (CSIC), Albacete, Spain.,School of Medicine, University of Castilla-La Mancha, Spanish National Research Council (CSIC), Albacete, Spain and.,Regional Center for Biomedical Research (CRIB)-Biomedicine Unit, University of Castilla-La Mancha, Spanish National Research Council (CSIC), Albacete, Spain
| | - Jorge Laborda
- Biochemistry and Molecular Biology Branch-Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha, Spanish National Research Council (CSIC), Albacete, Spain .,School of Pharmacy, University of Castilla-La Mancha, Spanish National Research Council (CSIC), Albacete, Spain
| |
Collapse
|
20
|
Traustadóttir GÁ, Jensen CH, Garcia Ramirez JJ, Beck HC, Sheikh SP, Andersen DC. The non-canonical NOTCH1 ligand Delta-like 1 homolog (DLK1) self interacts in mammals. Int J Biol Macromol 2017; 97:460-467. [DOI: 10.1016/j.ijbiomac.2017.01.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 12/11/2022]
|
21
|
Qi S, Yan Y, Wen Y, Li J, Wang J, Chen F, Tang X, Shang G, Xu Y, Wang R. The effect of delta-like 1 homologue on the proliferation and odontoblastic differentiation in human dental pulp stem cells. Cell Prolif 2017; 50. [PMID: 28205268 DOI: 10.1111/cpr.12335] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 01/03/2017] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION This study aimed to investigate the functions of delta-like homologue 1 (DLK1) in the proliferation and differentiation of human dental pulp stem cells (hDPSCs). METHODS Immunohistochemical analysis was used to determine the expression of alkaline phosphatase (ALP), dentin sialophosphoprotein (DSPP), DLK1, NOTCH1 and p-ERK1/2 in the mouse first maxillary molar. Recombinant lentivirus was constructed to overexpress DLK1 stably in hDPSCs. The cell viability and proliferation of hDPSCs were examined by CCK8 and EdU incorporation assay respectively. The odontoblastic differentiation of hDPSCs was determined by detection of ALPase activity assay, ALP and alizarin red staining and the expression of mineralization-related genes including ALP, DSPP and dental matrix protein. The mRNA and protein levels of DLK1 and p-ERK1/2 protein expression were detected. ERK inhibitor was used to test the differentiation effect of DLK1 on hDPSCs. RESULTS Delta-like homologue 1 was highly expressed on the odontoblasts and dental pulp cells on the first maxillary molar; the expression of p-ERK1/2 is similar with the DLK1 in the same process. The expression level of DLK1 increased significantly after the odontoblastic induction of hDPSCs. DLK1 overexpression increased the proliferation ability of hDPSCs and inhibited odontoblastic differentiation of hDPSCs. The protein level of p-ERK1/2 significantly increased in hDPSCs/dlk1-oe group. ERK signalling pathway inhibitor reversed the odontoblastic differentiation effects of DLK1 on hDPSCs. CONCLUSIONS The proliferation of hDPSCs was promoted after DLK1 overexpression. DLK1 inhibited the odontoblastic differentiation of hDPSCs, which maybe through ERK signalling pathway.
Collapse
Affiliation(s)
- Shengcai Qi
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanhong Yan
- Department of Pediatric Dentistry, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yue Wen
- Institute of Stomatology, Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jialiang Li
- Institute of Stomatology, Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jing Wang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fubo Chen
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoshan Tang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guangwei Shang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanzhi Xu
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Raorao Wang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Characterization of Cellular and Molecular Heterogeneity of Bone Marrow Stromal Cells. Stem Cells Int 2016; 2016:9378081. [PMID: 27610142 PMCID: PMC5004045 DOI: 10.1155/2016/9378081] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/26/2016] [Indexed: 01/04/2023] Open
Abstract
Human bone marrow-derived stromal stem cells (hBMSC) exhibit multiple functions, including differentiation into skeletal cells (progenitor function), hematopoiesis support, and immune regulation (nonprogenitor function). We have previously demonstrated the presence of morphological and functional heterogeneity of hBMSC cultures. In the present study, we characterized in detail two hTERT-BMSC clonal cell populations termed here CL1 and CL2 that represent an opposing phenotype with respect to morphology, markers expression: alkaline phosphatase (ALP) and CD146, and ex vivo differentiation potential. CL1 differentiated readily to osteoblasts, adipocytes, and chondrocytes as shown by expression of lineage specific genes and proteins. Whole genome transcriptome profiling of CL1 versus CL2 revealed enrichment in CL1 of bone-, mineralization-, and skeletal muscle-related genes, for example, ALP, POSTN, IGFBP5 BMP4, and CXCL12. On the other hand, CL2 transcriptome was enriched in immune modulatory genes, for example, CD14, CD99, NOTCH3, CXCL6, CFB, and CFI. Furthermore, gene expression microarray analysis of osteoblast differentiated CL1 versus CL2 showed significant upregulation in CL1 of bone development and osteoblast differentiation genes which included several homeobox genes: TBX15, HOXA2 and HOXA10, and IGF1, FGFR3, BMP6, MCAM, ITGA10, IGFBP5, and ALP. siRNA-based downregulation of the ALP gene in CL1 impaired osteoblastic and adipocytic differentiation. Our studies demonstrate the existence of molecular and functional heterogeneity in cultured hBMSC. ALP can be employed to identify osteoblastic and adipocytic progenitor cells in the heterogeneous hBMSC cultures.
