1
|
Rodríguez-Cano MM, González-Gómez MJ, Monsalve EM, Díaz-Guerra MJM, Kassem M, Laborda J, Nueda ML, Baladrón V. DLK1 and DLK2, two non-canonical ligands of NOTCH receptors, differentially modulate the osteogenic differentiation of mesenchymal C3H10T1/2 cells. Biol Res 2024; 57:77. [PMID: 39473022 PMCID: PMC11523663 DOI: 10.1186/s40659-024-00561-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND C3H10T1/2 is a mesenchymal cell line capable of differentiating into osteoblasts, adipocytes and chondrocytes. The differentiation of these cells into osteoblasts is modulated by various transcription factors, such as RUNX2. Additionally, several interconnected signaling pathways, including the NOTCH pathway, play a crucial role in modulating their differentiation into mature bone cells. We have investigated the roles of DLK1 and DLK2, two non-canonical inhibitory ligands of NOTCH receptors, in the osteogenic differentiation of C3H10T1/2 cells. RESULTS Our results corroborate existing evidence that DLK1 acts as an inhibitor of osteogenesis. In contrast, we demonstrate for the first time that DLK2 enhances this differentiation process. Additionally, our data suggest that NOTCH2, 3 and 4 receptors may promote osteogenesis, as indicated by their increased expression during this process, whereas NOTCH1 expression, which decreases during cell differentiation, might inhibit osteogenesis. Moreover, treatment with DAPT, a NOTCH signaling inhibitor, impeded osteogenic differentiation. We have confirmed the increase in ERK1/2 MAPK and p38 MAPK phosphorylation in C3H10T1/2 cells induced to differentiate to osteoblasts. Our new findings reveal increased ERK1/2 MAPK phosphorylation in differentiated C3H10T1/2 cells with a decrease in DLK1 expression or an overexpression of DLK2, which is coincident with the behavior of those transfectants where we have detected an increase in osteogenic differentiation. Additionally, p38 MAPK phosphorylation increases in differentiated C3H10T1/2 cells with reduced DLK1 levels. CONCLUSIONS Our results suggest that DLK1 may inhibit osteogenesis, while DLK2 may promote it, by modulating NOTCH signaling and the phosphorylation of ERK1/2 and p38 MAPK pathways. Given the established inhibitory effect of DLK proteins on NOTCH signaling, these new insights could pave the way for developing future therapeutic strategies aimed at treating bone diseases.
Collapse
Affiliation(s)
- María-Milagros Rodríguez-Cano
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Pharmacy School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - María-Julia González-Gómez
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, ETSIAMB/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - Eva-María Monsalve
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Medical School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - María-José M Díaz-Guerra
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Medical School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - Moustapha Kassem
- Molecular Endocrinology Unit (KMEB), Department of Endocrinology and Metabolism, University Hospital of Odense and Danish Institute for Advanced Study, University of Southern Denmark, Odense, Denmark
| | - Jorge Laborda
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Pharmacy School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - María-Luisa Nueda
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Pharmacy School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain.
| | - Victoriano Baladrón
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Medical School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain.
| |
Collapse
|
2
|
Tarapongpun T, Onlamoon N, Tabu K, Chuthapisith S, Taga T. The optimized priming effect of FGF-1 and FGF-2 enhances preadipocyte lineage commitment in human adipose-derived mesenchymal stem cells. Genes Cells 2024; 29:231-253. [PMID: 38253356 DOI: 10.1111/gtc.13095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/26/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024]
Abstract
The cell-assisted lipotransfer technique, integrating adipose-derived mesenchymal stem cells (ADMSCs), has transformed lipofilling, enhancing fat graft viability. However, the multipotent nature of ADMSCs poses challenges. To improve safety and graft vitality and to reduce unwanted lineage differentiation, this study refines the methodology by priming ADMSCs into preadipocytes-unipotent, self-renewing cells. We explored the impact of fibroblast growth factor-1 (FGF-1), fibroblast growth factor-2 (FGF-2), and epidermal growth factor (EGF), either alone or in combination, on primary human ADMSCs during the proliferative phase. FGF-2 emerged as a robust stimulator of cell proliferation, preserving stemness markers, especially when combined with EGF. Conversely, FGF-1, while not significantly affecting cell growth, influenced cell morphology, transitioning cells to a rounded shape with reduced CD34 expression. Furthermore, co-priming with FGF-1 and FGF-2 enhanced adipogenic potential, limiting osteogenic and chondrogenic tendencies, and possibly promoting preadipocyte commitment. These preadipocytes exhibited unique features: rounded morphology, reduced CD34, decreased preadipocyte factor 1 (Pref-1), and elevated C/EBPα and PPARγ, alongside sustained stemness markers (CD73, CD90, CD105). Mechanistically, FGF-1 and FGF-2 activated key adipogenic transcription factors-C/EBPα and PPARγ-while inhibiting GATA3 and Notch3, which are adipogenesis inhibitors. These findings hold the potential to advance innovative strategies for ADMSC-mediated lipofilling procedures.
Collapse
Affiliation(s)
- Tanakorn Tarapongpun
- Division of Head Neck and Breast Surgery, Faculty of Medicine Siriraj Hospital, Department of Surgery, Mahidol University, Bangkok, Thailand
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Nattawat Onlamoon
- Department of Research, Faculty of Medicine Siriraj Hospital, Siriraj Research Group in Immunobiology and Therapeutic Sciences, Mahidol University, Bangkok, Thailand
| | - Kouichi Tabu
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Suebwong Chuthapisith
- Division of Head Neck and Breast Surgery, Faculty of Medicine Siriraj Hospital, Department of Surgery, Mahidol University, Bangkok, Thailand
| | - Tetsuya Taga
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
3
|
Keleher MR, Shubhangi S, Brown A, Duensing AM, Lixandrão ME, Gavin KM, Smith HA, Kechris KJ, Yang IV, Dabelea D, Boyle KE. Adipocyte hypertrophy in mesenchymal stem cells from infants of mothers with obesity. Obesity (Silver Spring) 2023; 31:2090-2102. [PMID: 37475691 PMCID: PMC10372711 DOI: 10.1002/oby.23803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/17/2023] [Accepted: 04/26/2023] [Indexed: 07/22/2023]
Abstract
OBJECTIVE Fat content of adipocytes derived from infant umbilical cord mesenchymal stem cells (MSCs) predicts adiposity in children through 4 to 6 years of age. This study tested the hypothesis that MSCs from infants born to mothers with obesity (Ob-MSCs) exhibit adipocyte hypertrophy and perturbations in genes regulating adipogenesis compared with MSCs from infants of mothers with normal weight (NW-MSCs). METHODS Adipogenesis was induced in MSCs embedded in three-dimensional hydrogel structures, and cell size and number were measured by three-dimensional imaging. Proliferation and protein markers of proliferation and adipogenesis in undifferentiated and adipocyte differentiating cells were measured. RNA sequencing was performed to determine pathways linked to adipogenesis phenotype. RESULTS In undifferentiated MSCs, greater zinc finger protein (Zfp)423 protein content was observed in Ob- versus NW-MSCs. Adipocytes from Ob-MSCs were larger but fewer than adipocytes from NW-MSCs. RNA sequencing analysis showed that Zfp423 protein correlated with mRNA expression of genes enriched for cell cycle, MSC lineage specification, inflammation, and metabolism pathways. MSC proliferation was not different before differentiation but declined faster in Ob-MSCs upon adipogenic induction. CONCLUSIONS Ob-MSCs have an intrinsic propensity for adipocyte hypertrophy and reduced hyperplasia during adipogenesis in vitro, perhaps linked to greater Zfp423 content and changes in cell cycle pathway gene expression.
Collapse
Affiliation(s)
- Madeline Rose Keleher
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO USA
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO USA
| | - Shreya Shubhangi
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Asya Brown
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Allison M. Duensing
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Manoel E. Lixandrão
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Kathleen M. Gavin
- Division of Geriatric Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
- Eastern Colorado VA Geriatric, Research, Education, and Clinical Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Harry A. Smith
- Department of Biostatistics & Informatics, Colorado School of Public Health, Aurora, CO USA
| | - Katerina J. Kechris
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO USA
- Department of Biostatistics & Informatics, Colorado School of Public Health, Aurora, CO USA
| | - Ivana V. Yang
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO USA
| | - Dana Dabelea
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO USA
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Kristen E. Boyle
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO USA
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO USA
| |
Collapse
|
4
|
Zhang H, Yang Y, Li X, Yuan X, Chu Q. Targeting the Notch signaling pathway and the Notch ligand, DLL3, in small cell lung cancer. Biomed Pharmacother 2023; 159:114248. [PMID: 36645960 DOI: 10.1016/j.biopha.2023.114248] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
Small cell lung cancer (SCLC) is a highly aggressive and poorly differentiated cancer with high-grade neuroendocrine (NE) features, accounting for approximately 15 % of all lung cancers. For decades, chemotherapy and radiotherapy have predominated the treatment strategy for SCLC, but relapses ensue quickly and result in poor survival of patients. Immunotherapy has brought novel insights, yet the efficacy is still restricted to a limited population with SCLC. Notch signaling is identified to play a key role in the initiation and development of SCLC, and the Notch ligand, Delta-like ligand 3 (DLL3) is found broadly and specifically expressed in SCLC cells. Thus, Notch signaling is under active exploration as a potential therapeutic target in SCLC. Herein, we summarized and updated the functional relevance of Notch signaling in SCLC, discussed Notch signaling-targeted therapy for SCLC and the correspondent preclinical and clinical trials, and investigated the promising synergy effects of Notch signaling targeted therapy and immune checkpoint inhibitors (ICIs) treatment.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China.
| | - Yunkai Yang
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China.
| | - Xuchang Li
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China.
| | - Xun Yuan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China.
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China.
| |
Collapse
|
5
|
Vitamin A: A Key Inhibitor of Adipocyte Differentiation. PPAR Res 2023; 2023:7405954. [PMID: 36776154 PMCID: PMC9908342 DOI: 10.1155/2023/7405954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 02/04/2023] Open
Abstract
Inhibiting adipocyte differentiation, the conversion of preadipocytes to mature functional adipocytes, might represent a new approach to treating obesity and related metabolic disorders. Peroxisome proliferator-activated receptor γ and CCAAT-enhancer-binding protein α are two master coregulators controlling adipogenesis both in culture and in vivo. Many recent studies have confirmed the relationship between retinoic acid (RA) and the conversion of embryonic stem cells into adipocytes; however, these studies have shown that RA potently blocks the differentiation of preadipocytes into mature adipocytes. Nevertheless, the functional role of RA in early tissue development and stem cell differentiation, including in adipose tissue, remains unclear. This study highlights transcription factors that block adipocyte differentiation and maintain preadipocyte status, focusing on those controlled by RA. However, some of these novel adipogenesis inhibitors have not been validated in vivo, and their mechanisms of action require further clarification.
