1
|
Chai F, Wang G, Shen Y, Niu Y, Huang Y, Fu T, Yang T, Jiang Y, Zhang J. KGF impedes TRIM21-enhanced stabilization of keratin 10 mediating differentiation in hypopharyngeal cancer. Cell Signal 2025; 127:111614. [PMID: 39848455 DOI: 10.1016/j.cellsig.2025.111614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/03/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
KGF, also known as FGF7, is a member of the fibroblast growth factor (FGF) family that binds with high affinity to the FGF receptor 2b (FGFR2b) and regulates various cellular processes, including cell proliferation and differentiation in a variety of tumors. However, its potential role in hypopharyngeal cancer (HPC) remains largely unknown. In our study, we observed increased expression of FGFR2b in HPC. KGF treatment inhibited the expression of the differentiation marker keratin 10 (K10) protein at the post-transcriptional level in FaDu cells. Furthermore, treatment with the proteasome inhibitor MG132 was found to attenuate KGF-induced K10 reduction, suggesting the involvement of the ubiquitin-proteasome system. Using mass spectrometry and immunoprecipitation analysis, we identified the E3 ubiquitin ligase TRIM21 as a K10-interacting protein. Unexpectedly, instead of causing degradation, TRIM21 enhanced K10 protein stability through K6-linked ubiquitination of K10 at lysine 163 (K163) in the context of KGF exposure. Meanwhile, KGF treatment decreased TRIM21 protein levels, which were regulated by the p38 MAPK pathway, leading to K48-linked ubiquitination-mediated degradation of TRIM21. Notably, TRIM21 knockdown significantly promoted proliferation, inhibited differentiation and migration of FaDu cells, whereas TRIM21 overexpression had opposite effects in vitro and suppressed xenograft tumor growth in vivo. Our study demonstrates that TRIM21 may act as a tumor suppressor in HPC. However, TRIM21 overexpression decreased the sensitivity of FaDu cells to 5-fluorouracil, whereas TRIM21 knockdown or KGF administration significantly increased 5-fluorouracil sensitivity. Taken together, these findings highlight the intricate balance between protein stabilization and degradation orchestrated by KGF. This ubiquitination-mediated non-degradation mechanism of TRIM21 may provide novel therapeutic strategies for HPC and other cancers.
Collapse
Affiliation(s)
- Fangyu Chai
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China
| | - Guangyi Wang
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China
| | - Yibang Shen
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China
| | - Yanfang Niu
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yichuan Huang
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China
| | - Tao Fu
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China
| | - Tao Yang
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Higher Education Key Laboratory of Tumor Immunology & Targeted Drug Development in Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key laboratory of Digestive Disease & Organ Transplantation in Shanxi Province, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| | - Yan Jiang
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China.
| | - Jisheng Zhang
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China.
| |
Collapse
|
2
|
Feng N, Li Y, Guo F, Song J, Wang L, Li M, Gao K, Wang X, Chu D, Song Y, Wang L. Fibroblast growth factor 10 alleviates LPS-induced acute lung injury by promoting recruited macrophage M2 polarization. Inflammation 2024:10.1007/s10753-024-02158-4. [PMID: 39538090 DOI: 10.1007/s10753-024-02158-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/03/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
Acute lung injury (ALI) is characterized by damage to the alveoli and an overabundance of inflammation. Representing a serious inflammatory condition, ALI lacks a precise treatment approach. Despite the recognized benefit impacts of Fibroblast growth factor-10 (FGF10) on ALI, the underlying mechanisms remain unelucidated. To study the role of FGF10 in ALI, C57BL/6 J mice were intratracheally injected with 5 mg/kg Lipopolysaccharide (LPS) with FGF10 (5 mg/kg) or an equal volume of PBS. Inflammatory factors were quantified in bronchoalveolar lavage fluid (BALF) and plasma using ELISA. RNA sequencing of F4/80+Ly6G- macrophages in BALF explored changes in macrophage phenotype and potential mechanisms. Macrophage polarization in BALF was assessed using qRT-PCR, flow cytometry, and Western blot analysis. In vitro, a Transwell co-culture of mouse lung epithelial cells (MLE12) and bone marrow macrophages (BMDM) validated the role of FGF10 in modulating LPS-induced macrophage phenotypic changes. FGF10 ameliorated LPS-induced ALI by diminishing pro-inflammatory factors (IL-1β, TNF-α, and IL-6) and the neutrophil accumulation in BALF. FGF10 also increased the levels of anti-inflammatory factor IL-10. The FGF10 intervention group exhibited enhanced gene expression of macrophage arginine biosynthesis marker (ARG1), and expression of M2-type marker CD206 in monocytes and macrophages. In addition, phosphorylated STAT3 expression increased in isolated monocyte-derived macrophages. Experiments in vitro confirmed that FGF10 could elevate macrophage M2 marker ARG1 expression through the JAK2/STAT3 pathway. FGF10 ameliorates acute LPS-induced lung injury by modulating the polarization of monocyte-derived macrophages recruited in the alveolar space to the M2 type.
Collapse
Affiliation(s)
- Nana Feng
- Department of Respiratory and Critical Medicine, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Yufan Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Fengxia Guo
- Department of Respiratory and Critical Medicine, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Juan Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lu Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Miao Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Kaijing Gao
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaocen Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dejie Chu
- Department of Respiratory and Critical Medicine, Shanghai Eighth People's Hospital, Shanghai, 200235, China.
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 200032, China.
- Shanghai Respiratory Research Institute, Shanghai, 200032, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200032, China.