Collapse
|
23
|
Hu J, Zhao W, Zhan S, Xiao P, Zhou J, Wang L, Li L, Zhang H, Niu L, Zhong T. Delta-like 1 homolog in Capra hircus: molecular characteristics, expression pattern and phylogeny. Mol Biol Rep 2016; 43:563-71. [PMID: 27108112 DOI: 10.1007/s11033-016-3989-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 04/19/2016] [Indexed: 01/24/2023]
Abstract
To research the molecular characteristics, expression pattern and phylogeny of the Delta-like 1 homolog gene (Dlk1) in goats. Dlk1 transcripts were identified in the Jianyang Da'er goats by reverse-transcription polymerase chain reaction (RT-PCR). Phylogenetic trees were constructed by Bayesian inference and neighbor-joining methods. Quantitative real-time PCR (qPCR), western blotting and in situ hybridization were performed to analyze the expression pattern of Dlk1. Five alternatively transcripts were identified in different tissues and designated as Dlk1-AS1, 2, 3, 4 and 5. Compared with the normal transcript Dlk1-AS1, Dlk1-AS4 and Dlk1-AS5 retained the identical open reading frame (ORF) and encoded proteins with truncated epidermal-growth-factor like repeats of 121 and 83 amino acids, respectively. Using the Bayesian inference method, the consensus phylogenetic tree indicated that caprine Dlk1 had a closer relationship with bovine Dlk1 than with Dlk1 from pigs, humans and mice. qPCR revealed high expression levels of Dlk1 in the kidney (P < 0.01). However, mRNA and protein levels presented an inconsistent correlation, possibly because of post-transcriptional regulation. RNA in situ hybridization indicated that Dlk1 mRNA was localized in the interlobular bile duct and alongside the hepatocyte nuclei, in the epithelial cells of proximal and distal convoluted tubules and in the connective region between the mesothelium and myocardium in the heart. The Dlk1 gene in goats produces alternatively spliced transcripts, with specific expression and cellular localization patterns. These findings would lay the foundation for further study.
Collapse
Affiliation(s)
- Jiangtao Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Wei Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Siyuan Zhan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ping Xiao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jingxuan Zhou
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Linjie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lili Niu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Tao Zhong
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China.
| |
Collapse
|
24
|
García-Gallastegui P, Luzuriaga J, Aurrekoetxea M, Baladrón V, Ruiz-Hidalgo MJ, García-Ramírez JJ, Laborda J, Unda F, Ibarretxe G. Reduced salivary gland size and increased presence of epithelial progenitor cells in DLK1-deficient mice. Cell Tissue Res 2015; 364:513-525. [DOI: 10.1007/s00441-015-2344-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 12/10/2015] [Indexed: 01/23/2023]
|
25
|
Abdallah BM, Ditzel N, Laborda J, Karsenty G, Kassem M. DLK1 Regulates Whole-Body Glucose Metabolism: A Negative Feedback Regulation of the Osteocalcin-Insulin Loop. Diabetes 2015; 64:3069-80. [PMID: 25918236 DOI: 10.2337/db14-1642] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/21/2015] [Indexed: 11/13/2022]
Abstract
The endocrine role of the skeleton in regulating energy metabolism is supported by a feed-forward loop between circulating osteoblast (OB)-derived undercarboxylated osteocalcin (Glu-OCN) and pancreatic β-cell insulin; in turn, insulin favors osteocalcin (OCN) bioactivity. These data suggest the existence of a negative regulation of this cross talk between OCN and insulin. Recently, we identified delta like-1 (DLK1) as an endocrine regulator of bone turnover. Because DLK1 is colocalized with insulin in pancreatic β-cells, we examined the role of DLK1 in insulin signaling in OBs and energy metabolism. We show that Glu-OCN specifically stimulates Dlk1 expression by the pancreas. Conversely, Dlk1-deficient (Dlk1(-/-) ) mice exhibited increased circulating Glu-OCN levels and increased insulin sensitivity, whereas mice overexpressing Dlk1 in OB displayed reduced insulin secretion and sensitivity due to impaired insulin signaling in OB and lowered Glu-OCN serum levels. Furthermore, Dlk1(-/-) mice treated with Glu-OC experienced significantly lower blood glucose levels than Glu-OCN-treated wild-type mice. The data suggest that Glu-OCN-controlled production of DLK1 by pancreatic β-cells acts as a negative feedback mechanism to counteract the stimulatory effects of insulin on OB production of Glu-OCN, a potential mechanism preventing OCN-induced hypoglycemia.
Collapse
Affiliation(s)
- Basem M Abdallah
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital and University of Southern Denmark, Odense, Denmark
| | - Nicholas Ditzel
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital and University of Southern Denmark, Odense, Denmark
| | - Jorge Laborda
- Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha Medical School, Albacete, Spain
| | - Gerard Karsenty
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Moustapha Kassem
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital and University of Southern Denmark, Odense, Denmark DanStem (Danish Stem Cell Center), Panum Institute, University of Copenhagen, Copenhagen, Denmark Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
26
|
González MJ, Ruiz-García A, Monsalve EM, Sánchez-Prieto R, Laborda J, Díaz-Guerra MJM, Ruiz-Hidalgo MJ. DLK1 is a novel inflammatory inhibitor which interferes with NOTCH1 signaling in TLR-activated murine macrophages. Eur J Immunol 2015; 45:2615-27. [PMID: 26115479 DOI: 10.1002/eji.201545514] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/19/2015] [Accepted: 06/23/2015] [Indexed: 12/19/2022]
Abstract
Delta-like protein 1 (DLK1) is a noncanonical ligand that inhibits NOTCH1 receptor activity and regulates multiple differentiation processes. In macrophages, NOTCH signaling increases TLR-induced expression of key pro-inflammatory mediators. We have investigated the role of DLK1 in macrophage activation and inflammation using Dlk1-deficient mice and Raw 264.7 cells overexpressing Dlk1. In the absence of Dlk1, NOTCH1 expression is increased and the activation of macrophages with TLR3 or TLR4 agonists leads to higher production of IFN-β and other pro-inflammatory cytokines, including TNF-α, IL-12, and IL-23. The expression of key proteins involved in IFN-β signaling, such as IRF3, IRF7, IRF1, or STAT1, as well as cRel, or RelB, which are responsible for the generation of IL-12 and IL-23, is enhanced in Dlk1 KO macrophages. Consistently, Dlk1 KO mice are more sensitive to LPS-induced endotoxic shock. These effects seem to be mediated through the modulation of NOTCH1 signaling. TLR4 activation reduces DLK1 expression, whereas increases NOTCH1 levels. In addition, DLK1 expression diminishes during differentiation of human U937 cells to macrophages. Overall, these results reveal a novel role for DLK1 as a regulator of NOTCH-mediated, pro-inflammatory macrophage activation, which could help to ensure a baseline level preventing constant tissue inflammation.