Collapse
|
6
|
Huang D, Han Y, Tang T, Yang L, Jiang P, Qian W, Zhang Z, Qian X, Zeng X, Qian P. Dlk1 maintains adult mice long-term HSCs by activating Notch signaling to restrict mitochondrial metabolism. Exp Hematol Oncol 2023; 12:11. [PMID: 36653853 PMCID: PMC9850540 DOI: 10.1186/s40164-022-00369-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Adult hematopoietic stem cells (HSCs) homeostasis is critically important in maintaining lifelong hematopoiesis. However, how adult HSCs orchestrate its homeostasis remains not fully understood. Imprinted gene Dlk1 has been shown to play critical role in mouse embryonic hematopoiesis and in regulation of stem cells, but its physiological roles in adult HSCs are unknown. METHODS We performed gene expression analysis of Dlk1, and constructed conditional Dlk1 knockout (KO) mice by crossing Mx1 cre mice with Dlkflox/flox mice. Western blot and quantitative PCR were used to detect Dlk1 KO efficiency. Flow cytometry was performed to investigate the effects of Dlk1 KO on HSCs, progenitors and linage cells in primary mice. Competitive HSCs transplantation and secondary transplantation was used to examine the effects of Dlk1 KO on long-term hematopoietic repopulation potential of HSCs. RNA-Seq and cell metabolism assays was used to determine the underlying mechanisms. RESULTS Dlk1 was highly expressed in adult mice long-term HSCs (LT-HSCs) relative to progenitors and mature lineage cells. Dlk1 KO in adult mice HSCs drove HSCs enter active cell cycle, and expanded phenotypical LT-HSCs, but undermined its long-term hematopoietic repopulation potential. Dlk1 KO resulted in an increase in HSCs' metabolic activity, including glucose uptake, ribosomal translation, mitochondrial metabolism and ROS production, which impaired HSCs function. Further, Dlk1 KO in adult mice HSCs attenuated Notch signaling, and re-activation of Notch signaling under Dlk1 KO decreased the mitochondrial activity and ROS production, and rescued the changes in frequency and absolute number of HSCs. Scavenging ROS by antioxidant N-acetylcysteine could inhibit mitochondrial metabolic activity, and rescue the changes in HSCs caused by Dlk1 KO. CONCLUSION Our study showed that Dlk1 played an essential role in maintaining HSC homeostasis, which is realized by governing cell cycle and restricting mitochondrial metabolic activity.
Collapse
Affiliation(s)
- Deyu Huang
- grid.13402.340000 0004 1759 700XCenter of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China ,grid.13402.340000 0004 1759 700XInstitute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XDr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310012 Zhejiang People’s Republic of China
| | - Yingli Han
- grid.13402.340000 0004 1759 700XCenter of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China ,grid.13402.340000 0004 1759 700XInstitute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058 China
| | - Tian Tang
- grid.13402.340000 0004 1759 700XCenter of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China ,grid.13402.340000 0004 1759 700XInstitute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058 China ,grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR China
| | - Lin Yang
- grid.13402.340000 0004 1759 700XCenter of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China ,grid.13402.340000 0004 1759 700XInstitute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058 China
| | - Penglei Jiang
- grid.13402.340000 0004 1759 700XCenter of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China ,grid.13402.340000 0004 1759 700XInstitute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058 China
| | - Wenchang Qian
- grid.13402.340000 0004 1759 700XCenter of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China ,grid.13402.340000 0004 1759 700XInstitute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058 China
| | - Zhaoru Zhang
- grid.13402.340000 0004 1759 700XCenter of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China ,grid.13402.340000 0004 1759 700XInstitute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058 China
| | - Xinyue Qian
- grid.13402.340000 0004 1759 700XCenter of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China ,grid.13402.340000 0004 1759 700XInstitute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058 China
| | - Xin Zeng
- grid.13402.340000 0004 1759 700XCenter of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China ,grid.13402.340000 0004 1759 700XInstitute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058 China
| | - Pengxu Qian
- grid.13402.340000 0004 1759 700XCenter of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China ,grid.13402.340000 0004 1759 700XInstitute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058 China ,grid.13402.340000 0004 1759 700XDr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310012 Zhejiang People’s Republic of China
| |
Collapse
|
7
|
Gui Z, Lv M, Han M, Li S, Mo Z. Effect of CPP-related genes on GnRH secretion and Notch signaling pathway during puberty. Biomed J 2022; 46:100575. [PMID: 36528337 DOI: 10.1016/j.bj.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/11/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Puberty is a complex biological process of sexual development, influenced by genetic, metabolic-nutritional, environmental and socioeconomic factors, characterized by the development of secondary sexual characteristics, maturation of the gonads, leading to the acquisition of reproductive capacity. The onset of central precocious puberty (CPP) is mainly associated with the early activation of the hypothalamic-pituitary-gonadal (HPG) axis and increased secretion of gonadotropin-releasing hormone (GnRH), leading to increased pituitary secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) and activation of gonadal function. Due to the expense and invasiveness of current diagnostic testing and drug therapies for CPP, it would be helpful to find serum and genetic markers to facilitate diagnosis. In this paper, we summarized the related factors that may affect the expression of GnRH1 gene and the secretion and action pathway of GnRH and related sex hormones, and found several potential targets, such as MKRN3, DLK1 and KISS1. Although, the specific mechanism still needs to be further studied, we would be encouraged if the insights from this review could provide new insights for future research and clinical diagnosis and treatment of CPP.
Collapse
Affiliation(s)
- Zihao Gui
- Guangxi Provincial Postgraduate Co-training Base for Collaborative Innovation in Basic Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China; Clinical Medicine of Hengyang Medical School, University of South China, Hengyang, China
| | - Mei Lv
- Guangxi Provincial Postgraduate Co-training Base for Collaborative Innovation in Basic Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China; Anshun City People's Hospital, Anshun, Guizhou, China
| | - Min Han
- Clinical Medicine of Hengyang Medical School, University of South China, Hengyang, China
| | - Shan Li
- Guangxi Provincial Postgraduate Co-training Base for Collaborative Innovation in Basic Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China
| | - Zhongcheng Mo
- Guangxi Provincial Postgraduate Co-training Base for Collaborative Innovation in Basic Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China; Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China.
| |
Collapse
|
8
|
Kapeni C, Nitsche L, Kilpatrick AM, Wilson NK, Xia K, Mirshekar-Syahkal B, Chandrakanthan V, Malouf C, Pimanda JE, Göttgens B, Kirschner K, Tomlinson SR, Ottersbach K. p57Kip2 regulates embryonic blood stem cells by controlling sympathoadrenal progenitor expansion. Blood 2022; 140:464-477. [PMID: 35653588 PMCID: PMC9353151 DOI: 10.1182/blood.2021014853] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/13/2022] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are of major clinical importance, and finding methods for their in vitro generation is a prime research focus. We show here that the cell cycle inhibitor p57Kip2/Cdkn1c limits the number of emerging HSCs by restricting the size of the sympathetic nervous system (SNS) and the amount of HSC-supportive catecholamines secreted by these cells. This regulation occurs at the SNS progenitor level and is in contrast to the cell-intrinsic function of p57Kip2 in maintaining adult HSCs, highlighting profound differences in cell cycle requirements of adult HSCs compared with their embryonic counterparts. Furthermore, this effect is specific to the aorta-gonad-mesonephros (AGM) region and shows that the AGM is the main contributor to early fetal liver colonization, as early fetal liver HSC numbers are equally affected. Using a range of antagonists in vivo, we show a requirement for intact β2-adrenergic signaling for SNS-dependent HSC expansion. To gain further molecular insights, we have generated a single-cell RNA-sequencing data set of all Ngfr+ sympathoadrenal cells around the dorsal aorta to dissect their differentiation pathway. Importantly, this not only defined the relevant p57Kip2-expressing SNS progenitor stage but also revealed that some neural crest cells, upon arrival at the aorta, are able to take an alternative differentiation pathway, giving rise to a subset of ventrally restricted mesenchymal cells that express important HSC-supportive factors. Neural crest cells thus appear to contribute to the AGM HSC niche via 2 different mechanisms: SNS-mediated catecholamine secretion and HSC-supportive mesenchymal cell production.
Collapse
Affiliation(s)
- Chrysa Kapeni
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Leslie Nitsche
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicola K Wilson
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kankan Xia
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Bahar Mirshekar-Syahkal
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Vashe Chandrakanthan
- School of Medical Sciences, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Camille Malouf
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - John E Pimanda
- School of Medical Sciences, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
- Department of Haematology, The Prince of Wales Hospital, Sydney, NSW, Australia
| | - Berthold Göttgens
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kristina Kirschner
- Institute of Cancer Sciences and
- CRUK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom
| | - Simon R Tomlinson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Katrin Ottersbach
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
9
|
Fu Y, Hao X, Shang P, Chamba Y, Zhang B, Zhang H. Functional Identification of Porcine DLK1 during Muscle Development. Animals (Basel) 2022; 12:ani12121523. [PMID: 35739860 PMCID: PMC9219491 DOI: 10.3390/ani12121523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Skeletal muscle is the largest tissue and serves as a protein reservoir and energy reservoir in the human and animal body. It also serves as the main metabolic activity site. The formation of skeletal muscle mainly depends on the differentiation and fusion of myocytes and other complex ordered processes; each step is regulated by various factors. In this study, we investigated the expression profiles, functional identification, and regulatory pathways of Delta-like 1 homolog (DLK1) in pigs and myocytes. We found that DLK1 was highly expressed in the muscle tissues of pigs. DLK1 promoted myocyte proliferation, migration, differentiation, fusion, and muscular hypertrophy, but suppressed muscle degradation. DLK1 also inhibited the Notch signaling pathway by regulating the expression of key factors in the pathway, thereby producing a phenotype in which DLK1 promotes muscle development. These findings provide valuable information to improve our understanding of the functional mechanisms of DLK1 that underly myogenesis to accelerate the process of animal genetic improvement. Abstract DLK1 is paternally expressed and is involved in metabolism switching, stem cell maintenance, cell proliferation, and differentiation. Porcine DLK1 was identified in our previous study as a candidate gene that regulates muscle development. In the present study, we characterized DLK1 expression in pigs, and the results showed that DLK1 was highly expressed in the muscles of pigs. In-vitro cellular tests showed that DLK1 promoted myoblast proliferation, migration, and muscular hypertrophy, and at the same time inhibited muscle degradation. The expression of myogenic and fusion markers and the formation of multinucleated myotubes were both upregulated in myoblasts with DLK1 overexpression. DLK1 levels in cultured myocytes were negatively correlated with the expression of key factors in the Notch pathway, suggesting that the suppression of Notch signaling pathways may mediate these processes. Collectively, our results suggest a biological function of DLK1 as an enhancer of muscle development by the inhibition of Notch pathways.