- Jinshan Hospital of Fudan University, Shanghai, 201508, China.
| | - Linlin Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Chen S, Zhu H, Lin L, Lu L, Chen L, Zeng L, Yue W, Kong X, Zhang H. Apelin-13 improves pulmonary epithelial barrier function in a mouse model of LPS-induced acute lung injury by inhibiting Chk1-mediated DNA damage. Biochem Pharmacol 2024; 226:116297. [PMID: 38801925 DOI: 10.1016/j.bcp.2024.116297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 04/10/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Apelin-13, a type of active peptide, can alleviate lipopolysaccharide (LPS)-induced acute lung injury (ALI). However, the specific mechanism is unclear. Cell cycle checkpoint kinase 1 (Chk1) plays an important role in DNA damage. Here, we investigated the regulatory effect of Apelin on Chk1 in ALI. Chk1-knockout and -overexpression mice were used to explore the role of Chk1 in LPS-induced ALI mice treated with or without Apelin-13. In addition, A549 cells were also treated with LPS to establish a cell model. Chk1 knockdown inhibited the destruction of alveolar structure, the damage of lung epithelial barrier function, and DNA damage in the ALI mouse model. Conversely, Chk1 overexpression had the opposite effect. Furthermore, Apelin-13 reduced Chk1 expression and DNA damage to improve the impaired lung epithelial barrier function in the ALI model. However, the high expression of Chk1 attenuated the protective effect of Apelin-13 on ALI. Notably, Apelin-13 promoted Chk1 degradation through autophagy to regulate DNA damage in LPS-treated A549 cells. In summary, Apelin-13 regulates the expression of Chk1 by promoting autophagy, thereby inhibiting epithelial DNA damage and repairing epithelial barrier function.
Collapse
Affiliation(s)
- Siyue Chen
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China; School of Basic Medical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315302, PR China
| | - Huihui Zhu
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China
| | - Lidan Lin
- School of Basic Medical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315302, PR China
| | - Liling Lu
- Children's Hospital, Zhejiang University School of Medicine, Zhejiang 310000, PR China
| | - Lin Chen
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China; School of Basic Medical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315302, PR China
| | - Luyao Zeng
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China
| | - Wei Yue
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China
| | - Xiaoxia Kong
- School of Basic Medical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315302, PR China.
| | - Hailin Zhang
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China.
| |
Collapse
|
4
|
Yoshioka H, Kagawa K, Minamizaki T, Nakano M, Aubin JE, Kozai K, Tsuga K, Yoshiko Y. Developmental impairments of craniofacial bone and cartilage in transgenic mice expressing FGF10. Bone Rep 2023; 18:101692. [PMID: 37275784 PMCID: PMC10236464 DOI: 10.1016/j.bonr.2023.101692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/07/2023] Open
Abstract
Mutations in a common extracellular domain of fibroblast growth factor receptor (FGFR)-2 isoforms (type IIIb and IIIc) cause craniosynostosis syndrome and chondrodysplasia syndrome. FGF10, a major ligand for FGFR2-IIIb and FGFR1-IIIb, is a key participant in the epithelial-mesenchymal interactions required for morphogenetic events. FGF10 also regulates preadipocyte differentiation and early chondrogenesis in vitro, suggesting that FGF10-FGFR signaling may be involved in craniofacial skeletogenesis in vivo. To test this hypothesis, we used a tet-on doxycycline-inducible transgenic mouse model (FGF10 Tg) to overexpress Fgf10 from embryonic day 12.5. Fgf10 expression was 73.3-fold higher in FGF10 Tg than in wild-type mice. FGF10 Tg mice exhibited craniofacial anomalies, such as a short rostrum and mandible, an underdeveloped (cleft) palate, and no tympanic ring. Opposite effects on chondrogenesis in different anatomical regions were seen, e.g., hyperplasia in the nasal septum and hypoplasia in the mandibular condyle. We found an alternative splicing variant of Fgfr2-IIIb with a predicted translation product lacking the transmembrane domain, and suggesting a soluble form of FGFR2-IIIb (sFGFR2-IIIb), differentially expressed in some of the craniofacial bones and cartilages. Thus, excessive FGF10 may perturb signal transduction of the FGF-FGFR, leading to craniofacial skeletal abnormalities in FGF10 Tg mice.
Collapse
Affiliation(s)
- Hirotaka Yoshioka
- Department of Anatomy, School of Medicine, International University of Health and Welfare, Chiba, Japan
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuko Kagawa
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoko Minamizaki
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masashi Nakano
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Pediatric Dentistry, Division of Oral Health and Development, Hiroshima University Hospital, Hiroshima, Japan
| | - Jane E. Aubin
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Katsuyuki Kozai
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhiro Tsuga
- Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuji Yoshiko
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
5
|
Guo K, Wang L, Zhong Y, Gao S, Jing R, Ye J, Zhang K, Fu M, Hu Z, Zhao W, Xu N. Cucurbitacin promotes hair growth in mice by inhibiting the expression of fibroblast growth factor 18. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1104. [PMID: 36388783 PMCID: PMC9652544 DOI: 10.21037/atm-22-4423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/08/2022] [Indexed: 08/03/2023]
Abstract
BACKGROUND The inhibition of fibroblast growth factor 18 (FGF18) promotes the transition of hair follicles (HFs) from the telogen phase to the anagen phase. Cucurbitacin has been shown to have a good effect in promoting hair cell growth. This study explored the potential effect of cucurbitacin on hair growth and its effect on FGF18 expression in mice. METHODS Male C57BL/6J mice were randomly divided into the following two groups: (I) the vehicle group; and (II) the cucurbitacin group. Matrix cream and cucurbitacin cream were applied to the depilated skin on the back of the vehicle group mice and the cucurbitacin group mice, respectively. On days 3, 6, 9, 12, 15, and 18, the hair growth in the depilated dorsal skin of the mice was recorded with a digital camera and a HF detector, and the HF cycle status of the mice was observed by hematoxylin and eosin (H&E) staining. In addition, the level of FGF18 messenger ribonucleic acid (mRNA) in the dorsal skin was measured on days 15 and 18 by quantitative real-time polymerase chain reaction (qRT-PCR), while the level of FGF18 protein was measured by western blot and immunofluorescence staining. RESULTS The dorsal skin to which the cucurbitacin cream was applied began to darken on day 6 and grew hairs on day 9, which was 3 days earlier than the dorsal skin to which the matrix cream was applied. The H&E staining revealed a transition from the telogen phase to the anagen phase 3 days earlier for the cucurbitacin cream-treated skin than the matrix cream-treated skin. In addition, the skin treated with cucurbitacin cream also showed a significant decrease in FGF18 mRNA as seen by qRT-PCR, and reduced FGF18 protein levels as detected by western blot and immunofluorescence staining compared to the skin treated with matrix cream only. CONCLUSIONS Cucurbitacin significantly reduced the levels of FGF18 mRNA and protein in the dorsal skin of mice to accelerate the HFs to enter the anagen phase earlier, thereby promoting the regeneration of hair. Thus, cucurbitacin can be considered a new and valuable agent for the development of anti-hair loss products.