Collapse
Affiliation(s)
- María J González
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Almudena Ruiz-García
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Eva M Monsalve
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Ricardo Sánchez-Prieto
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Jorge Laborda
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - María J M Díaz-Guerra
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - María J Ruiz-Hidalgo
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| |
Collapse
|
27
|
Howard M, Charalambous M. Molecular basis of imprinting disorders affecting chromosome 14: lessons from murine models. Reproduction 2015; 149:R237-49. [DOI: 10.1530/rep-14-0660] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Uniparental inheritance of chromosome 14q32 causes developmental failure during gestation and early postnatal development due to mis-expression of a cluster of imprinted genes under common epigenetic control. Two syndromes associated with chromosome 14q32 abnormalities have been described, Kagami–Ogata and Temple syndromes. Both of these syndromes are characterised by specific impairments of intrauterine development, placentation and early postnatal survival. Such abnormalities arise because the processes of intrauterine growth and postnatal adaptation are critically modulated by the dosage of imprinted genes in the chromosome 14q32 cluster. Much of our understanding of how the imprinted genes in this cluster are regulated, as well as their individual functions in the molecular pathways controlling growth and postnatal adaptation, has come from murine models. Mouse chromosome 12qF1 contains an imprinted region syntenic to human chromosome 14q32, collectively referred to as the Dlk1–Dio3 cluster. In this review, we will summarise the wealth of information derived from animal models of chromosome 12 imprinted gene mis-regulation, and explore the relationship between the functions of individual genes and the phenotypic result of their mis-expression. As there is often a considerable overlap between the functions of genes in the Dlk1–Dio3 cluster, we propose that the expression dosage of these genes is controlled by common regulatory mechanisms to co-ordinate the timing of growth and postnatal adaptation. While the diseases associated with mis-regulated chromosome 14 imprinting are rare, studies carried out in mice on the functions of the affected genes as well as their normal regulatory mechanisms have revealed new mechanistic pathways for the control of growth and survival in early life.
Collapse
|
28
|
Liechti R, Ducray AD, Jensen P, Di Santo S, Seiler S, Jensen CH, Meyer M, Widmer HR. Characterization of fetal antigen 1/delta-like 1 homologue expressing cells in the rat nigrostriatal system: effects of a unilateral 6-hydroxydopamine lesion. PLoS One 2015; 10:e0116088. [PMID: 25723595 PMCID: PMC4344227 DOI: 10.1371/journal.pone.0116088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/04/2014] [Indexed: 11/18/2022] Open
Abstract
Fetal antigen 1/delta-like 1 homologue (FA1/dlk1) belongs to the epidermal growth factor superfamily and is considered to be a non-canonical ligand for the Notch receptor. Interactions between Notch and its ligands are crucial for the development of various tissues. Moreover, FA1/dlk1 has been suggested as a potential supplementary marker of dopaminergic neurons. The present study aimed at investigating the distribution of FA1/dlk1-immunoreactive (-ir) cells in the early postnatal and adult midbrain as well as in the nigrostriatal system of 6-hydroxydopamine (6-OHDA)-lesioned hemiparkinsonian adult rats. FA1/dlk1-ir cells were predominantly distributed in the substantia nigra (SN) pars compacta (SNc) and in the ventral tegmental area. Interestingly, the expression of FA1/dlk1 significantly increased in tyrosine hydroxylase (TH)-ir cells during early postnatal development. Co-localization and tracing studies demonstrated that FA1/dlk1-ir cells in the SNc were nigrostriatal dopaminergic neurons, and unilateral 6-OHDA lesions resulted in loss of both FA1/dlk1-ir and TH-ir cells in the SNc. Surprisingly, increased numbers of FA1/dlk1-ir cells (by 70%) were detected in dopamine-depleted striata as compared to unlesioned controls. The higher number of FA1/dlk1-ir cells was likely not due to neurogenesis as colocalization studies for proliferation markers were negative. This suggests that FA1/dlk1 was up-regulated in intrinsic cells in response to the 6-OHDA-mediated loss of FA1/dlk1-expressing SNc dopaminergic neurons and/or due to the stab wound. Our findings hint to a significant role of FA1/dlk1 in the SNc during early postnatal development. The differential expression of FA1/dlk1 in the SNc and the striatum of dopamine-depleted rats could indicate a potential involvement of FA1/dlk1 in the cellular response to the degenerative processes.
Collapse
Affiliation(s)
- Rémy Liechti
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster University of Bern, Inselspital, CH-3010 Berne, Switzerland
| | - Angélique D. Ducray
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster University of Bern, Inselspital, CH-3010 Berne, Switzerland
| | - Pia Jensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Winsløwparken 21, DK-5000 Odense C, Denmark
| | - Stefano Di Santo
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster University of Bern, Inselspital, CH-3010 Berne, Switzerland
| | - Stefanie Seiler
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster University of Bern, Inselspital, CH-3010 Berne, Switzerland
| | - Charlotte H. Jensen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Sdr. Boulevard 29, DK-5000, Odense C, Denmark
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Winsløwparken 21, DK-5000 Odense C, Denmark
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Winsløwparken 21, DK-5000 Odense C, Denmark
| | - Hans Rudolf Widmer
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster University of Bern, Inselspital, CH-3010 Berne, Switzerland
- * E-mail:
| |
Collapse
|
29
|
Abdallah BM, Jafari A, Zaher W, Qiu W, Kassem M. Skeletal (stromal) stem cells: an update on intracellular signaling pathways controlling osteoblast differentiation. Bone 2015; 70:28-36. [PMID: 25138551 DOI: 10.1016/j.bone.2014.07.028] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 07/22/2014] [Accepted: 07/24/2014] [Indexed: 01/06/2023]
Abstract
Skeletal (marrow stromal) stem cells (BMSCs) are a group of multipotent cells that reside in the bone marrow stroma and can differentiate into osteoblasts, chondrocytes and adipocytes. Studying signaling pathways that regulate BMSC differentiation into osteoblastic cells is a strategy for identifying druggable targets for enhancing bone formation. This review will discuss the functions and the molecular mechanisms of action on osteoblast differentiation and bone formation; of a number of recently identified regulatory molecules: the non-canonical Notch signaling molecule Delta-like 1/preadipocyte factor 1 (Dlk1/Pref-1), the Wnt co-receptor Lrp5 and intracellular kinases. This article is part of a Special Issue entitled: Stem Cells and Bone.