Collapse
Affiliation(s)
- Yu Fu
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.F.); (X.H.)
| | - Xin Hao
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.F.); (X.H.)
| | - Peng Shang
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi 860000, China; (P.S.); (Y.C.)
| | - Yangzom Chamba
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi 860000, China; (P.S.); (Y.C.)
| | - Bo Zhang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.F.); (X.H.)
- Correspondence: (B.Z.); (H.Z.); Tel.: +86-010-62734852 (H.Z.)
| | - Hao Zhang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.F.); (X.H.)
- Correspondence: (B.Z.); (H.Z.); Tel.: +86-010-62734852 (H.Z.)
| |
Collapse
|
10
|
Lee MG, Lee YK, Huang SC, Chang CL, Ko CY, Lee WC, Chen TY, Tzou SJ, Huang CY, Tai MH, Lin YW, Kung ML, Tsai MC, Chen YL, Chang YC, Wen ZH, Huang CC, Chu TH. DLK2 Acts as a Potential Prognostic Biomarker for Clear Cell Renal Cell Carcinoma Based on Bioinformatics Analysis. Genes (Basel) 2022; 13:genes13040629. [PMID: 35456435 PMCID: PMC9030291 DOI: 10.3390/genes13040629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 02/07/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common RCC subtype with a high mortality. It has been reported that delta-like 1 homologue (DLK1) participates in the tumor microenvironmental remodeling of ccRCC, but the relationship between delta-like 2 homologue (DLK2, a DLK1 homologue) and ccRCC is still unclear. Thus, this study aims to investigate the role of DLK2 in the biological function and disease prognosis of ccRCC using bioinformatics analysis. The TNMplot database showed that DLK2 was upregulated in ccRCC tissues. From the UALCAN analysis, the overexpression of DLK2 was associated with advanced stage and high grade in ccRCC. Moreover, the Kaplan-Meier plotter (KM Plotter) database showed that DLK2 upregulation was associated with poor survival outcome in ccRCC. By the LinkedOmics analysis, DLK2 signaling may participated in the modulation of ccRCC extracellular matrix (ECM), cell metabolism, ribosome biogenesis, TGF-β signaling and Notch pathway. Besides, Tumor Immune Estimation Resource (TIMER) analysis showed that the macrophage and CD8+ T cell infiltrations were associated with good prognosis in ccRCC patients. Finally, DLK2 overexpression was associated with the reduced macrophage recruitments and the M1–M2 polarization of macrophage in ccRCC tissues. Together, DLK2 may acts as a novel biomarker, even therapeutic target in ccRCC. However, this study lacks experimental validation, and further studies are required to support this viewpoint.
Collapse
Affiliation(s)
- Man-Gang Lee
- Department of Surgery, Division of Urology, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- Department of Surgery, Division of Urology, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 81342, Taiwan
| | - Yung-Kuo Lee
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
| | - Shih-Chung Huang
- Department of Internal Medicine, Division of Cardiology, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- Department of Internal Medicine, Division of Cardiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-L.C.); (C.-Y.K.); (S.-J.T.)
| | - Chen-Lin Chang
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-L.C.); (C.-Y.K.); (S.-J.T.)
- Department of Psychiatry, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Chou-Yuan Ko
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-L.C.); (C.-Y.K.); (S.-J.T.)
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
| | - Wen-Chin Lee
- Department of Internal Medicine, Division of Nephrology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Tung-Yuan Chen
- Department of Surgery, Division of Colorectal Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
| | - Shiow-Jyu Tzou
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-L.C.); (C.-Y.K.); (S.-J.T.)
- Department of Nursing, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Cheng-Yi Huang
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-Y.H.); (M.-H.T.)
- Department of Pathology, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Ming-Hong Tai
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-Y.H.); (M.-H.T.)
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Yu-Wei Lin
- Department of Radiation Oncology, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan;
| | - Mei-Lang Kung
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan;
| | - Ming-Chao Tsai
- Department of Internal Medicine, Division of Hepato-Gastroenterology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Yung-Lung Chen
- Section of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Yi-Chen Chang
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan;
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, Asia-Pacific Ocean Research Center, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
| | - Chao-Cheng Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Biobank and Tissue Bank, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Correspondence: (C.-C.H.); (T.-H.C.); Tel.: +886-7-731-7123 (ext. 2557) (C.-C.H.); +886-7-749-6751 (ext. 726201) (T.-H.C.)
| | - Tian-Huei Chu
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- Correspondence: (C.-C.H.); (T.-H.C.); Tel.: +886-7-731-7123 (ext. 2557) (C.-C.H.); +886-7-749-6751 (ext. 726201) (T.-H.C.)
| |
Collapse
|
11
|
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: 1.0] [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
|
12
|
Abstract
DLK1 is a maternally imprinted, paternally expressed gene coding for the transmembrane protein Delta-like homologue 1 (DLK1), a non-canonical NOTCH ligand with well-described roles during development, and tumor-supportive functions in several aggressive cancer forms. Here, we review the many functions of DLK1 as a regulator of stem cell pools and tissue differentiation in tissues such as brain, muscle, and liver. Furthermore, we review recent evidence supporting roles for DLK1 in the maintenance of aggressive stem cell characteristics of tumor cells, specifically focusing on central nervous system tumors, neuroblastoma, and hepatocellular carcinoma. We discuss NOTCH -dependent as well as NOTCH-independent functions of DLK1, and focus particularly on the complex pattern of DLK1 expression and cleavage that is finely regulated from a spatial and temporal perspective. Progress in recent years suggest differential functions of extracellular, soluble DLK1 as a paracrine stem cell niche-secreted factor, and has revealed a role for the intracellular domain of DLK1 in cell signaling and tumor stemness. A better understanding of DLK1 regulation and signaling may enable therapeutic targeting of cancer stemness by interfering with DLK1 release and/or intracellular signaling.
Collapse
Affiliation(s)
- Elisa Stellaria Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alexander Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
13
|
Weickert MT, Hecker JS, Buck MC, Schreck C, Rivière J, Schiemann M, Schallmoser K, Bassermann F, Strunk D, Oostendorp RAJ, Götze KS. Bone marrow stromal cells from MDS and AML patients show increased adipogenic potential with reduced Delta-like-1 expression. Sci Rep 2021; 11:5944. [PMID: 33723276 PMCID: PMC7961144 DOI: 10.1038/s41598-021-85122-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 02/24/2021] [Indexed: 12/29/2022] Open
Abstract
Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are clonal hematopoietic stem cell disorders with a poor prognosis, especially for elderly patients. Increasing evidence suggests that alterations in the non-hematopoietic microenvironment (bone marrow niche) can contribute to or initiate malignant transformation and promote disease progression. One of the key components of the bone marrow (BM) niche are BM stromal cells (BMSC) that give rise to osteoblasts and adipocytes. It has been shown that the balance between these two cell types plays an important role in the regulation of hematopoiesis. However, data on the number of BMSC and the regulation of their differentiation balance in the context of hematopoietic malignancies is scarce. We established a stringent flow cytometric protocol for the prospective isolation of a CD73+ CD105+ CD271+ BMSC subpopulation from uncultivated cryopreserved BM of MDS and AML patients as well as age-matched healthy donors. BMSC from MDS and AML patients showed a strongly reduced frequency of CFU-F (colony forming unit-fibroblast). Moreover, we found an altered phenotype and reduced replating efficiency upon passaging of BMSC from MDS and AML samples. Expression analysis of genes involved in adipo- and osteogenic differentiation as well as Wnt- and Notch-signalling pathways showed significantly reduced levels of DLK1, an early adipogenic cell fate inhibitor in MDS and AML BMSC. Matching this observation, functional analysis showed significantly increased in vitro adipogenic differentiation potential in BMSC from MDS and AML patients. Overall, our data show BMSC with a reduced CFU-F capacity, and an altered molecular and functional profile from MDS and AML patients in culture, indicating an increased adipogenic lineage potential that is likely to provide a disease-promoting microenvironment.
Collapse
Affiliation(s)
- Marie-Theresa Weickert
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Judith S Hecker
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Michèle C Buck
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Christina Schreck
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Jennifer Rivière
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Matthias Schiemann
- Flow Cytometry Unit (CyTUM-MIH), Institute of Microbiology, Immunology, and Hygiene, Technical University of Munich, Munich, Germany
| | - Katharina Schallmoser
- Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria.,Department for Blood Group Serology and Transfusion Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Florian Bassermann
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Partner Site, Munich, Germany
| | - Dirk Strunk
- Experimental and Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - Robert A J Oostendorp
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Katharina S Götze
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany. .,German Cancer Consortium (DKTK), Heidelberg, Partner Site, Munich, Germany.
| |
Collapse
|
14
|
Cottrez F, Leblanc V, Boitel E, Groux H, Alépée N. The EyeIRR-IS assay: Development and evaluation of an in vitro assay to measure the eye irritation sub-categorization of liquid chemicals. Toxicol In Vitro 2021; 71:105072. [DOI: 10.1016/j.tiv.2020.105072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/27/2020] [Accepted: 12/17/2020] [Indexed: 11/30/2022]
|
15
|
Baulina N, Kiselev I, Favorova O. Imprinted Genes and Multiple Sclerosis: What Do We Know? Int J Mol Sci 2021; 22:1346. [PMID: 33572862 PMCID: PMC7866243 DOI: 10.3390/ijms22031346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune neurodegenerative disease of the central nervous system that arises from interplay between non-genetic and genetic risk factors. The epigenetics functions as a link between these factors, affecting gene expression in response to external influence, and therefore should be extensively studied to improve the knowledge of MS molecular mechanisms. Among others, the epigenetic mechanisms underlie the establishment of parent-of-origin effects that appear as phenotypic differences depending on whether the allele was inherited from the mother or father. The most well described manifestation of parent-of-origin effects is genomic imprinting that causes monoallelic gene expression. It becomes more obvious that disturbances in imprinted genes at the least affecting their expression do occur in MS and may be involved in its pathogenesis. In this review we will focus on the potential role of imprinted genes in MS pathogenesis.