Collapse
Affiliation(s)
- Keke Guo
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Lusheng Wang
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Yulan Zhong
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Shuang Gao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Rongrong Jing
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Jiabin Ye
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Kaini Zhang
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Mengli Fu
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Zhenlin Hu
- School of Medicine, Shanghai University, Shanghai, China
| | - Wengang Zhao
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Nuo Xu
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| |
Collapse
|
6
|
Peak KE, Mohr-Allen SR, Gleghorn JP, Varner VD. Focal sources of FGF-10 promote the buckling morphogenesis of the embryonic airway epithelium. Biol Open 2022; 11:bio059436. [PMID: 35979841 PMCID: PMC9536751 DOI: 10.1242/bio.059436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/11/2022] [Indexed: 12/01/2022] Open
Abstract
During airway branching morphogenesis, focal regions of FGF-10 expression in the pulmonary mesenchyme are thought to provide a local guidance cue, which promotes chemotactically the directional outgrowth of the airway epithelium. Here, however, we show that an ectopic source of FGF-10 induces epithelial buckling morphogenesis and the formation of multiple new supernumerary buds. FGF-10-induced budding can be modulated by altered epithelial tension and luminal fluid pressure. Increased tension suppresses the formation of ectopic branches, while a collapse of the embryonic airway promotes more expansive buckling and additional FGF-10-induced supernumerary buds. Our results indicate that a focal source of FGF-10 can promote epithelial buckling and suggest that the overall branching pattern cannot be explained entirely by the templated expression of FGF-10. Both FGF-10-mediated cell behaviors and exogenous mechanical forces must be integrated to properly shape the bronchial tree.
Collapse
Affiliation(s)
- Kara E Peak
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shelby R Mohr-Allen
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Victor D Varner
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
7
|
Deuper L, Meuser M, Thiesler H, Jany UWH, Rudat C, Hildebrandt H, Trowe MO, Kispert A. Mesenchymal FGFR1 and FGFR2 control patterning of the ureteric mesenchyme by balancing SHH and BMP4 signaling. Development 2022; 149:276592. [DOI: 10.1242/dev.200767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 08/19/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The coordinated development of the mesenchymal and epithelial progenitors of the murine ureter depends on a complex interplay of diverse signaling activities. We have recently shown that epithelial FGFR2 signaling regulates stratification and differentiation of the epithelial compartment by enhancing epithelial Shh expression, and mesenchymal SHH and BMP4 activity. Here, we show that FGFR1 and FGFR2 expression in the mesenchymal primordium impinges on the SHH/BMP4 signaling axis to regulate mesenchymal patterning and differentiation. Mouse embryos with conditional loss of Fgfr1 and Fgfr2 in the ureteric mesenchyme exhibited reduced mesenchymal proliferation and prematurely activated lamina propria formation at the expense of the smooth muscle cell program. They also manifested hydroureter at birth. Molecular profiling detected increased SHH, WNT and retinoic acid signaling, whereas BMP4 signaling in the mesenchyme was reduced. Pharmacological activation of SHH signaling in combination with inhibition of BMP4 signaling recapitulated the cellular changes in explant cultures of wild-type ureters. Additional experiments suggest that mesenchymal FGFR1 and FGFR2 act as a sink for FGF ligands to dampen activation of Shh and BMP receptor gene expression by epithelial FGFR2 signaling.
Collapse
Affiliation(s)
- Lena Deuper
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Max Meuser
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover 2 , 30625 Hannover , Germany
| | - Ulrich W. H. Jany
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Carsten Rudat
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover 2 , 30625 Hannover , Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| |
Collapse
|
8
|
Willie D, Holmes G, Jabs EW, Wu M. Cleft Palate in Apert Syndrome. J Dev Biol 2022; 10:jdb10030033. [PMID: 35997397 PMCID: PMC9397066 DOI: 10.3390/jdb10030033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/06/2022] [Accepted: 08/07/2022] [Indexed: 11/17/2022] Open
Abstract
Apert syndrome is a rare genetic disorder characterized by craniosynostosis, midface retrusion, and limb anomalies. Cleft palate occurs in a subset of Apert syndrome patients. Although the genetic causes underlying Apert syndrome have been identified, the downstream signaling pathways and cellular mechanisms responsible for cleft palate are still elusive. To find clues for the pathogenic mechanisms of palatal defects in Apert syndrome, we review the clinical characteristics of the palate in cases of Apert syndrome, the palatal phenotypes in mouse models, and the potential signaling mechanisms involved in palatal defects. In Apert syndrome patients, cleft of the soft palate is more frequent than of the hard palate. The length of the hard palate is decreased. Cleft palate is associated most commonly with the S252W variant of FGFR2. In addition to cleft palate, high-arched palate, lateral palatal swelling, or bifid uvula are common in Apert syndrome patients. Mouse models of Apert syndrome display palatal defects, providing valuable tools to understand the underlying mechanisms. The mutations in FGFR2 causing Apert syndrome may change a signaling network in epithelial–mesenchymal interactions during palatogenesis. Understanding the pathogenic mechanisms of palatal defects in Apert syndrome may shed light on potential novel therapeutic solutions.