Collapse
Affiliation(s)
- Basem M Abdallah
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Abbas Jafari
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; DanStem (Danish Stem Cell Center), Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Walid Zaher
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Saudi Arabia
| | - Weimin Qiu
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Moustapha Kassem
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; DanStem (Danish Stem Cell Center), Panum Institute, University of Copenhagen, Copenhagen, Denmark; Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Saudi Arabia.
| |
Collapse
|
30
|
Madon-Simon M, Cowley M, Garfield AS, Moorwood K, Bauer SR, Ward A. Antagonistic roles in fetal development and adult physiology for the oppositely imprinted Grb10 and Dlk1 genes. BMC Biol 2014; 12:771. [PMID: 25551289 PMCID: PMC4280702 DOI: 10.1186/s12915-014-0099-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/07/2014] [Indexed: 12/14/2022] Open
Abstract
Background Despite being a fundamental biological problem the control of body size and proportions during development remains poorly understood, although it is accepted that the insulin-like growth factor (IGF) pathway has a central role in growth regulation, probably in all animals. The involvement of imprinted genes has also attracted much attention, not least because two of the earliest discovered were shown to be oppositely imprinted and antagonistic in their regulation of growth. The Igf2 gene encodes a paternally expressed ligand that promotes growth, while maternally expressed Igf2r encodes a cell surface receptor that restricts growth by sequestering Igf2 and targeting it for lysosomal degradation. There are now over 150 imprinted genes known in mammals, but no other clear examples of antagonistic gene pairs have been identified. The delta-like 1 gene (Dlk1) encodes a putative ligand that promotes fetal growth and in adults restricts adipose deposition. Conversely, Grb10 encodes an intracellular signalling adaptor protein that, when expressed from the maternal allele, acts to restrict fetal growth and is permissive for adipose deposition in adulthood. Results Here, using knockout mice, we present genetic and physiological evidence that these two factors exert their opposite effects on growth and physiology through a common signalling pathway. The major effects are on body size (particularly growth during early life), lean:adipose proportions, glucose regulated metabolism and lipid storage in the liver. A biochemical pathway linking the two cell signalling factors remains to be defined. Conclusions We propose that Dlk1 and Grb10 define a mammalian growth axis that is separate from the IGF pathway, yet also features an antagonistic imprinted gene pair. Electronic supplementary material The online version of this article (doi:10.1186/s12915-014-0099-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | - Andrew Ward
- Department of Biology & Biochemistry and Centre for Regenerative Medicine, University of Bath, Building 4 South, Claverton Down, Bath BA2 7AY, UK.
| |
Collapse
|
31
|
Delta-Like Homologue 1 and Its Role in the Bone Marrow Niche and Hematologic Malignancies. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2014; 14:451-5. [DOI: 10.1016/j.clml.2014.06.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 06/08/2014] [Accepted: 06/17/2014] [Indexed: 01/08/2023]
|
32
|
Delta-like 1 homolog (DLK1) inhibits proliferation and myotube formation of avian QM7 myoblasts. Comp Biochem Physiol B Biochem Mol Biol 2014; 179:37-43. [PMID: 25250736 DOI: 10.1016/j.cbpb.2014.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/12/2014] [Accepted: 09/13/2014] [Indexed: 11/23/2022]
Abstract
Delta-like 1 homolog (DLK1) has been implicated as an important regulator in mammalian muscle development. Our previous studies showed that different alternative splicing isoforms have distinct functions in the regulation of myogenesis in mice. Unlike most mammals, including mice, pigs, cattle, and sheep, DLK1 mRNA for avian species has a single form without alternative splicing. In the current study, we have used QM7 cells, a quail myoblast, to study the role of DLK1 in the regulation of avian myogenesis. Overexpression of DLK1 inhibited myogenesis with a lower fusion rate and thinner myotube compared to the control QM7 cells. Comparison of relative levels of protein and mRNA showed down-regulation of PAX7, MYOG, and MHC, and up-regulation of MYOD by DLK1, suggesting that quail DLK1 inhibits myogenesis at later stages of myogenic differentiation and myotube formation. DLK1 reduced the QM7 cell growth rate which is accompanied by a lower percentage of bromodeoxyuridine positive cells, indicating an inhibitory role of DLK1 in proliferation. During the early post-hatch ages, the relatively slower increase in the amount of total DNA mass in breast muscle of the heavy weight quail line, that has been selected for over 40 generations, could be partially explained by the higher expression of DLK1 compared to the control quail. Taken together, DLK1 inhibits myogenic differentiation and proliferation by regulating the expression levels of myogenic factors in quail. In addition, the regulation of expression level and cleavage of full-length DLK1 may be important factors for regulating myogenesis in quail having no splicing variants of DLK1.
Collapse
|
33
|
Abstract
The ATDC5 cell line is derived from mouse teratocarcinoma cells and characterized as a chondrogenic cell line which goes through a sequential process analogy to chondrocyte differentiation. Thus, it is regarded as a promising in vitro model to study the factors that influence cell behaviors during chondrogenesis. It also provides insights in exploring signaling pathways related to skeletal development as well as interactions with innovative materials. To date, over 200 studies have utilized ATDC5 to obtain lots of significant findings. In this review, we summarized the literature of ATDC5 related studies and emphasized the application of ATDC5 in chondrogenesis. In addition, the general introduction of ATDC5 including its derivation and characterization is covered in this article.