Collapse
Affiliation(s)
- Natalia Baulina
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (I.K.); (O.F.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Ivan Kiselev
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (I.K.); (O.F.)
| | - Olga Favorova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (I.K.); (O.F.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
16
|
Masoudzadeh SH, Mohammadabadi M, Khezri A, Stavetska RV, Oleshko VP, Babenko OI, Yemets Z, Kalashnik OM. Effects of diets with different levels of fennel (Foeniculum vulgare) seed powder on DLK1 gene expression in brain, adipose tissue, femur muscle and rumen of Kermani lambs. Small Rumin Res 2020. [DOI: 10.1016/j.smallrumres.2020.106276] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
17
|
Rodríguez-Cano MM, González-Gómez MJ, Sánchez-Solana B, Monsalve EM, Díaz-Guerra MJM, Laborda J, Nueda ML, Baladrón V. NOTCH Receptors and DLK Proteins Enhance Brown Adipogenesis in Mesenchymal C3H10T1/2 Cells. Cells 2020; 9:cells9092032. [PMID: 32899774 PMCID: PMC7565505 DOI: 10.3390/cells9092032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 12/26/2022] Open
Abstract
The NOTCH family of receptors and ligands is involved in numerous cell differentiation processes, including adipogenesis. We recently showed that overexpression of each of the four NOTCH receptors in 3T3-L1 preadipocytes enhances adipogenesis and modulates the acquisition of the mature adipocyte phenotype. We also revealed that DLK proteins modulate the adipogenesis of 3T3-L1 preadipocytes and mesenchymal C3H10T1/2 cells in an opposite way, despite their function as non-canonical inhibitory ligands of NOTCH receptors. In this work, we used multipotent C3H10T1/2 cells as an adipogenic model. We used standard adipogenic procedures and analyzed different parameters by using quantitative-polymerase chain reaction (qPCR), quantitative reverse transcription-polymerase chain reaction (qRT-PCR), luciferase, Western blot, and metabolic assays. We revealed that C3H10T1/2 multipotent cells show higher levels of NOTCH receptors expression and activity and lower Dlk gene expression levels than 3T3-L1 preadipocytes. We found that the overexpression of NOTCH receptors enhanced C3H10T1/2 adipogenesis levels, and the overexpression of NOTCH receptors and DLK (DELTA-like homolog) proteins modulated the conversion of cells towards a brown-like adipocyte phenotype. These and our prior results with 3T3-L1 preadipocytes strengthen the idea that, depending on the cellular context, a precise and highly regulated level of global NOTCH signaling is necessary to allow adipogenesis and determine the mature adipocyte phenotype.
Collapse
Affiliation(s)
- María-Milagros Rodríguez-Cano
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (M.-M.R.-C.); (M.-J.G.-G.)
| | - María-Julia González-Gómez
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (M.-M.R.-C.); (M.-J.G.-G.)
| | - Beatriz Sánchez-Solana
- National Institutes of Health, Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA;
| | - Eva-María Monsalve
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (E.-M.M.); (M.-J.M.D.-G.)
| | - María-José M. Díaz-Guerra
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (E.-M.M.); (M.-J.M.D.-G.)
| | - Jorge Laborda
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (M.-M.R.-C.); (M.-J.G.-G.)
- Correspondence: (J.L.); (M.-L.N.); (V.B.); Tel.: +34-967-599-200 (ext. 2926) (V.B.); Fax: +34-967-599-327 (V.B.)
| | - María-Luisa Nueda
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (M.-M.R.-C.); (M.-J.G.-G.)
- Correspondence: (J.L.); (M.-L.N.); (V.B.); Tel.: +34-967-599-200 (ext. 2926) (V.B.); Fax: +34-967-599-327 (V.B.)
| | - Victoriano Baladrón
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (E.-M.M.); (M.-J.M.D.-G.)
- Correspondence: (J.L.); (M.-L.N.); (V.B.); Tel.: +34-967-599-200 (ext. 2926) (V.B.); Fax: +34-967-599-327 (V.B.)
| |
Collapse
|
18
|
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: 60] [Impact Index Per Article: 15.0] [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
|
19
|
Rahim F, Abbasi Pashaki P, Jafarisani M, Ghorbani F, Ebrahimi A. Runx2 silencing promotes adipogenesis via down-regulation of DLK1 in chondrogenic differentiating MSCs. J Gene Med 2020; 22:e3244. [PMID: 32559818 DOI: 10.1002/jgm.3244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND For cartilage regeneration, stem cells are a promising cell source; however, even the advances made in the differentiation of stem cells into precursor-differentiated cartilage cells have not been successful with respect to reprograming these cells to achieve complete differentiation and fully functioning cells until now. Previous findings suggest that Runx2 plays a major role in chondrocyte differentiation and maturation. Although targeting Runx2 has enhanced some chondrocyte properties, the adipogenic lineage shift has eventually occurred in these cells. The present study mainly aimed to reveal the mechanism of this adipogenesis. METHODS To create inducible artificial shRNA-miR expressing vectors, the designed short hairpin RNAs (shRNAs) were inserted into the pri-mir-30 backbone, cloned into lentiviral pLVET-Tet-on, and transducted into mesenchymal stem cells (MSCs). Runx2 gene was silenced in MSCs either for 1 week or 4 weeks and cultured in the chondrogenic medium. At days 7, 14 and 28, cells were harvested, and chondrogenesis, adipogenesis and hypertrophic states were examined using histochemical staining and a real-time polymerase chain reaction assay. RESULTS The results showed that the designed shRNA-miR effectively targeted Runx2 in mRNA and protein levels. Chondrogenic markers were up-regulated in constantly silenced Runx2 group; however, adipogenic markers and fat droplets appeared gradually. DLK1 gene was also significantly down-regulated in this group, and overexpression of DLK1 abrogated adipogenesis in the Runx2 targeted group. CONCLUSIONS Based on these results, it can be concluded that DLK1 is responsible for the lineage shift in Runx2 targeted chondrogenic differentiating MSCs.
Collapse
Affiliation(s)
- Fakher Rahim
- Thalassemia and Hemoglobinopathy Research Centre, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Moslem Jafarisani
- Department of Biochemistry, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Fatemeh Ghorbani
- Student Research Committee, Guilan University of Medical Sciences, Rasht, Iran
| | - Ammar Ebrahimi
- Department of Medical Biotechnology, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
20
|
The Role of Pref-1 during Adipogenic Differentiation: An Overview of Suggested Mechanisms. Int J Mol Sci 2020; 21:ijms21114104. [PMID: 32526833 PMCID: PMC7312882 DOI: 10.3390/ijms21114104] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 12/15/2022] Open
Abstract
Obesity contributes significantly to the global health burden. A better understanding of adipogenesis, the process of fat formation, may lead to the discovery of novel treatment strategies. However, it is of concern that the regulation of adipocyte differentiation has predominantly been studied using the murine 3T3-L1 preadipocyte cell line and murine experimental animal models. Translation of these findings to the human setting requires confirmation using experimental models of human origin. The ability of mesenchymal stromal/stem cells (MSCs) to differentiate into adipocytes is an attractive model to study adipogenesis in vitro. Differences in the ability of MSCs isolated from different sources to undergo adipogenic differentiation, may be useful in investigating elements responsible for regulating adipogenic differentiation potential. Genes involved may be divided into three broad categories: early, intermediate and late-stage regulators. Preadipocyte factor-1 (Pref-1) is an early negative regulator of adipogenic differentiation. In this review, we briefly discuss the adipogenic differentiation potential of MSCs derived from two different sources, namely adipose-derived stromal/stem cells (ASCs) and Wharton’s Jelly derived stromal/stem cells (WJSCs). We then discuss the function and suggested mechanisms of action of Pref-1 in regulating adipogenesis, as well as current findings regarding Pref-1’s role in human adipogenesis.
Collapse
|
21
|
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: 0.4] [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
|
22
|
Jensen CH, Kosmina R, Rydén M, Baun C, Hvidsten S, Andersen MS, Christensen LL, Gastaldelli A, Marraccini P, Arner P, Jørgensen CD, Laborda J, Holst JJ, Andersen DC. The imprinted gene Delta like non-canonical notch ligand 1 (Dlk1) associates with obesity and triggers insulin resistance through inhibition of skeletal muscle glucose uptake. EBioMedicine 2019; 46:368-380. [PMID: 31383551 PMCID: PMC6711890 DOI: 10.1016/j.ebiom.2019.07.070] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 07/29/2019] [Accepted: 07/29/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The imprinted gene Delta like non-canonical Notch ligand 1 (Dlk1) is considered an inhibitor of adipogenesis, but its in vivo impact on fat mass indeed remains elusive and controversial. METHODS Fat deposits were assessed by MRI and DXA scanning in two cohorts of non-diabetic men, whereas glucose disposal rate (GDR) was determined during euglycemic hyperinsulinemic clamp. Blood analyte measurements were used for correlation and mediation analysis to investigate how age, BMI, and fat percentage affect the relation between DLK1 and GDR. Confirmatory animal studies performed in normal (NC) and high fat diet (HFD) fed Dlk1+/+ and Dlk1-/- mice included DXA scanning, glucose tolerance tests (GTTs), blood measurements, and skeletal muscle glucose uptake studies by positron emission tomography (PET), histology, qRT-PCR, and in vitro cell studies. FINDINGS Overall, DLK1 is positively correlated with fat amounts, which is consistent with a negative linear relationship between DLK1 and GDR. This relationship is not mediated by age, BMI, or fat percentage. In support, DLK1 also correlates positively with HOMA-IR and ADIPO-IR in these humans, but has no linear relationship with the early diabetic inflammation marker MCP-1. In Dlk1-/- mice, the increase in fat percentage and adipocyte size induced by HFD is attenuated, and these animals are protected against insulin resistance. These Dlk1 effects seem independent of gluconeogenesis, but at least partly relies on increased in vivo glucose uptake in skeletal muscles by Dlk1 regulating the major glucose transporter Glut4 in vivo as well as in two independent cell lines. INTERPRETATION Thus, instead of an adipogenic inhibitor, Dlk1 should be regarded as a factor causally linked to obesity and insulin resistance, and may be used to predict development of type 2 diabetes. FUND: The Danish Diabetes Academy supported by the Novo Nordisk Foundation, The Danish National Research Council (#09-073648), The Lundbeck Foundation, University of Southern Denmark, and Dep. Of Clinical Biochemistry and Pharmacology/Odense University Hospital, the Swedish Research Council, the Swedish Diabetes Foundation, the Strategic Research Program in Diabetes at Karolinska Institute and an EFSD/Lilly grant.