Collapse
|
9
|
Yeganeh PM, Tahmasebi S, Esmaeilzadeh A. Cellular and biological factors involved in healing wounds and burns and treatment options in tissue engineering. Regen Med 2022; 17:401-418. [PMID: 35545963 DOI: 10.2217/rme-2022-0029] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Severe traumatic wounds and burns have a high chance of mortality and can leave survivors with many functional disabilities and cosmetic problems, including scars. The healing process requires a harmonious interplay of various cells and growth factors. Different structures of the skin house numerous cells, matrix components and growth factors. Any disturbance in the balance between these components can impair the healing process. The function of cells and growth factors can be manipulated and facilitated to aid tissue repair. In the current review, the authors focus on the importance of the skin microenvironment, the pathophysiology of various types of burns, mechanisms and factors involved in skin repair and wound healing and regeneration of the skin using tissue engineering approaches.
Collapse
Affiliation(s)
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolreza Esmaeilzadeh
- Department of immunology, School of Medicine, Zanjan University of Medical Science, Zanjan, 4513956111, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Science, Zanjan, Iran
| |
Collapse
|
10
|
Karasawa Y, Shinomiya N, Takeuchi M, Ito M. Growth factor dependence of the proliferation and survival of cultured lacrimal gland epithelial cells isolated from late-embryonic mice. Dev Growth Differ 2022; 64:138-149. [PMID: 35149991 DOI: 10.1111/dgd.12776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 12/03/2021] [Accepted: 12/11/2021] [Indexed: 11/30/2022]
Abstract
Epidermal growth factor (EGF) and hepatocyte growth factor (HGF) regulate the growth and morphogenesis of various exocrine glands with branched morphologies. Their roles in lacrimal gland (LG) development remain unknown, but fibroblast growth factor (FGF) 10 is crucial for early LG organogenesis. To clarify the roles of EGF, HGF, and FGF10 in LG development, LG epithelial cells were isolated from late-embryonic and neonatal mice; cultured; and treated with EGF, HGF, or FGF10 and their respective receptor tyrosine kinase (RTK) inhibitors AG1478, PHA665752, or SU5402. EGF and HGF increased the number of viable cells by enhancing DNA synthesis, FGF10 and SU5402 showed no such effect, and RTK inhibitors exhibited the opposite effect. EGF and HGF receptors were immunostained in cultured late-embryonic LG epithelial cells and terminal LG acini from late embryos and adult mice. HGF was detected in neonatal LG epithelial cell culture supernatants by western blotting. In the absence of EGF and HGF RTK inhibitors, growth factor addition increased the number of viable cells and suppressed cell death. However, when one RTK was inhibited and a growth factor targeting an intact RTK was added, the number of dead cells increased as the number of viable cells increased. No cells survived when both RTKs were inhibited. In explant cultures of LGs from embryos, AG1478 or PHA665752 decreased the number of Ki67-positive proliferating epithelial cells in terminal acini. Thus, EGF and HGF may function in a cooperative autocrine manner, supporting cell proliferation and survival during LG development in late-embryonic and neonatal mice.
Collapse
Affiliation(s)
- Yoko Karasawa
- Department of Ophthalmology, National Defense Medical College, Saitama, Japan
| | | | - Masaru Takeuchi
- Department of Ophthalmology, National Defense Medical College, Saitama, Japan
| | - Masataka Ito
- Department of Developmental Anatomy and Regenerative Biology, National Defense Medical College, Saitama, Japan
| |
Collapse
|
11
|
Meuser M, Deuper L, Rudat C, Aydoğdu N, Thiesler H, Zarnovican P, Hildebrandt H, Trowe MO, Kispert A. FGFR2 signaling enhances the SHH-BMP4 signaling axis in early ureter development. Development 2022; 149:273983. [DOI: 10.1242/dev.200021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/09/2021] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The patterned array of basal, intermediate and superficial cells in the urothelium of the mature ureter arises from uncommitted epithelial progenitors of the distal ureteric bud. Urothelial development requires signaling input from surrounding mesenchymal cells, which, in turn, depend on cues from the epithelial primordium to form a layered fibro-muscular wall. Here, we have identified FGFR2 as a crucial component in this reciprocal signaling crosstalk in the murine ureter. Loss of Fgfr2 in the ureteric epithelium led to reduced proliferation, stratification, intermediate and basal cell differentiation in this tissue, and affected cell survival and smooth muscle cell differentiation in the surrounding mesenchyme. Loss of Fgfr2 impacted negatively on epithelial expression of Shh and its mesenchymal effector gene Bmp4. Activation of SHH or BMP4 signaling largely rescued the cellular defects of mutant ureters in explant cultures. Conversely, inhibition of SHH or BMP signaling in wild-type ureters recapitulated the mutant phenotype in a dose-dependent manner. Our study suggests that FGF signals from the mesenchyme enhance, via epithelial FGFR2, the SHH-BMP4 signaling axis to drive urothelial and mesenchymal development in the early ureter.
Collapse
Affiliation(s)
- Max Meuser
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Lena Deuper
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Carsten Rudat
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Nurullah Aydoğdu
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Patricia Zarnovican
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| |
Collapse
|
12
|
Stenhouse C, Seo H, Wu G, Johnson GA, Bazer FW. Insights into the Regulation of Implantation and Placentation in Humans, Rodents, Sheep, and Pigs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1354:25-48. [PMID: 34807435 DOI: 10.1007/978-3-030-85686-1_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Precise cell-specific spatio-temporal molecular signaling cascades regulate the establishment and maintenance of pregnancy. Importantly, the mechanisms regulating uterine receptivity, conceptus apposition and adhesion to the uterine luminal epithelia/superficial glandular epithelia and, in some species, invasion into the endometrial stroma and decidualization of stromal cells, are critical prerequisite events for placentation which is essential for the appropriate regulation of feto-placental growth for the remainder of pregnancy. Dysregulation of these signaling cascades during this critical stage of pregnancy can lead to pregnancy loss, impaired growth and development of the conceptus, and alterations in the transplacental exchange of gasses and nutrients. While many of these processes are conserved across species, significant variations in the molecular mechanisms governing maternal recognition of pregnancy, conceptus implantation, and placentation exist. This review addresses the complexity of key mechanisms that are critical for the establishment and maintenance of a successful pregnancy in humans, rodents, sheep, and pigs. Improving understanding of the molecular mechanisms governing these processes is critical to enhancing the fertility and reproductive health of humans and livestock species.