Collapse
Affiliation(s)
- Yongchang Yao
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | | |
Collapse
|
34
|
The proteins DLK1 and DLK2 modulate NOTCH1-dependent proliferation and oncogenic potential of human SK-MEL-2 melanoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2674-84. [PMID: 25093684 DOI: 10.1016/j.bbamcr.2014.07.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 07/25/2014] [Accepted: 07/28/2014] [Indexed: 01/06/2023]
Abstract
NOTCH receptors regulate cell proliferation and survival in several types of cancer cells. Depending on the cellular context, NOTCH1 can function as an oncogene or as a tumor suppressor gene. DLK1 is also involved in the regulation of cell growth and cancer, but nothing is known about the role of DLK2 in these processes. Recently, the proteins DLK1 and DLK2 have been reported to interact with NOTCH1 and to inhibit NOTCH1 activation and signaling in different cell lines. In this work, we focused on the role of DLK proteins in the control of melanoma cell growth, where NOTCH1 is known to exert an oncogenic effect. We found that human DLK proteins inhibit NOTCH signaling in SK-MEL-2 metastatic melanoma cells. Moreover, the proliferation rate of these cells was dependent upon the level of NOTCH activation and signaling as regulated by DLK proteins. In particular, high levels of NOTCH inhibition resulted in a decrease, whereas lower levels of NOTCH inhibition led to an increase in melanoma cell proliferation rates, both in vitro and in vivo. Finally, our data revealed additive NOTCH-mediated effects of DLK proteins and the γ-secretase inhibitor DAPT on cell proliferation. The data presented in this work suggest that a fine regulation of NOTCH signaling plays an important role in the control of metastatic melanoma cell proliferation. Our results open the way to new research on the role of DLK proteins as potential therapeutic tools for the treatment of human melanoma.
Collapse
|
35
|
Lottrup G, Nielsen J, Maroun L, Møller L, Yassin M, Leffers H, Skakkebæk N, Rajpert-De Meyts E. Expression patterns of DLK1 and INSL3 identify stages of Leydig cell differentiation during normal development and in testicular pathologies, including testicular cancer and Klinefelter syndrome. Hum Reprod 2014; 29:1637-50. [DOI: 10.1093/humrep/deu124] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
36
|
Doublecortin may play a role in defining chondrocyte phenotype. Int J Mol Sci 2014; 15:6941-60. [PMID: 24758934 PMCID: PMC4013671 DOI: 10.3390/ijms15046941] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/03/2014] [Accepted: 04/14/2014] [Indexed: 01/01/2023] Open
Abstract
Embryonic development of articular cartilage has not been well understood and the role of doublecortin (DCX) in determination of chondrocyte phenotype is unknown. Here, we use a DCX promoter-driven eGFP reporter mouse model to study the dynamic gene expression profiles in mouse embryonic handplates at E12.5 to E13.5 when the condensed mesenchymal cells differentiate into either endochondral chondrocytes or joint interzone cells. Illumina microarray analysis identified a variety of genes that were expressed differentially in the different regions of mouse handplate. The unique expression patterns of many genes were revealed. Cytl1 and 3110032G18RIK were highly expressed in the proximal region of E12.5 handplate and the carpal region of E13.5 handplate, whereas Olfr538, Kctd15, and Cited1 were highly expressed in the distal region of E12.5 and the metacarpal region of E13.5 handplates. There was an increasing gradient of Hrc expression in the proximal to distal direction in E13.5 handplate. Furthermore, when human DCX protein was expressed in human adipose stem cells, collagen II was decreased while aggrecan, matrilin 2, and GDF5 were increased during the 14-day pellet culture. These findings suggest that DCX may play a role in defining chondrocyte phenotype.
Collapse
|
37
|
Membrane-bound delta-like 1 homolog (Dlk1) promotes while soluble Dlk1 inhibits myogenesis in C2C12 cells. FEBS Lett 2014; 588:1100-8. [PMID: 24582655 DOI: 10.1016/j.febslet.2014.02.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 02/07/2014] [Accepted: 02/13/2014] [Indexed: 11/24/2022]
Abstract
Delta-like 1 homolog (Dlk1) is important in myogenesis. However, the roles of different Dlk1 isoforms have not been investigated. In C2C12 cell lines producing different Dlk1 isoforms, membrane-bound Dlk1 promoted the hypertrophic phenotype and a higher fusion rate, whereas soluble Dlk1 inhibited myotube formation. Inversed expression patterns of genes related to myogenic differentiation further support these phenotypic changes. In addition, temporal expression and balance between the Dlk1 isoforms have a regulatory role in myogenesis in vivo. Collectively, Dlk1 isoforms have distinctive effects on myogenesis, and its regulation during myogenesis is critical for normal muscle development.
Collapse
|
38
|
Begum A, Lin Q, Yu C, Kim Y, Yun Z. Interaction of delta-like 1 homolog (Drosophila) with prohibitins and its impact on tumor cell clonogenicity. Mol Cancer Res 2013; 12:155-64. [PMID: 24249679 DOI: 10.1158/1541-7786.mcr-13-0360] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
UNLABELLED Cancer stem cell characteristics, especially their self-renewal and clonogenic potentials, play an essential role in malignant progression and response to anticancer therapies. Currently, it remains largely unknown what pathways are involved in the regulation of cancer cell stemness and differentiation. Previously, we found that delta-like 1 homolog (Drosophila) or DLK1, a developmentally regulated gene, plays a critical role in the regulation of differentiation, self-renewal, and tumorigenic growth of neuroblastoma cells. Here, we show that DLK1 specifically interacts with the prohibitin 1 (PHB1) and PHB2, two closely related genes with pleiotropic functions, including regulation of mitochondrial function and gene transcription. DLK1 interacts with the PHB1-PHB2 complex via its cytoplasmic domain and regulates mitochondrial functions, including mitochondrial membrane potential and production of reactive oxygen species. We have further found that PHB1 and especially PHB2 regulate cancer cell self-renewal as well as their clonogenic potential. Hence, the DLK1-PHB interaction constitutes a new signaling pathway that maintains clonogenicity and self-renewal potential of cancer cells. IMPLICATIONS This study provides a new mechanistic insight into the regulation of the stem cell characteristics of cancer cells.