Collapse
Affiliation(s)
- Charlotte Harken Jensen
- Laboratory of Molecular and Cellular Cardiology, Dep. of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Danish Center for Regenerative Medicine (danishcrm.com), Odense University Hospital, Denmark
| | - Rok Kosmina
- Laboratory of Molecular and Cellular Cardiology, Dep. of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; The Danish Diabetes Academy, Denmark; Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Mikael Rydén
- Dep. of Medicine-H7, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Christina Baun
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Svend Hvidsten
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | | | | | | | | | - Peter Arner
- Dep. of Medicine-H7, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Jorge Laborda
- Department of Inorganic and Organic Chemistry and Biochemistry, University of Castilla-La Mancha, Pharmacy School, Albacete, Spain
| | - Jens Juul Holst
- Department of Endocrinology and Metabolism, Section for Translational Metabolic Physiology, University of Copenhagen, Denmark
| | - Ditte Caroline Andersen
- Laboratory of Molecular and Cellular Cardiology, Dep. of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Danish Center for Regenerative Medicine (danishcrm.com), Odense University Hospital, Denmark; Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Clinical Institute, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
23
|
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: 4.2] [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]
|
24
|
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: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2019] [Indexed: 10/27/2022]
|
25
|
Zhao BH, Jiang Y, Zhu H, Xi FF, Chen Y, Xu YT, Liu F, Wang YY, Hu WS, Lv WG, Luo Q. Placental Delta-Like 1 Gene DNA Methylation Levels Are Related to Mothers' Blood Glucose Concentration. J Diabetes Res 2019; 2019:9521510. [PMID: 31886292 PMCID: PMC6927055 DOI: 10.1155/2019/9521510] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 11/04/2019] [Indexed: 12/20/2022] Open
Abstract
PURPOSE We aim to identify the methylation status of delta-like 1 (DLK1) in the placenta and the correlation between DLK1 methylation and maternal serum glucose level and fetal birth weight. METHODS We analyzed the gene expression of DLK1 gene in both maternal and fetal sides of the placenta in a GDM group (n = 15) and a control group (n = 15) using real-time polymerase chain reaction. With MethylTargetTM technique, we detected the methylation status of DLK1 promotor in the placenta. Furthermore, Pearson's correlation was used to confirm the association of methylation alteration of DLK1 promoter and maternal 2 h OGTT glucose level and fetal birth weight. RESULTS In our study, we found that DLK1 expression in both maternal and fetal sides of the placenta decreased significantly in GDM group compared with control group, and it was caused by hypermethylation of DLK1 promoter region. Additionally, the methylation status of DLK1 gene in the maternal side of the placenta highly correlated with maternal 2 h OGTT glucose level (coefficient = 0.7968, P < 0.0001), while the methylation status in the fetal side of the placenta was closely related to fetal birth weight (coefficient = 0.6233, P < 0.0001). CONCLUSIONS Our results demonstrated that altered expression of DLK1 was caused by the hypermethylation of DLK1 promoter region in the placenta, and intrauterine exposure to GDM has long-lasting effects on the epigenome of the offspring.
Collapse
Affiliation(s)
- Bai-Hui Zhao
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Jiang
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Zhu
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital of China Welfare institute, Shanghai, China
| | - Fang-Fang Xi
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Chen
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ye-Tao Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fang Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ya-Yun Wang
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen-Sheng Hu
- Department of Obstetrics, Maternal and Child Health Care Hospital, Hangzhou, China
| | - Wei-Guo Lv
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiong Luo
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
26
|
Nueda ML, González-Gómez MJ, Rodríguez-Cano MM, Monsalve EM, Díaz-Guerra MJM, Sánchez-Solana B, Laborda J, Baladrón V. DLK proteins modulate NOTCH signaling to influence a brown or white 3T3-L1 adipocyte fate. Sci Rep 2018; 8:16923. [PMID: 30446682 PMCID: PMC6240076 DOI: 10.1038/s41598-018-35252-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 11/01/2018] [Indexed: 02/05/2023] Open
Abstract
The role of NOTCH signaling in adipogenesis is highly controversial, with data indicating null, positive or negative effects on this differentiation process. We hypothesize that these contradictory results could be due to the different global NOTCH signaling levels obtained in different experimental settings, because of a specific modulation of NOTCH receptors’ activity by their ligands. We have previously demonstrated that DLK1 and DLK2, two non-canonical NOTCH1 ligands that inhibit NOTCH1 signaling in a dose-dependent manner, modulate the adipogenesis process of 3T3-L1 preadipocytes. In this work, we show that over-expression of any of the four NOTCH receptors enhanced adipogenesis of 3T3-L1 preadipocytes. We also determine that DLK proteins inhibit not only the activity of NOTCH1, but also the activity of NOTCH2, 3 and 4 receptors to different degrees. Interestingly, we have observed, by different approaches, that NOTCH1 over-expression seems to stimulate the differentiation of 3T3-L1 cells towards a brown-like adipocyte phenotype, whereas cells over-expressing NOTCH2, 3 or 4 receptors or DLK proteins would rather differentiate towards a white-like adipocyte phenotype. Finally, our data also demonstrate a complex feed-back mechanism involving Notch and Dlk genes in the regulation of their expression, which suggest that a precise level of global NOTCH expression and NOTCH-dependent transcriptional activity of specific targets could be necessary to determine the final phenotype of 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- María-Luisa Nueda
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica y Bioquímica, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC. C/Almansa 14, 02008, Albacete, Spain
| | - María-Julia González-Gómez
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica y Bioquímica, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC. C/Almansa 14, 02008, Albacete, Spain
| | - María-Milagros Rodríguez-Cano
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica y Bioquímica, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC. C/Almansa 14, 02008, Albacete, Spain
| | - Eva-María Monsalve
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica, Orgánica y Bioquímica, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - María José M Díaz-Guerra
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica, Orgánica y Bioquímica, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - Beatriz Sánchez-Solana
- Laboratory of Cellular Oncology, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jorge Laborda
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica y Bioquímica, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC. C/Almansa 14, 02008, Albacete, Spain
| | - Victoriano Baladrón
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica, Orgánica y Bioquímica, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain.
| |
Collapse
|
27
|
García-Gutiérrez MS, Navarrete F, Laborda J, Manzanares J. Deletion of Dlk1 increases the vulnerability to developing anxiety-like behaviors and ethanol consumption in mice. Biochem Pharmacol 2018; 158:37-44. [PMID: 30268817 DOI: 10.1016/j.bcp.2018.09.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/26/2018] [Indexed: 12/18/2022]
Abstract
Anxiety and alcohol use disorders (AUD) often present together, constituting a significant public health problem worldwide. In this study, we investigated the role of DLK1, a ligand of the Delta/NOTCH epidermal growth factor (EGF)-like protein family, reported to play a role in DA neurons differentiation in the striatum, as a neurobiological factor involved in the mechanisms regulating this psychiatric comorbidity. We exposed Dlk1 knockout mice (Dlk1-/- mice) to the open-field (OF), the light-dark box (LBD) and the elevated plus maze (EPM) tests, evaluating motivation to drink and ethanol consumption using the oral ethanol self-administration (OEA) paradigm. Quantitative real time polymerase chain reaction (qPCR) studies were carried out to evaluate alterations in targets closely related to DA neurotransmission in the reward system, tyrosine hydroxylase (Th) in the ventral tegmental area (VTA), and μ-opioid receptor (Oprm1) in the nucleus accumbens (NAc). No differences were observed in the total or peripheral distances travelled by Dlk1-/- compared to wild-type (WT) mice in OF. However, central distance travelled significantly decreased in Dlk1-/- mice. Deletion of Dlk1 increased anxiety-like behaviors in the LDB and EPM, and, Dlk1-/- mice also presented higher ethanol intake and motivation to drink (number of effective responses) in the OEA. In addition, Th and Oprm1 gene expression was reduced in the VTA and NAc of Dlk1-/- mice. We conclude that deletion of Dlk1 increases anxiety-related behaviors and vulnerability to ethanol consumption and modifies the gene expression of key targets closely related with DA neurotransmission involved in the reinforcing actions of ethanol.
Collapse
Affiliation(s)
- María S García-Gutiérrez
- Institute of Neurosciences, Miguel Hernández University-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; Topic-based Network for Cooperative Health Research (RETICS), Substance Abuse Network, Health Institute Carlos III, MICINN and FEDER, Madrid, Spain
| | - Francisco Navarrete
- Institute of Neurosciences, Miguel Hernández University-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; Topic-based Network for Cooperative Health Research (RETICS), Substance Abuse Network, Health Institute Carlos III, MICINN and FEDER, Madrid, Spain
| | - Jorge Laborda
- School of Pharmacy, Regional Center for Biomedical Research (CRIB), Biomedicine Unit UCLM-CSIC, Albacete, Spain
| | - Jorge Manzanares
- Institute of Neurosciences, Miguel Hernández University-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; Topic-based Network for Cooperative Health Research (RETICS), Substance Abuse Network, Health Institute Carlos III, MICINN and FEDER, Madrid, Spain.
| |
Collapse
|
28
|
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: 1.3] [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
|
29
|
Drake KA, Adam M, Mahoney R, Potter SS. Disruption of Hox9,10,11 function results in cellular level lineage infidelity in the kidney. Sci Rep 2018; 8:6306. [PMID: 29679048 PMCID: PMC5910417 DOI: 10.1038/s41598-018-24782-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/10/2018] [Indexed: 01/09/2023] Open
Abstract
Hox genes are important regulators of development. The 39 mammalian Hox genes have considerable functional overlap, greatly confounding their study. In this report, we generated mice with multiple combinations of paralogous and flanking Abd-B Hox gene mutations to investigate functional redundancies in kidney development. The resulting mice developed a number of kidney abnormalities, including hypoplasia, agenesis, and severe cysts, with distinct Hox functions observed in early metanephric kidney formation and nephron progenitor maintenance. Most surprising, however, was that extensive removal of Hox shared function in these kidneys resulted in cellular level lineage infidelity. Strikingly, mutant nephron tubules consisted of intermixed cells with proximal tubule, loop of Henle, and collecting duct identities, with some single cells expressing markers associated with more than one nephron segment. These results indicate that Hox genes are required for proper lineage selection/maintenance and full repression of genes involved in cell fate restriction in the developing kidney.