Collapse
Affiliation(s)
- Claire Stenhouse
- Department of Animal Science and Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Heewon Seo
- Department of Animal Science and Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Guoyao Wu
- Department of Animal Science and Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Gregory A Johnson
- Department of Animal Science and Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Fuller W Bazer
- Department of Animal Science and Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
13
|
Guadagnin AR, Velasco-Acosta DA, Stella SL, Luchini D, Cardoso FC. Methionine supply during the peripartum period and early lactation alter immunometabolic gene expression in cytological smear and endometrial tissue of holstein cows. Theriogenology 2021; 173:102-111. [PMID: 34365138 DOI: 10.1016/j.theriogenology.2021.07.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
The objective of the present study was to evaluate the effect of feeding rumen-protected methionine (RPM) during the peripartal period and early lactation on mRNA gene expression profiles of uterine cytological smear and endometrial samples of Holstein cows (n = 20). Treatments consisted of a supplementation with RPM [MET; n = 11; RPM at a rate of 0.08 % of DM: Lys:Met = 2.8:1, (Smartamine® M Adisseo, Alpharetta, GA, USA)] and no supplementation (CON; n = 9; Lys:Met = 3.5:1). Uterine cytology smears and endometrial samples were collected at 15, 30, and 73 days in milk (DIM) and analyzed for expression of genes related with metabolism, inflammation, and methionine metabolism. Regarding the cytological smear samples, RPM supplementation tended to increase mRNA expression of methionine adenosyltransferase 1 alpha (MAT1A) and increased the mRNA expression of fibroblast growth factor 7 (FGF7), with an effect of time for the latter. On the other hand, RPM decreased mRNA expression for glucose transporter 4 (GLUT4), interleukin 1 beta (IL-1β), interleukin 6 (IL-6), interleukin 8 (IL-8), prostaglandin E synthase 3 (PTGES3), translocator protein 18 kDa (TSPO), mucin 1 (MUC1) and superoxide dismutase (SOD1) in cytological smear samples. There was an effect of time for all variables except MAT1A, with decreasing expression over time. There was a TRT × TIME interaction for GLUT4 mRNA expression, with higher GLUT4 mRNA expression for cows fed CON than for cows fed RPM at time 15 and a tendency to higher expression for cows fed CON on time 30 when compared with cows fed RPM. For uterine tissue samples, feeding RPM increased the mRNA expression of lecithin-cholesterol acyltransferase (LCAT), S-adenosyl-l-homocysteine hydrolase (SAAH), FGF7, GLUT4, and apolipoproteins 3 (APOL3), with an effect of time for APOL3 where its expression increased over time. There was a tendency for cows fed RPM to have decreased IL1β mRNA expression. In conclusion, feeding RPM during transition period and early lactation is beneficial for uterine immune response and metabolism in early lactation as indicated by the favorable expressions of genes affecting the uterine immunometabolism during such a challenging period.
Collapse
Affiliation(s)
- A R Guadagnin
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | - D A Velasco-Acosta
- The Colombian Corporation for Agricultural Research (AGROSAVIA), Mosquera, Colombia
| | - S L Stella
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | | | - F C Cardoso
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA.
| |
Collapse
|
14
|
Eguchi A, Ueki A, Hoshiyama J, Kuwata K, Chikaoka Y, Kawamura T, Nagatoishi S, Tsumoto K, Ueki R, Sando S. A DNA Aptamer That Inhibits the Aberrant Signaling of Fibroblast Growth Factor Receptor in Cancer Cells. JACS AU 2021; 1:578-585. [PMID: 34467321 PMCID: PMC8395645 DOI: 10.1021/jacsau.0c00121] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Indexed: 06/13/2023]
Abstract
Growth factor receptors are activated through dimerization by the binding of their ligands and play pivotal roles in normal cell function. However, the aberrant activity of the receptors has been associated with cancer malignancy. One of the main causes of the aberrant receptor activation is the overexpression of receptors and the resultant formation of unliganded receptor dimers, which can be activated in the absence of external ligand molecules. Thus, the unliganded receptor dimer is a promising target to inhibit aberrant signaling in cancer. Here, we report an aptamer that specifically binds to fibroblast growth factor receptor 2b and inhibits the aberrant receptor activation and signaling. Our investigation suggests that this aptamer inhibits the formation of the receptor dimer occurring in the absence of external ligand molecules. This work presents a new inhibitory function of aptamers and the possibility of oligonucleotide-based therapeutics for cancer.