Collapse
Affiliation(s)
- Asma Begum
- Department of Therapeutic Radiology, Yale University School of Medicine, P.O. Box 208040, New Haven, CT 06520-8040.
| | | | | | | | | |
Collapse
|
39
|
Traustadottir GA, Kosmina R, Sheikh SP, Jensen CH, Andersen DC. Preadipocytes proliferate and differentiate under the guidance of Delta-like 1 homolog (DLK1). Adipocyte 2013; 2:272-5. [PMID: 24052905 PMCID: PMC3774705 DOI: 10.4161/adip.24994] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 05/08/2013] [Accepted: 05/09/2013] [Indexed: 12/20/2022] Open
Abstract
Obesity occurs when an excessive dietary fat intake leads to expansion of adipose tissue, which mainly consists of adipocytes that arise from proliferating and differentiating adipose stem cells, the preadipocytes. Obesity is a consequence of both adipocyte hypertrophy and hyperplasia. Knowledge about preadipocyte differentiation is relatively well established, whereas the mechanism responsible for preadipocyte proliferation is incompletely understood and only in the early stage of comprehension. In this regard, we have recently identified that Delta-like 1 homolog (Dlk1) (also known as Preadipocyte factor 1 [Pref-1]) inhibits preadipocyte proliferation by regulating their entry into G1/S-phase. This novel disclosure, adding to the previous published data on Dlk1 repression of preadipocyte differentiation, has given us the chance to firmly place Dlk1 as a master regulator of preadipocyte homeostasis and adipose tissue expansion. Dlk1 manipulation may, therefore, open new perspectives in obesity treatments.
Collapse
|
40
|
Andersen DC, Laborda J, Baladron V, Kassem M, Sheikh SP, Jensen CH. Dual role of delta-like 1 homolog (DLK1) in skeletal muscle development and adult muscle regeneration. Development 2013; 140:3743-53. [DOI: 10.1242/dev.095810] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Muscle development and regeneration is tightly orchestrated by a specific set of myogenic transcription factors. However, factors that regulate these essential myogenic inducers remain poorly described. Here, we show that delta-like 1 homolog (Dlk1), an imprinted gene best known for its ability to inhibit adipogenesis, is a crucial regulator of the myogenic program in skeletal muscle. Dlk1-/- mice were developmentally retarded in their muscle mass and function owing to inhibition of the myogenic program during embryogenesis. Surprisingly however, Dlk1 depletion improves in vitro and in vivo adult skeletal muscle regeneration by substantial enhancement of the myogenic program and muscle function, possibly by means of an increased number of available myogenic precursor cells. By contrast, Dlk1 fails to alter the adipogenic commitment of muscle-derived progenitors in vitro, as well as intramuscular fat deposition during in vivo regeneration. Collectively, our results suggest a novel and surprising dual biological function of DLK1 as an enhancer of muscle development, but as an inhibitor of adult muscle regeneration.
Collapse
Affiliation(s)
- Ditte Caroline Andersen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Winsloewparken 21 3rd, 5000 Odense C, Denmark
- Insitute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jorge Laborda
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, Avenida de Almansa 14, 02006 Albacete, Spain
| | - Victoriano Baladron
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, Avenida de Almansa 14, 02006 Albacete, Spain
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Odense University Hospital, Odense, Denmark
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Søren Paludan Sheikh
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Winsloewparken 21 3rd, 5000 Odense C, Denmark
- Department of Cardiovascular and Renal Research, Insitute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Charlotte Harken Jensen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Winsloewparken 21 3rd, 5000 Odense C, Denmark
| |
Collapse
|
41
|
Chen J, Crawford R, Chen C, Xiao Y. The key regulatory roles of the PI3K/Akt signaling pathway in the functionalities of mesenchymal stem cells and applications in tissue regeneration. TISSUE ENGINEERING PART B-REVIEWS 2013; 19:516-28. [PMID: 23651329 DOI: 10.1089/ten.teb.2012.0672] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into various cell types and have been widely used in tissue engineering application. In tissue engineering, a scaffold, MSCs and growth factors are used as essential components and their interactions have been regarded to be important for regeneration of tissues. A critical problem for MSCs in tissue engineering is their low survival ability and functionality. Most MSCs are going to be apoptotic after transplantation. Therefore, increasing MSC survival ability and functionalities is the key for potential applications of MSCs. Several approaches have been studied to increase MSC tissue forming capacity including application of growth factors, overexpression of stem cell regulatory genes, and improvement of biomaterials for scaffolds. The effects of these approaches on MSCs have been associated with activation of the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. The pathway plays central regulatory roles in MSC survival, proliferation, migration, angiogenesis, cytokine production, and differentiation. In this review, we summarize and discuss the literatures related to the roles of the PI3K/Akt pathway in the functionalities of MSCs and the involvement of the pathway in biomaterials-increased MSC functionalities. Biomaterials have been modified in their properties and surface structure and loaded with growth factors to increase MSC functionalities. Several studies demonstrated that the biomaterials-increased MSC functionalities are mediated by the activation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Jiezhong Chen
- 1 Institute of Health and Biomedical Innovation, Queensland University of Technology , Brisbane, Australia
| | | | | | | |
Collapse
|
42
|
Potential application of cord blood-derived stromal cells in cellular therapy and regenerative medicine. JOURNAL OF BLOOD TRANSFUSION 2012; 2012:365182. [PMID: 24066257 PMCID: PMC3771124 DOI: 10.1155/2012/365182] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 11/05/2012] [Indexed: 01/08/2023]
Abstract
Neonatal stromal cells from umbilical cord blood (CB) are promising alternatives to bone marrow- (BM-) derived multipotent stromal cells (MSCs). In comparison to BM-MSC, the less mature CB-derived stromal cells have been described as a cell population with higher differentiation and proliferation potential that might be of potential interest for clinical application in regenerative medicine. Recently, it has become clear that cord blood contains different stromal cell populations, and as of today, a clear distinction between unrestricted somatic stromal cells (USSCs) and CB-MSC has been established. This classification is based on the expression of DLK-1, HOX, and CD146, as well as functional examination of the adipogenic differentiation potential and the capacity to support haematopoiesis in vitro and in vivo. However, a marker enabling a prospective isolation of the rare cell populations directly out of cord blood is yet to be found. Further analysis may help to reveal even more subpopulations with different properties, which could be useful for the directed application of these cells in preclinical models.