Collapse
Affiliation(s)
- Keri A Drake
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Robert Mahoney
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| |
Collapse
|
30
|
Wang X, Yuan W, Wang X, Qi J, Qin Y, Shi Y, Zhang J, Gong J, Dong Z, Liu X, Sun C, Chai R, Le Noble F, Liu D. The somite-secreted factor Maeg promotes zebrafish embryonic angiogenesis. Oncotarget 2018; 7:77749-77763. [PMID: 27780917 PMCID: PMC5363618 DOI: 10.18632/oncotarget.12793] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 10/12/2016] [Indexed: 01/06/2023] Open
Abstract
MAM and EGF containing gene (MAEG), also called Epidermal Growth Factor-like domain multiple 6 (EGFL6), belongs to the epidermal growth factor repeat superfamily. The role of Maeg in zebrafish angiogenesis remains unclear. It was demonstrated that maeg was dynamically expressed in zebrafish developing somite during a time window encompassing many key steps in embryonic angiogenesis. Maeg loss-of-function embryos showed reduced endothelial cell number and filopodia extensions of intersegmental vessels (ISVs). Maeg gain-of-function induced ectopic sprouting evolving into a hyperbranched and functional perfused vasculature. Mechanistically we demonstrate that Maeg promotes angiogenesis dependent on RGD domain and stimulates activation of Akt and Erk signaling in vivo. Loss of Maeg or Itgb1, augmented expression of Notch receptors, and inhibiting Notch signaling or Dll4 partially rescued angiogenic phenotypes suggesting that Notch acts downstream of Itgb1. We conclude that Maeg acts as a positive regulator of angiogenic cell behavior and formation of functional vessels.
Collapse
Affiliation(s)
- Xin Wang
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Wei Yuan
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Xueqian Wang
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Jialing Qi
- Medical College, Nantong University, Nantong, China
| | - Yinyin Qin
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Yunwei Shi
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Jie Zhang
- Medical College, Nantong University, Nantong, China
| | - Jie Gong
- School of life science, Nantong University, Nantong, China
| | - Zhangji Dong
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaoyu Liu
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Chen Sun
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Renjie Chai
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China.,Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Ferdinand Le Noble
- Department of Cell and Developmental Biology, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Dong Liu
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
31
|
Comparative transcriptome analysis reveals potentially novel roles of Homeobox genes in adipose deposition in fat-tailed sheep. Sci Rep 2017; 7:14491. [PMID: 29101335 PMCID: PMC5670210 DOI: 10.1038/s41598-017-14967-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 10/18/2017] [Indexed: 12/12/2022] Open
Abstract
Adipose tissues are phenotypically, metabolically and functionally heterogeneous based on the sites of their deposition. Undesirable fat deposits in the body are often detrimental to animal and human health. To unravel the potential underlying mechanisms governing accumulation of adipose tissues in various regions of the body, i.e., subcutaneous (SAT), visceral (VAT) and tail (TAT), we profiled transcriptomes from Tan sheep, a Chinese indigenous breed with notable fat tail using RNA-seq. Upon comparison, we identified a total of 1,058 differentially expressed genes (DEGs) between the three adipose types (218, 324, and 795 in SAT/VAT, SAT/TAT, and VAT/TAT, respectively), from which several known key players were identified that are involved in lipid metabolic process, Wnt signals, Vitamin A metabolism, and transcriptional regulation of adipocyte differentiation. We also found that many elevated genes in VAT were notably enriched for key biological processes such as cytokine secretion, signaling molecule interaction and immune systems. Several developmental genes including HOXC11, HOXC12 and HOXC13, and adipose-expressed genes in the tail region, such as HOTAIR_2, HOTAIR_3 and SP9 were specially highlighted, indicating their strong associations with tail fat development in fat-tailed sheep. Our results provide new insight into exploring the specific fat deposition in tail, also contribute to the understanding of differences between adipose depots.
Collapse
|
32
|
The adaptor SASH1 acts through NOTCH1 and its inhibitor DLK1 in a 3D model of lumenogenesis involving CEACAM1. Exp Cell Res 2017; 359:384-393. [DOI: 10.1016/j.yexcr.2017.08.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 08/02/2017] [Accepted: 08/13/2017] [Indexed: 12/21/2022]
|
33
|
Chean J, Chen CJ, Shively JE. ETS transcription factor ELF5 induces lumen formation in a 3D model of mammary morphogenesis and its expression is inhibited by Jak2 inhibitor TG101348. Exp Cell Res 2017; 359:62-75. [PMID: 28800960 DOI: 10.1016/j.yexcr.2017.08.008] [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: 04/01/2017] [Revised: 08/02/2017] [Accepted: 08/05/2017] [Indexed: 02/06/2023]
Abstract
The loss of expression of a single gene can revert normal tissue to a malignant phenotype. For example, while normal breast has high lumenal expression of CEACAM1, the majority of breast cancers exhibit the early loss of this gene with the concurrent loss of their lumenal phenotype. MCF7 cells that lack CEACAM1 expression and fail to form lumena in 3D culture, regain the normal phenotype when transfected with CEACAM1. In order to probe the mechanism of this gain of function, we treated these cells with the clinically relevant Jak2 inhibitor TG101348 (TG), expecting that disruption of the prolactin receptor signaling pathway would interfere with the positive effects of transfection of MCF7 cells with CEACAM1. Indeed, lumen formation was inhibited, resulting in the down regulation of a set of genes, likely involved in the complex process of lumen formation. As expected, inhibition of the expression of many of these genes also inhibited lumen formation, confirming their involvement in a single pathway. Among the genes identified by the inhibition assay, ETS transcription factor ELF5 stood out, since it has been identified as a master regulator of mammary morphogenesis, and is associated with prolactin receptor signaling. When ELF5 was transfected into the parental MCF7 cells that lack CEACAM1, lumen formation was restored, indicating that ELF5 can replace CEACAM1 in this model system of lumenogenesis. We conclude that the event(s) that led to the loss of expression of CEACAM1 is epistatic in that multiple genes associated with a critical pathway were affected, but that restoration of the normal phenotype can be achieved with reactivation of certain genes at various nodal points in tissue morphogenesis.
Collapse
Affiliation(s)
- Jennifer Chean
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, 1450 E. Duarte Road, Duarte, CA 91010, USA
| | - Charng-Jui Chen
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, 1450 E. Duarte Road, Duarte, CA 91010, USA
| | - John E Shively
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, 1450 E. Duarte Road, Duarte, CA 91010, USA.
| |
Collapse
|
34
|
Bello AR, Puertas‐Avendaño RA, González‐Gómez MJ, González‐Gómez M, Laborda J, Damas C, Ruiz‐Hidalgo M, Diaz C. Delta-like protein 1 in the pituitary-adipose axis in the adult male mouse. J Neuroendocrinol 2017; 29:e12507. [PMID: 28718206 PMCID: PMC6084355 DOI: 10.1111/jne.12507] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 06/21/2017] [Accepted: 07/12/2017] [Indexed: 12/18/2022]
Abstract
With the aim of studying delta-like protein 1 (DLK1) with respect to the relationship between adipocyte leptin and adenohypophyseal hormones, we carried out an immunohistochemical study analysing the presence of receptors for these hormones in the pituitary and adipose cells of male wild-type (WT) mice (Dlk1+/+ ) compared to knockout (KO) mice (Dlk1-/- ). The mRNA expression of these molecules was also determined using the reverse transcriptase-polymerase chain reaction. The results obtained showed that, in WT adipose cells, all of the adenohypophyseal hormone receptors were present, with a higher mRNA expression for growth hormone (GH) receptor and thyroid-stimulating hormone (TSH) receptor. Of the total cells in the anterior pituitary lobe, 17.09±0.9% were leptin receptor (LEPR) immunoreactive (-IR), mainly in GH-IR and prolactin (PRL)-IR cells (41.5±3.8%; 13.5±1.7%, respectively). In Dlk1-/- mice, adipocyte cells showed a significant increase in the TSH receptor mRNA expression level. Moreover, the percentage of LEPR-IR GH cells showed a statistically significant increase compared to controls, from 41.5±3.8% to 53.1±4.0%. By contrast, only 3.0±0.6% of LEP-IR anterior pituitary cells were detected in Dlk1 KO mice, as opposed to 6.8±1.1% observed in WT mice. The results suggest that relationships exist between adipocytes and pituitary GH, PRL and TSH cells, in addition to an influence with respect to the synthesis and release of pituitary leptin, particularly in PRL cells.
Collapse
Affiliation(s)
- A. R. Bello
- Cell Biology SectionSchool of Sciences/Institute for Tropical Diseases and Public HealthUniversity of La LagunaTenerifeSpain
| | - R. A. Puertas‐Avendaño
- Cell Biology SectionSchool of Sciences/Institute for Tropical Diseases and Public HealthUniversity of La LagunaTenerifeSpain
| | - M. J. González‐Gómez
- Department of Inorganic and Organic Chemistry and BiochemistrySchool of Medicine/Regional Centre for Biomedical ResearchBiomedicine Unit Spanish National Research Council/University of Castilla‐La ManchaAlbaceteSpain
| | - M. González‐Gómez
- Department of Basic Medical SciencesSchool of MedicineUniversity of La LagunaTenerifeSpain
| | - J. Laborda
- Department of Inorganic and Organic Chemistry and BiochemistrySchool of Medicine/Regional Centre for Biomedical ResearchBiomedicine Unit Spanish National Research Council/University of Castilla‐La ManchaAlbaceteSpain
| | - C. Damas
- Department of PsychobiologySchool of PsychologyUniversity of La LagunaTenerifeSpain
| | - M. Ruiz‐Hidalgo
- Department of Inorganic and Organic Chemistry and BiochemistrySchool of Medicine/Regional Centre for Biomedical ResearchBiomedicine Unit Spanish National Research Council/University of Castilla‐La ManchaAlbaceteSpain
| | - C. Diaz
- Department of Medical SciencesSchool of Medicine/Institute for Research in Neurological DisabilitiesUniversity of Castilla‐La ManchaAlbaceteSpain
| |
Collapse
|
35
|
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: 2.0] [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
|
36
|
Zhong Z, Ye Y, Guo W, He Y, Hu W. Relationship between DLK1 gene promoter region DNA methylation and non-small cell lung cancer biological behavior. Oncol Lett 2017; 13:4123-4126. [PMID: 28588700 PMCID: PMC5452895 DOI: 10.3892/ol.2017.6019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 02/10/2017] [Indexed: 02/06/2023] Open
Abstract
We investigated the possible association between DLKI gene promoter region methylation and the increased invasion capacity of non-small cell lung cancer (NSCLC). Lung cancer cell line H1299, as well as the gene transfection and RNA interference technology were used to build DLK gene overexpression and knockdown cells. An in vitro invasion assay was performed to observe the changes in the invasion ability of lung cancer cells. Western blot analysis was used to verify Notchl and matrix metalloproteinase-9 (MMP-9) expression levels and a sulfurous acid sequencing technique was used to test the DNA methylation level in the promoter region. Our results showed that the invasion ability of cells in the overexpression group was significantly enhanced. This ability was considerably reduced in the knockdown group. The Notchl and MMP-9 expression level increased significantly in the overexpression group, while it was reduced considerably in the knockdown group. We detected significantly lower levels of DNA methylation in the promoter region in the overexpression group. It was concluded that methylation of the DLK1 gene promoter region increased the invasion ability of NSCLC. Furthermore, it is possible that this process is related to the Notch signaling pathway.