Collapse
Affiliation(s)
- Akihiro Eguchi
- Department of Chemistry and Biotechnology andDepartment of Bioengineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Ayaka Ueki
- Department of Chemistry and Biotechnology andDepartment of Bioengineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Junya Hoshiyama
- Department of Chemistry and Biotechnology andDepartment of Bioengineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Keiko Kuwata
- Institute
of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8601, Japan
| | - Yoko Chikaoka
- Proteomics
Laboratory, Isotope Science Center, The
University of Tokyo, 2-11-16, Yayoi, Bunkyo-Ku, Tokyo 113-0032, Japan
| | - Takeshi Kawamura
- Proteomics
Laboratory, Isotope Science Center, The
University of Tokyo, 2-11-16, Yayoi, Bunkyo-Ku, Tokyo 113-0032, Japan
| | - Satoru Nagatoishi
- The
Institute of Medical Science, The University
of Tokyo, 4-6-1 Shirokanedai,
Minato-ku, Tokyo 108-8639, Japan
| | - Kouhei Tsumoto
- Department of Chemistry and Biotechnology andDepartment of Bioengineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- The
Institute of Medical Science, The University
of Tokyo, 4-6-1 Shirokanedai,
Minato-ku, Tokyo 108-8639, Japan
| | - Ryosuke Ueki
- Department of Chemistry and Biotechnology andDepartment of Bioengineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology andDepartment of Bioengineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
15
|
Tomita H, Tanaka K, Hirata A, Okada H, Imai H, Shirakami Y, Ohnishi K, Sugie S, Aoki H, Hatano Y, Noguchi K, Kanayama T, Niwa A, Suzui N, Miyazaki T, Tanaka T, Akiyama H, Shimizu M, Yoshida K, Hara A. Inhibition of FGF10-ERK signal activation suppresses intraductal papillary neoplasm of the bile duct and its associated carcinomas. Cell Rep 2021; 34:108772. [PMID: 33626352 DOI: 10.1016/j.celrep.2021.108772] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 12/31/2020] [Accepted: 01/28/2021] [Indexed: 02/07/2023] Open
Abstract
Evidence regarding intraductal papillary neoplasm of the bile duct (IPNB) as a type of precancerous lesion of cholangiocarcinoma is limited. Moreover, a reproducible in vivo model is lacking, and IPNB pathogenesis remains unclear. Here, we use a doxycycline-inducible tetracycline (Tet)-on mice model to control fibroblast growth factor 10 (FGF10) expression, which regulates branching and tubule formation. FGF10-induced IPNB mimics the multifocal and divergent human IPNB phenotypes via the FGF10-FGF receptor 2 (FGFR2)-RAS-extracellular-signal-regulated kinase (ERK) signaling pathway. A paracrine/autocrine growth factor is sufficient to initiate and maintain IPNB originating from the peribiliary glands, including biliary stem/progenitor cells. With KrasG12D, p53, or p16 mutations or both, Fgf10-induced IPNB shows stepwise carcinogenesis, causing associated invasive carcinoma. Fgf10-induced papillary changes and progression are suppressed by the inhibition of the FGF10-FGFR2-RAS-ERK signaling pathway, demonstrating that the signal is a therapeutic target for IPNB and associated carcinoma.
Collapse
Affiliation(s)
- Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan.
| | - Kaori Tanaka
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Surgical Oncology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Akihiro Hirata
- Division of Animal Experiment, Life Science Research Center, Gifu University, Gifu 501-1194, Japan
| | - Hideshi Okada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Hisashi Imai
- Department of Surgical Oncology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Yohei Shirakami
- Department of Gastroenterology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Kotaro Ohnishi
- Department of Gastroenterology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Shigeyuki Sugie
- Department of Pathology, Asahi University Hospital, Gifu 500-8523, Japan
| | - Hitomi Aoki
- Department of Tissue and Organ Development, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Yuichiro Hatano
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Kei Noguchi
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Tomohiro Kanayama
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Ayumi Niwa
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Natsuko Suzui
- Department of Pathology, Gifu University Hospital, Gifu 501-1194, Japan
| | | | - Takuji Tanaka
- Department of Diagnostic Pathology (DDP) and Research Center of Diagnostic Pathology (RC-DiP), Gifu Municipal Hospital, Gifu 500-8513, Japan
| | - Haruhiko Akiyama
- Department of Orthopedic Surgery, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Masahito Shimizu
- Department of Gastroenterology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Kazuhiro Yoshida
- Department of Surgical Oncology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| |
Collapse
|
16
|
Wang X, Wang S, Guo B, Su Y, Tan Z, Chang M, Diao J, Zhao Y, Wang Y. Human primary epidermal organoids enable modeling of dermatophyte infections. Cell Death Dis 2021; 12:35. [PMID: 33414472 PMCID: PMC7790817 DOI: 10.1038/s41419-020-03330-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/15/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022]
Abstract
Technology of generating human epidermal derivatives with physiological relevance to in vivo epidermis is continuously investigated for improving their effects on modeling of human natural dermatological status in basic and clinical studies. Here, we report a method of robust establishment and expansion of human primary epidermal organoids (hPEOs) under a chemically defined condition. hPEOs reconstruct morphological, molecular, and functional features of human epidermis and can expand for 6 weeks. Remarkably, hPEOs are permissive for dermatophyte infections caused by Trichophyton Rubrum (T. rubrum). The T. rubrum infections on hPEOs reflect many aspects of known clinical pathological reactions and reveal that the repression on IL-1 signaling may contribute to chronic and recurrent infections with the slight inflammation caused by T. rubrum in human skin. Thus, our present study provides a new insight into the pathogenesis of T. rubrum infections and indicates that hPEOs are a potential ex vivo model for both basic studies of skin diseases and clinical studies of testing potential antifungal drugs.
Collapse
Affiliation(s)
- Xuan Wang
- Translational Medicine Research Center, Beijing Tsinghua Chang Gung Hospital, Beijing, 102218, China
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Shuyong Wang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, the 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Baolin Guo
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Yuxin Su
- Translational Medicine Research Center, Beijing Tsinghua Chang Gung Hospital, Beijing, 102218, China
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Zuolong Tan
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Mingyang Chang
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Jinmei Diao
- Translational Medicine Research Center, Beijing Tsinghua Chang Gung Hospital, Beijing, 102218, China
| | - Yi Zhao
- Department of Dermatology, Beijing Tsinghua Chang Gung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Yunfang Wang
- Translational Medicine Research Center, Beijing Tsinghua Chang Gung Hospital, Beijing, 102218, China.