Collapse
|
43
|
Kang N, Liu X, Guan Y, Wang J, Gong F, Yang X, Yan L, Wang Q, Fu X, Cao Y, Xiao R. Effects of co-culturing BMSCs and auricular chondrocytes on the elastic modulus and hypertrophy of tissue engineered cartilage. Biomaterials 2012; 33:4535-44. [DOI: 10.1016/j.biomaterials.2012.03.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 03/04/2012] [Indexed: 10/28/2022]
|
44
|
Taipaleenmäki H, Harkness L, Chen L, Larsen KH, Säämänen AM, Kassem M, Abdallah BM. The crosstalk between transforming growth factor-β1 and delta like-1 mediates early chondrogenesis during embryonic endochondral ossification. Stem Cells 2012; 30:304-13. [PMID: 22102178 DOI: 10.1002/stem.792] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Delta like-1 (Dlk1)/preadipocyte factor-1 (Pref-1)/fetal antigen-1 (FA1) is a novel surface marker for embryonic chondroprogenitor cells undergoing lineage progression from proliferation to prehypertrophic stages. However, mechanisms mediating control of its expression during chondrogenesis are not known. Thus, we examined the effect of a number of signaling molecules and their inhibitors on Dlk1 expression during in vitro chondrogenic differentiation in mouse embryonic limb bud mesenchymal micromass cultures and mouse embryonic fibroblast (MEF) pellet cultures. Dlk1/Pref-1 was initially expressed during mesenchymal condensation and chondrocyte proliferation, in parallel with expression of Sox9 and Col2a1, and was downregulated upon the expression of Col10a1 by hypertrophic chondrocytes. Among a number of molecules that affected chondrogenesis, transforming growth factor-β1 (TGF-β1)-induced proliferation of chondroprogenitors was associated with decreased Dlk1 expression. This effect was abolished by TGF-β signaling inhibitor SB431542, suggesting regulation of Dlk1/FA1 by TGF-β1 signaling in chondrogenesis. TGF-β1-induced Smad phosphorylation and chondrogenesis were significantly increased in Dlk1(-/-) MEF, while they were blocked in Dlk1 overexpressing MEF, in comparison with wild-type MEF. Furthermore, overexpression of Dlk1 or addition of its secreted form FA1 dramatically inhibited TGF-β1-induced Smad reporter activity. In conclusion, our data identified Dlk1/FA1 as a downstream target of TGF-β1 signaling molecule that mediates its function in embryonic chondrogenesis. The crosstalk between TGF-β1 and Dlk1/FA1 was shown to promote early chondrogenesis during the embryonic endochondral ossification process.
Collapse
Affiliation(s)
- Hanna Taipaleenmäki
- Endocrine Research Laboratory (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital, Odense, Denmark
| | | | | | | | | | | | | |
Collapse
|
45
|
Rogers ED, Ramalie JR, McMurray EN, Schmidt JV. Localizing transcriptional regulatory elements at the mouse Dlk1 locus. PLoS One 2012; 7:e36483. [PMID: 22606264 PMCID: PMC3350532 DOI: 10.1371/journal.pone.0036483] [Citation(s) in RCA: 13] [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: 11/01/2011] [Accepted: 04/06/2012] [Indexed: 11/25/2022] Open
Abstract
Much effort has focused recently on determining the mechanisms that control the allele-specific expression of genes subject to genomic imprinting, yet imprinting regulation is only one aspect of configuring appropriate expression of these genes. Imprinting control mechanisms must interact with those regulating the tissue-specific expression pattern of each imprinted gene in a cluster. Proper expression of the imprinted Delta-like 1 (Dlk1)-Maternally expressed gene 3 (Meg3) gene pair is required for normal fetal development in mammals, yet the mechanisms that control tissue-specific expression of these genes are unknown. We have used a combination of in vivo and in vitro expression assays to localize cis-regulatory elements that may regulate Dlk1 expression in the mouse embryo. A bacterial artificial chromosome transgene encompassing the Dlk1 gene and 77 kb of flanking sequence conferred expression in most endogenous Dlk1-expressing tissues. In combination with previous transgenic data, these experiments localize the majority of Dlk1 cis-regulatory elements to a 41 kb region upstream of the gene. Cross-species sequence conservation was used to further define potential regulatory elements, several of which functioned as enhancers in a luciferase expression assay. Two of these elements were able to drive expression of a lacZ reporter transgene in Dlk1-expressing tissues in the mouse embryo. The sequence proximal to Dlk1 therefore contains at least two discrete regions that may regulate tissue-specificity of Dlk1 expression.