Collapse
Affiliation(s)
- Zhaokui Zhong
- Department of Thoracic Surgery, Central Hospital of Zhumadian, Zhumadian, Henan 463000, P.R. China
| | - Yongting Ye
- Department of Internal Medicine, The Fourth People's Hospital of Zhumadian, Zhumadian, Henan 463000, P.R. China
| | - Wei Guo
- Department of Thoracic Surgery, Central Hospital of Zhumadian, Zhumadian, Henan 463000, P.R. China
| | - Yi He
- Department of Thoracic Surgery, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Weicai Hu
- Department of Thoracic Surgery, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
37
|
Plikus MV, Guerrero-Juarez CF, Ito M, Li YR, Dedhia PH, Zheng Y, Shao M, Gay DL, Ramos R, Hsi TC, Oh JW, Wang X, Ramirez A, Konopelski SE, Elzein A, Wang A, Supapannachart RJ, Lee HL, Lim CH, Nace A, Guo A, Treffeisen E, Andl T, Ramirez RN, Murad R, Offermanns S, Metzger D, Chambon P, Widgerow AD, Tuan TL, Mortazavi A, Gupta RK, Hamilton BA, Millar SE, Seale P, Pear WS, Lazar MA, Cotsarelis G. Regeneration of fat cells from myofibroblasts during wound healing. Science 2017; 355:748-752. [PMID: 28059714 DOI: 10.1126/science.aai8792] [Citation(s) in RCA: 387] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/19/2016] [Indexed: 12/14/2022]
Abstract
Although regeneration through the reprogramming of one cell lineage to another occurs in fish and amphibians, it has not been observed in mammals. We discovered in the mouse that during wound healing, adipocytes regenerate from myofibroblasts, a cell type thought to be differentiated and nonadipogenic. Myofibroblast reprogramming required neogenic hair follicles, which triggered bone morphogenetic protein (BMP) signaling and then activation of adipocyte transcription factors expressed during development. Overexpression of the BMP antagonist Noggin in hair follicles or deletion of the BMP receptor in myofibroblasts prevented adipocyte formation. Adipocytes formed from human keloid fibroblasts either when treated with BMP or when placed with human hair follicles in vitro. Thus, we identify the myofibroblast as a plastic cell type that may be manipulated to treat scars in humans.
Collapse
Affiliation(s)
- Maksim V Plikus
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA. .,Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Mayumi Ito
- The Ronald O. Perelman Department of Dermatology, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Yun Rose Li
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Priya H Dedhia
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying Zheng
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mengle Shao
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Denise L Gay
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,INSERM U967, Commissariat à L'énergie Atomique et aux Énergies Alternatives, Institut de Radiobiologie Cellulaire et Moléculaire 92265 Fontenay-aux-Roses Cedex, France
| | - Raul Ramos
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Tsai-Ching Hsi
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Ji Won Oh
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.,Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Xiaojie Wang
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Amanda Ramirez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Sara E Konopelski
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Arijh Elzein
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Anne Wang
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rarinthip June Supapannachart
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hye-Lim Lee
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Chae Ho Lim
- The Ronald O. Perelman Department of Dermatology, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Arben Nace
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amy Guo
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elsa Treffeisen
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 328116, USA
| | - Ricardo N Ramirez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Rabi Murad
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964, Université de Strasbourg, Illkirch 67404, France
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964, Institut d'Etudes Avancées de l'Université de Strasbourg, Collège de France, Illkirch 67404, France
| | - Alan D Widgerow
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, Irvine, CA 92868, USA
| | - Tai-Lan Tuan
- The Saban Research Institute of Children's Hospital Los Angeles and Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bruce A Hamilton
- Departments of Medicine and Cellular and Molecular Medicine, Moores Cancer Center and Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sarah E Millar
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick Seale
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Warren S Pear
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitchell A Lazar
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - George Cotsarelis
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
38
|
Traustadóttir GÁ, Jensen CH, Thomassen M, Beck HC, Mortensen SB, Laborda J, Baladrón V, Sheikh SP, Andersen DC. Evidence of non-canonical NOTCH signaling: Delta-like 1 homolog (DLK1) directly interacts with the NOTCH1 receptor in mammals. Cell Signal 2016; 28:246-54. [PMID: 26791579 DOI: 10.1016/j.cellsig.2016.01.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/08/2016] [Accepted: 01/08/2016] [Indexed: 12/21/2022]
Abstract
Canonical NOTCH signaling, known to be essential for tissue development, requires the Delta-Serrate-LAG2 (DSL) domain for NOTCH to interact with its ligand. However, despite lacking DSL, Delta-like 1 homolog (DLK1), a protein that plays a significant role in mammalian development, has been suggested to interact with NOTCH1 and act as an antagonist. This non-canonical interaction is, however controversial, and evidence for a direct interaction, still lacking in mammals. In this study, we elucidated the putative DLK1-NOTCH1 interaction in a mammalian context. Taking a global approach and using Dlk1(+/+) and Dlk1(-/-) mouse tissues at E16.5, we demonstrated that several NOTCH signaling pathways indeed are affected by DLK1 during tissue development, and this was supported by a lower activation of NOTCH1 protein in Dlk1(+/+) embryos. Likewise, but using a distinct Dlk1-manipulated (siRNA) setup in a mammalian cell line, NOTCH signaling was substantially inhibited by DLK1. Using a mammalian two-hybrid system, we firmly established that the effect of DLK1 on NOTCH signaling was due to a direct interaction between DLK1 and NOTCH1. By careful dissection of this mechanism, we found this interaction to occur between EGF domains 5 and 6 of DLK1 and EGF domains 10-15 of NOTCH1. Thus, our data provide the first evidence for a direct interaction between DLK1 and NOTCH1 in mammals, and substantiate that non-canonical NOTCH ligands exist, adding to the complexity of NOTCH signaling.
Collapse
Affiliation(s)
- Gunnhildur Ásta Traustadóttir
- Laboratory of Molecular and Cellular Cardiology, Dep. of Clinical Biochemistry and Pharmacology, Odense University Hospital (OUH), Winsloewparken 21 3rd, 5000 Odense C, Denmark; Cardiovascular and Renal Research (University of Southern Denmark), OUH, Denmark
| | - Charlotte H Jensen
- Laboratory of Molecular and Cellular Cardiology, Dep. of Clinical Biochemistry and Pharmacology, Odense University Hospital (OUH), Winsloewparken 21 3rd, 5000 Odense C, Denmark; The Danish Centre for Regenerative Medicine (danishcrm@com), OUH, Denmark
| | - Mads Thomassen
- Dep. of Clinical Genetics (OUH), OUH, Denmark; Clinical Institute (University of Southern Denmark), OUH, Denmark
| | - Hans Christian Beck
- Clinical Institute (University of Southern Denmark), OUH, Denmark; Dep. of Clinical Biochemistry and Pharmacology (OUH), OUH, Denmark
| | - Sussi B Mortensen
- Laboratory of Molecular and Cellular Cardiology, Dep. of Clinical Biochemistry and Pharmacology, Odense University Hospital (OUH), Winsloewparken 21 3rd, 5000 Odense C, Denmark; Dep. of Clinical Immunology (OUH), Denmark
| | - Jorge Laborda
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, Albacete, Spain
| | - Victoriano Baladrón
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, Albacete, Spain
| | - Søren P Sheikh
- Laboratory of Molecular and Cellular Cardiology, Dep. of Clinical Biochemistry and Pharmacology, Odense University Hospital (OUH), Winsloewparken 21 3rd, 5000 Odense C, Denmark; Cardiovascular and Renal Research (University of Southern Denmark), OUH, Denmark; The Danish Centre for Regenerative Medicine (danishcrm@com), OUH, Denmark
| | - Ditte C Andersen
- Laboratory of Molecular and Cellular Cardiology, Dep. of Clinical Biochemistry and Pharmacology, Odense University Hospital (OUH), Winsloewparken 21 3rd, 5000 Odense C, Denmark; Clinical Institute (University of Southern Denmark), OUH, Denmark; The Danish Centre for Regenerative Medicine (danishcrm@com), OUH, Denmark.
| |
Collapse
|
39
|
Jagtap S, Khare P, Mangal P, Kondepudi KK, Bishnoi M, Bhutani KK. Protective effects of phyllanthin, a lignan from Phyllanthus amarus, against progression of high fat diet induced metabolic disturbances in mice. RSC Adv 2016. [DOI: 10.1039/c6ra10774e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Phyllanthin delayed the progression of high fat diet induced changes affecting lipid and glucose metabolism such as adiposity, hypertriglyceridemia, fatty liver, inflammation, lipid peroxidation and insulin resistance.
Collapse
Affiliation(s)
- Sneha Jagtap
- Department of Natural Products
- National Institute of Pharmaceutical Education and Research (NIPER)
- S.A.S. Nagar
- India
| | - Pragyanshu Khare
- National Agri-Food Biotechnology Institute (NABI)
- S.A.S. Nagar
- India
| | - Priyanka Mangal
- Department of Natural Products
- National Institute of Pharmaceutical Education and Research (NIPER)
- S.A.S. Nagar
- India
| | | | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute (NABI)
- S.A.S. Nagar
- India
| | - Kamlesh Kumar Bhutani
- Department of Natural Products
- National Institute of Pharmaceutical Education and Research (NIPER)
- S.A.S. Nagar
- India
| |
Collapse
|
40
|
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: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 12/10/2015] [Indexed: 01/23/2023]
|
41
|
Gil-García B, Baladrón V. The complex role of NOTCH receptors and their ligands in the development of hepatoblastoma, cholangiocarcinoma and hepatocellular carcinoma. Biol Cell 2015; 108:29-40. [DOI: 10.1111/boc.201500029] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 11/24/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Borja Gil-García
- Laboratory of Biochemistry and Molecular Biology; Department of Inorganic and Organic Chemistry and Biochemistry; Medical School/CRIB/Biomedicine Unit; University of Castilla-La Mancha (UCLM)/CSIC; 02008, Albacete Spain
| | - Victoriano Baladrón
- Laboratory of Biochemistry and Molecular Biology; Department of Inorganic and Organic Chemistry and Biochemistry; Medical School/CRIB/Biomedicine Unit; University of Castilla-La Mancha (UCLM)/CSIC; 02008, Albacete Spain
| |
Collapse
|
42
|
Delta-Like-1 Changes the Immunomodulatory Property of OP9 Cells. Stem Cells Int 2015; 2016:1628352. [PMID: 26649045 PMCID: PMC4663344 DOI: 10.1155/2016/1628352] [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: 01/29/2015] [Revised: 04/10/2015] [Accepted: 04/20/2015] [Indexed: 01/10/2023] Open
Abstract
As stromal cells and recently confirmed mesenchymal stem cells, OP9 cells support hematopoiesis stem cell (HSC) differentiation into the B lymphocyte lineage, yet Delta-like-1 (DL1) overexpressing OP9 (OP9DL1) cells promote the development of early T lymphocytes from HSC. However, the immunomodulatory capacity of OP9 or OP9DL1 on mature B and T cell proliferation has not been elucidated. Here, we show that OP9 and OP9DL1 have similar proliferation capacities and immunophenotypes except DL1 expression. Compared with OP9, OP9DL1 displayed more osteogenesis and less adipogenesis when cultured in the respective induction media. Both OP9 and OP9DL1 inhibited mature B and T cell proliferation. Furthermore, OP9 showed stronger inhibition on B cell proliferation and OP9DL1 exhibited stronger inhibition on T cell proliferation. With stimulation, both OP9 and OP9DL1 showed increased nitrate oxide (NO) production. The NO levels of OP9 were higher than that of OP9DL1 when stimulated with TNFα/IFNγ or LPS/IL4. Taken together, our study reveals a previously unrecognized role of OP9 and OP9DL1 in mature B and T cell proliferation. DL1 overexpression alone changed the properties of OP9 cells in addition to their role in early B cell development.