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.
| |
Collapse
|
17
|
Walker H, Akula M, West-Mays JA. Corneal development: Role of the periocular mesenchyme and bi-directional signaling. Exp Eye Res 2020; 201:108231. [PMID: 33039457 DOI: 10.1016/j.exer.2020.108231] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/04/2020] [Accepted: 09/05/2020] [Indexed: 01/08/2023]
Abstract
The cornea is a highly specialized transparent tissue located at the anterior most surface of the eye. It consists of three main layers, the outer stratified squamous epithelium, the inner endothelium, and the intermediate stroma. Formation of these layers during development involves a complex interaction between ectodermal-derived structures, such as the overlying head ectoderm with the periocular mesenchyme (POM), the latter of which is comprised of neural crest cells (NCC) and mesoderm-derived progenitor cells. Regulation of corneal epithelial development, including both epithelial cell fate and stratification, has been shown to depend on numerous bi-directional mesenchymal-epithelial signaling pathways. In this review we pay particular attention to the genes and signaling pathways that involve the POM.
Collapse
Affiliation(s)
- Haydn Walker
- McMaster University, Health Sciences Centre, 1280 Main St. W., L8S 4L8, Hamilton, ON, Canada
| | - Monica Akula
- McMaster University, Health Sciences Centre, 1280 Main St. W., L8S 4L8, Hamilton, ON, Canada
| | - Judith A West-Mays
- McMaster University, Health Sciences Centre, 1280 Main St. W., L8S 4L8, Hamilton, ON, Canada.
| |
Collapse
|
18
|
Jimi S, Jaguparov A, Nurkesh A, Sultankulov B, Saparov A. Sequential Delivery of Cryogel Released Growth Factors and Cytokines Accelerates Wound Healing and Improves Tissue Regeneration. Front Bioeng Biotechnol 2020; 8:345. [PMID: 32426341 PMCID: PMC7212449 DOI: 10.3389/fbioe.2020.00345] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Growth factors and cytokines that are secreted by cells play a crucial role in the complex physiological reaction to tissue injury. The ability to spatially and temporally control their actions to maximize regenerative benefits and minimize side effects will help accelerate wound healing and improve tissue regeneration. In this study, the sequential targeted delivery of growth factor/cytokine combinations with regulatory functions on inflammation and tissue regeneration was examined using an internal splint wound healing model. Four examined growth factors and cytokines were effectively incorporated into a novel chitosan-based cryogel, which offered a controlled and sustained release of all factors while maintaining their biological activities. The cryogels incorporated with inflammation modulatory factors (IL-10 and TGF-β) and with wound healing factors (VEGF and FGF) were placed on the wound surface on day 0 and day 3, respectively, after wound initiation. Although wound area gradually decreased in all groups over time, the area in the cryogel group with growth factor/cytokine combinations was significantly reduced starting on day 7 and reached about 10% on day 10, as compared to 60-65% in the control groups. Sequential delivery of inflammation modulatory and wound healing factors enhanced granulation tissue formation, as well as functional neovascularization, leading to regenerative epithelialization. Collectively, the chitosan-based cryogel can serve as a controlled release system for sequential delivery of several growth factors and cytokines to accelerate tissue repair and regeneration.
Collapse
Affiliation(s)
- Shiro Jimi
- Central Laboratory for Pathology and Morphology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Alexandr Jaguparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Ayan Nurkesh
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Bolat Sultankulov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
19
|
Jones MLM, Sarila G, Chapuis P, Hutson JM, King SK, Teague WJ. The Role of Fibroblast Growth Factor 10 Signaling in Duodenal Atresia. Front Pharmacol 2020; 11:250. [PMID: 32210824 PMCID: PMC7076179 DOI: 10.3389/fphar.2020.00250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/24/2020] [Indexed: 11/24/2022] Open
Abstract
Introduction Duodenal atresia (DA) is a congenital bowel obstruction requiring major surgery in the first week of life. Three morphological phenotypes are described, reflecting increasing degrees of obstruction and discontinuity of the duodenum. The cause of DA is not known. Tandler’s original “solid cord” hypothesis conflicts with recent biological evidence, and is unable to account for differing DA types. In humans, a genetic etiology is supported by the association between Trisomy 21 and DA, and reports of familial inheritance patterns. Interruption of FGF10/FGFR2b signaling is the best demonstrated genetic link to DA in mice, with 35–75% of homozygous knockout embryos developing DA. Purpose This review examines the current evidence surrounding the etiology of DA. We focus on research regarding FGF10/FGFR2b signaling and its role in duodenal and other intestinal atresia. Further, we outline planned future research in this area, that we consider necessary to validate and better understand this murine model in order to successfully translate this research into clinical practice. Conclusion Determining the etiology of DA in humans is a clinical and scientific imperative. Fgf10/Fgfr2b murine models represent current science’s best key to unlocking this mystery. However, further research is required to understand the complex role of FGF10/FGFR2b signaling in DA development. Such complexity is expected, given the lethality of their associated defects makes ubiquitous interruption of either Fgf10 or Fgfr2b genes an unlikely cause of DA in humans. Rather, local or tissue-specific mutation in Fgf10, Fgfr2b, or their downstream targets, is the hypothesized basis of DA etiology.