Collapse
MESH Headings
- Animals
- Base Sequence
- Calcium-Binding Proteins
- Chromosomes, Artificial, Bacterial/genetics
- Conserved Sequence
- DNA Primers/genetics
- Enhancer Elements, Genetic
- Female
- Gene Expression Regulation, Developmental
- Genomic Imprinting
- Intercellular Signaling Peptides and Proteins/genetics
- Lac Operon
- Mice
- Mice, Transgenic
- Muscle, Skeletal/embryology
- Muscle, Skeletal/metabolism
- RNA, Long Noncoding
- RNA, Untranslated/genetics
- Regulatory Elements, Transcriptional
- Tissue Distribution
Collapse
Affiliation(s)
- Eric D. Rogers
- The Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jenniffer R. Ramalie
- The Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Erin N. McMurray
- The Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jennifer V. Schmidt
- The Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
46
|
Mitterberger MC, Lechner S, Mattesich M, Kaiser A, Probst D, Wenger N, Pierer G, Zwerschke W. DLK1(PREF1) is a negative regulator of adipogenesis in CD105⁺/CD90⁺/CD34⁺/CD31⁻/FABP4⁻ adipose-derived stromal cells from subcutaneous abdominal fat pats of adult women. Stem Cell Res 2012; 9:35-48. [PMID: 22640926 DOI: 10.1016/j.scr.2012.04.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 04/04/2012] [Accepted: 04/06/2012] [Indexed: 12/20/2022] Open
Abstract
The main physiological function of adipose-derived stromal/progenitor cells (ASC) is to differentiate into adipocytes. ASC are most likely localized at perivascular sites in adipose tissues and retain the capacity to differentiate into multiple cell types. Although cell surface markers for ASC have been described, there is no complete consensus on the antigen expression pattern that will precisely define these cells. DLK1(PREF1) is an established marker for mouse adipocyte progenitors which inhibits adipogenesis. This suggests that DLK1(PREF1) could be a useful marker to characterize human ASC. The DLK1(PREF1) status of human ASC is however unknown. In the present study we isolated ASC from the heterogeneous stromal vascular fraction of subcutaneous abdominal fat pats of adult women. These cells were selected by their plastic adherence and expanded to passage 5. The ASC were characterized as relatively homogenous cell population with the capacity to differentiate in vitro into adipocytes, chondrocytes, and osteoblasts and the immunophenotype CD105⁺/CD90⁺/CD34⁺/CD31⁻/FABP4⁻. The ASC were positive for DLK1(PREF1) which was well expressed in proliferating and density arrested cells but downregulated in the course of adipogenic differentiation. To investigate whether DLK1(PREF1) plays a role in the regulation of adipogenesis in these cells RNAi-mediated knockdown experiments were conducted. Knockdown of DLK1(PREF1) in differentiating ASC resulted in a significant increase of the expression of the adipogenic key regulator PPARγ2 and of the terminal adipogenic differentiation marker FABP4. We conclude that DLK1(PREF1) is well expressed in human ASC and acts as a negative regulator of adipogenesis. Moreover, DLK1(PREF1) could be a functional marker contributing to the characterization of human ASC.
Collapse
Affiliation(s)
- Maria C Mitterberger
- Department of Cell Metabolism and Differentiation Research, Institute for Biomedical Aging Research of the Austrian Academy of Sciences, Rennweg 10, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Begum A, Kim Y, Lin Q, Yun Z. DLK1, delta-like 1 homolog (Drosophila), regulates tumor cell differentiation in vivo. Cancer Lett 2011; 318:26-33. [PMID: 22142700 DOI: 10.1016/j.canlet.2011.11.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 11/16/2011] [Accepted: 11/25/2011] [Indexed: 02/08/2023]
Abstract
The stem cell-like characteristics of tumor cells are not only essential for tumor development and malignant progression, but also significantly contribute to therapy resistance. However, it remains poorly understood how cancer cell differentiation or stemness is regulated in vivo. We investigated the role of the stem cell gene DLK1, or delta-like 1 homolog (Drosophila), in the regulation of cancer cell differentiation in vivo using neuroblastoma (NB) xenografts as a model. We found that loss-of-function mutants of DLK1 significantly enhanced NB cell differentiation in vivo likely by increasing the basal phosphorylation of MEK and ERK kinases, a mechanism that has been shown to facilitate neuronal differentiation. We also found that DLK1(+) cells are preferentially located in hypoxic regions. These results clearly demonstrate that DLK1 plays an important role in the maintenance of undifferentiated, stem cell-like phenotypes of NB cells in vivo.
Collapse
Affiliation(s)
- Asma Begum
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|
48
|
Rodríguez P, Higueras MA, González-Rajal A, Alfranca A, Fierro-Fernández M, García-Fernández RA, Ruiz-Hidalgo MJ, Monsalve M, Rodríguez-Pascual F, Redondo JM, de la Pompa JL, Laborda J, Lamas S. The non-canonical NOTCH ligand DLK1 exhibits a novel vascular role as a strong inhibitor of angiogenesis. Cardiovasc Res 2011; 93:232-41. [PMID: 22068159 DOI: 10.1093/cvr/cvr296] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS The epidermal growth factor-like protein Delta-like 1 (DLK1) regulates multiple differentiation processes. It resembles NOTCH ligands structurally and is considered a non-canonical ligand. Given the crucial role of the NOTCH pathway in angiogenesis, we hypothesized that DLK1 could regulate angiogenesis by interfering with NOTCH. We therefore investigated the expression and function of DLK1 in the vascular endothelium and its role in the regulation of angiogenesis. METHODS AND RESULTS We report DLK1 expression in the endothelium of different species, including human, cow, pig, and mouse. Angiogenesis was studied by using in vitro and in vivo models of angiotube formation in endothelial cells, retinal phenotypes in Dlk1-null mice, and vessel development in zebrafish. DLK1 overexpression strongly inhibited angiotube formation, whereas lung endothelial cells from Dlk1-null mice were highly angiogenic. In vivo studies demonstrated DLK1-mediated inhibition of neovessel formation and revealed an altered pattern of angiogenesis in the retinas of Dlk1-null mice. The expression of human DLK1 in zebrafish embryos severely altered the formation of intersegmental vessels, while knockdown of the orthologous gene was associated with ectopic and increased tumour-induced angiogenesis. NOTCH-dependent signalling as determined by gene expression reporters was inhibited by the presence of DLK1 in vascular endothelial cells. In contrast, Dlk1-null mice showed increased levels of NOTCH downstream targets, such as Snail and Slug. CONCLUSION Our results unveil a novel inhibitory role for DLK1 in the regulation of angiogenesis, mediated by antagonism of the NOTCH pathway, and establish the basis for investigating its action in pathological settings.
Collapse
Affiliation(s)
- Patricia Rodríguez
- Laboratorio Mixto Consejo Superior de Investigaciones Científicas-Fundación Renal Iñigo Alvarez de Toledo, Centro de Biología Molecular Severo Ochoa, Nicolás Cabrera 1, 28029 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|