Collapse
|
43
|
Exogenous administration of DLK1 ameliorates hepatic steatosis and regulates gluconeogenesis via activation of AMPK. Int J Obes (Lond) 2015; 40:356-65. [PMID: 26315841 DOI: 10.1038/ijo.2015.173] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 08/07/2015] [Accepted: 08/13/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND/OBJECTIVES Activation of Notch signaling pathologically enhances lipogenesis and gluconeogenesis in the liver causing non-alcoholic fatty liver disease (NAFLD) and diabetes. Delta-like 1 homolog (DLK1), an imprinted gene that can modulate adipogenesis and muscle development in mice, was found as an inhibitory regulator of Notch signaling. Therefore, we investigated the metabolic effect of exogenous DLK1 in vitro and in vivo. SUBJECTS/METHODS A soluble DLK1 peptide was generated with fusion between a human Fc fragment and extracellular domain of DLK1. Male db/db mice were randomly assigned to two groups: vehicle treated and DLK1-treated group (25 mg kg(-1), intraperitoneal injection, twice a week for 4 weeks). Primary mice hepatocytes and HepG2 cells were used for in vitro experiments. RESULTS After 4 weeks of DLK1 administration, hepatic triglyceride content and lipid droplets in liver tissues, as well as serum levels of liver enzymes, were markedly decreased in db/db mice. DLK1 treatment induced phosphorylation of AMPK and ACC and suppressed nuclear expression of SREBP-1c in the mouse liver or hepatocytes, indicating regulation of fatty acid oxidation and synthesis pathways. Furthermore, DLK1-treated mice showed significantly lower levels of fasting and random glucose, with improved glucose and insulin tolerance compared with the vehicle-treated group. Macrophage infiltration and proinflammatory cytokine levels in the epididymal fat were decreased in DLK1-treated db/db mice. Moreover, DLK1 suppressed glucose production from hepatocytes, which was blocked after co-administration of an AMPK inhibitor, compound C. DLK1-treated hepatocytes and mouse liver tissues showed lower PEPCK and G6Pase expression. DLK1 triggered AKT phosphorylation followed by cytosolic translocation of FOXO1 from the nucleus in hepatocytes. CONCLUSIONS The present study demonstrated that exogenous administration of DLK1 reduced hepatic steatosis and hyperglycemia via AMPK activation in the liver. This result suggests that DLK1 may be a novel therapeutic approach for treating NAFLD and diabetes.
Collapse
|
44
|
Prenatal notch1 receptor blockade by protein delta homolog 1 (DLK1) modulates adipocyte size in vivo. Int J Obes (Lond) 2015; 40:698-705. [PMID: 26499442 DOI: 10.1038/ijo.2015.227] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 09/12/2015] [Accepted: 10/01/2015] [Indexed: 12/21/2022]
Abstract
INTRODUCTION/OBJECTIVES The protein delta homolog 1 (DLK1) has been reported to have an important role as inhibitor of adipogenesis. Understanding its mode of action can be a promising approach to cope with the formation of obesity. However, data on DLK1 signaling are not consistent, and especially its role as negative regulator of Notch receptors is discussed controversially. METHODS DLK1 effects have been investigated in differentiated 3T3-L1 cells by Adipokine Profiler Array, enzyme-linked immunosorbent assay and quantitative real-time PCR (qRT-PCR). In vivo effects of DLK1 on adipogenesis have been studied by the DLK1 treatment of pregnant C57BL/6NTac mice and the phenotypical characterization of the offspring fed on chow or high-fat diet (HFD). Furthermore, gene expression of key adipogenesis genes in adipose tissue (AT) samples was observed by qRT-PCR. RESULTS In 3T3-L1 cells, DLK1 was found to be an inhibitor of Notch1 signaling. Gene expression of Notch1 and Hes1 was lowered by 53% and 65%, respectively, and the expression of protein target PAI-1 was decreased by 51%. The offspring of DLK1-treated pregnant mice were fed chow or HFD starting from week 4. At week 18, a larger proportion of visceral AT was determined on HFD after DLK1 treatment (P=0.011), whereas adipocyte size was reduced (P=0.007 for maximal size). This was affiliated to an upregulation of adipocyte differentiation. The underlying mechanism was found in an increased expression of the Notch1 receptor gene and protein in AT of the offsprings independently of the diet. However, feeding a chow diet resulted in a decreased expression of Notch1 target genes Hes1 and RBP-Jκ, whereas under HFD these genes were upregulated. CONCLUSIONS Treatment of mice with recombinant human DLK1 during pregnancy has significant effects on AT of the offspring. This can be associated with counter-regulatory changes in the Notch1 signaling cascade.
Collapse
|
45
|
Wang X, Wang X, Yuan W, Chai R, Liu D. Egfl6 is involved in zebrafish notochord development. FISH PHYSIOLOGY AND BIOCHEMISTRY 2015; 41:961-969. [PMID: 25952972 DOI: 10.1007/s10695-015-0061-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/23/2015] [Indexed: 06/04/2023]
Abstract
The epidermal growth factor (EGF) repeat motif defines a superfamily of diverse protein involved in regulating a variety of cellular and physiological processes, such as cell cycle, cell adhesion, proliferation, migration, and neural development. Egfl6, an EGF protein, also named MAGE was first cloned in human tissue. Up to date, the study of zebrafish Egfl6 expression pattern and functional analysis of Egfl6 involved in embryonic development of vertebrate in vivo is thus far lacking. Here we reported that Egfl6 was involved in zebrafish notochord development. It was shown that Egfl6 mRNA was expressed in zebrafish, developing somites, fin epidermis, pharyngeal arches, and hindbrain region. Particularly the secreted Egfl6 protein was significantly accumulated in notochord. Loss of Egfl6 function in zebrafish embryos resulted in curved body with distorted notochord in the posterior trunk. It was observed that expression of all Notch ligand and receptors in notochord of 28 hpf Egfl6 morphants was not affected, except notch2, which was up-regulated. We found that inhibition of Notch signaling by DAPT efficiently rescued notochord developmental defect of Egfl6 deficiency embryos.
Collapse
Affiliation(s)
- Xueqian Wang
- Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Qixiu Road 19, 226001, Nantong, China
| | | | | | | | | |
Collapse
|
46
|
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: 1.7] [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
|
47
|
Ruhrmann S, Stridh P, Kular L, Jagodic M. Genomic imprinting: A missing piece of the Multiple Sclerosis puzzle? Int J Biochem Cell Biol 2015; 67:49-57. [PMID: 26002250 DOI: 10.1016/j.biocel.2015.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/10/2015] [Accepted: 05/11/2015] [Indexed: 12/14/2022]
Abstract
Evidence for parent-of-origin effects in complex diseases such as Multiple Sclerosis (MS) strongly suggests a role for epigenetic mechanisms in their pathogenesis. In this review, we describe the importance of accounting for parent-of-origin when identifying new risk variants for complex diseases and discuss how genomic imprinting, one of the best-characterized epigenetic mechanisms causing parent-of-origin effects, may impact etiology of complex diseases. While the role of imprinted genes in growth and development is well established, the contribution and molecular mechanisms underlying the impact of genomic imprinting in immune functions and inflammatory diseases are still largely unknown. Here we discuss emerging roles of imprinted genes in the regulation of inflammatory responses with a particular focus on the Dlk1 cluster that has been implicated in etiology of experimental MS-like disease and Type 1 Diabetes. Moreover, we speculate on the potential wider impact of imprinting via the action of imprinted microRNAs, which are abundantly present in the Dlk1 locus and predicted to fine-tune important immune functions. Finally, we reflect on how unrelated imprinted genes or imprinted genes together with non-imprinted genes can interact in so-called imprinted gene networks (IGN) and suggest that IGNs could partly explain observed parent-of-origin effects in complex diseases. Unveiling the mechanisms of parent-of-origin effects is therefore likely to teach us not only about the etiology of complex diseases but also about the unknown roles of this fascinating phenomenon underlying uneven genetic contribution from our parents. This article is part of a Directed Issue entitled: Epigenetics dynamics in development and disease.
Collapse
Affiliation(s)
- Sabrina Ruhrmann
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Pernilla Stridh
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lara Kular
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
48
|
A Monoclonal Antibody to Human DLK1 Reveals Differential Expression in Cancer and Absence in Healthy Tissues. Antibodies (Basel) 2015. [DOI: 10.3390/antib4020071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
49
|
Winter J. MicroRNAs of the miR379-410 cluster: New players in embryonic neurogenesis and regulators of neuronal function. NEUROGENESIS 2015; 2:e1004970. [PMID: 27504472 PMCID: PMC4973610 DOI: 10.1080/23262133.2015.1004970] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 09/26/2014] [Accepted: 01/03/2015] [Indexed: 01/29/2023]
Abstract
The imprinted miR379–410 cluster contains 38 microRNAs (miRNAs) that are involved in diverse neurodevelopmental processes and are important regulators of neuronal function. The implications of these miRNAs in neurological diseases have been recently recognized.In the present minireview, the current findings regarding the brain-specific functions of miR379–410 cluster miRNAs are summarized and discussed.
Collapse
Affiliation(s)
- Jennifer Winter
- Institute of Human Genetics; University Medical Centre Mainz ; Mainz, Germany
| |
Collapse
|
50
|
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: 76] [Impact Index Per Article: 8.4] [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
|