Collapse
Affiliation(s)
- Matthew L M Jones
- F. Douglas Stephens Surgical Research Laboratory, Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Discipline of Surgery, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Department of Paediatric Surgery, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Gulcan Sarila
- F. Douglas Stephens Surgical Research Laboratory, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Pierre Chapuis
- Discipline of Surgery, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - John M Hutson
- F. Douglas Stephens Surgical Research Laboratory, Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia.,Department of Urology, The Royal Children's Hospital Melbourne, Melbourne, VIC, Australia
| | - Sebastian K King
- F. Douglas Stephens Surgical Research Laboratory, Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Discipline of Surgery, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Department of Paediatric Surgery, The Royal Children's Hospital, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Warwick J Teague
- F. Douglas Stephens Surgical Research Laboratory, Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Discipline of Surgery, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Department of Paediatric Surgery, The Royal Children's Hospital, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
20
|
Samuel RZ, Lei P, Nam K, Baker OJ, Andreadis ST. Engineering the mode of morphogenetic signal presentation to promote branching from salivary gland spheroids in 3D hydrogels. Acta Biomater 2020; 105:121-130. [PMID: 31988042 DOI: 10.1016/j.actbio.2020.01.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/24/2022]
Abstract
Previously we developed a fibrin hydrogel (FH) decorated with laminin-111 peptides (L1p-FH) and supports three-dimensional (3D) gland microstructures containing polarized acinar cells. Here we expand on these results and show that co-culture of rat parotid Par-C10 cells with mesenchymal stem cells produces migrating branches of gland cells into the L1p-FH and we identify FGF-7 as the principal morphogenetic signal responsible for branching. On the other hand, another FGF family member and gland morphogen, FGF-10 increased proliferation but did not promote migration and therefore, limited the number and length of branched structures grown into the gel. By controlling the mode of growth factor presentation and delivery, we can control the length and cellularity of branches as well as formation of new nodes/clusters within the hydrogel. Such spatial delivery of two or more morphogens may facilitate engineering of anatomically complex tissues/mini organs such as salivary glands that can be used to address developmental questions or as platforms for drug discovery. STATEMENT OF SIGNIFICANCE: Hyposalivation leads to the development of a host of oral diseases. Current treatments only provide temporary relief. Tissue engineering may provide promising permanent solutions. Yet current models are limited to salivary spheroids with no branching networks. Branching structures are vital to an effective functioning gland as they increase the surface area/glandular volume ratio of the tissue, allowing a higher output from the small-sized gland. We describe a strategy that controls branch network formation in salivary glands that is a key in advancing the field of salivary gland tissue engineering.
Collapse
|
21
|
Polymorphisms in FGF3, FGF10, and FGF13 May Contribute to the Presence of Temporomandibular Disorders in Patients Who Required Orthognathic Surgery. J Craniofac Surg 2019; 30:2082-2084. [DOI: 10.1097/scs.0000000000006029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
22
|
Farahat M, Kazi GAS, Taketa H, Hara ES, Oshima M, Kuboki T, Matsumoto T. Fibronectin-induced ductal formation in salivary gland self-organization model. Dev Dyn 2019; 248:813-825. [PMID: 31237723 DOI: 10.1002/dvdy.78] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Recent advances in tissue regeneration approaches including 3D organoids, were based on various 3D organogenesis models. However, 3D models are generally technique-sensitive and time-consuming. Thus, we utilized an existing model of submandibular salivary gland (SMG) to modify a simple and highly reproducible in vitro 3D culture model of primary SMG cells self-organization into a well-developed cell spheroid inside Matrigel substrate. We used this model to observe the collective multicellular behavior during spheroid formation. Further, we applied various quantitative approaches including real-time live imaging and immune histochemical image analysis to dissect the cellular dynamics during tissue patterning. RESULTS On a time-scale of hours, we observed marked size and shape transformations in the developed 3D spheroid which resulted in a spatially-controlled growth differential from the canter to the periphery of the formed aggregates. Moreover, we investigated the effect of fibronectin (FN) on SMG cells self-organization using our simplified culture model. Interestingly, we discovered a novel role of FN in inducing duct-like elongation during initial stages of SMG bud formation. CONCLUSION This in vitro model provides an excellent tool for analyzing the intercellular dynamics during early SMG tissue development as well as revealing a novel role of FN in SMG ductal expansion.
Collapse
Affiliation(s)
- Mahmoud Farahat
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Gulsan A S Kazi
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroaki Taketa
- Center for the Development of Medical and Health Care Education, Okayama University, Okayama, Japan
| | - Emilio S Hara
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masamitsu Oshima
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuya Matsumoto
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
23
|
Thotakura S, Basova L, Makarenkova HP. FGF Gradient Controls Boundary Position Between Proliferating and Differentiating Cells and Regulates Lacrimal Gland Growth Dynamics. Front Genet 2019; 10:362. [PMID: 31191595 PMCID: PMC6546953 DOI: 10.3389/fgene.2019.00362] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/04/2019] [Indexed: 12/17/2022] Open
Abstract
Fibroblast growth factor (FGF) signaling plays an important role in controlling cell proliferation, survival, and cell movements during branching morphogenesis of many organs. In mammals branching morphogenesis is primarily regulated by members of the FGF7-subfamily (FGF7 and FGF10), which are expressed in the mesenchyme, and signal to the epithelial cells through the “b” isoform of fibroblast growth factor receptor-2 (FGFR2). Our previous work demonstrated that FGF7 and FGF10 form different gradients in the extracellular matrix (ECM) and induce distinct cellular responses and gene expression profiles in the lacrimal and submandibular glands. The last finding was the most surprising since both FGF7 and FGF10 bind signal most strongly through the same fibroblast growth factor receptor-2b isoform (FGFR2b). Here we revisit this question to gain an explanation of how the different FGFs regulate gene expression. For this purpose, we employed our ex vivo epithelial explant migration assay in which isolated epithelial explants are grown near the FGF loaded beads. We demonstrate that the graded distribution of FGF induces activation of ERK1/2 MAP kinases that define the position of the boundary between proliferating “bud” and differentiating “stalk” cells of growing lacrimal gland epithelium. Moreover, we showed that gene expression profiles of the epithelial explants exposed to distinct FGFs strictly depend on the ratio between “bud” and “stalk” area. Our data also suggests that differentiation of “stalk” and “bud” regions within the epithelial explants is necessary for directional and persistent epithelial migration. Gaining a better understanding of FGF functions is important for development of new approaches to enhance tissue regeneration.
Collapse
Affiliation(s)
- Suharika Thotakura
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA, United States
| | - Liana Basova
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA, United States
| | - Helen P Makarenkova
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA, United States
| |
Collapse
|
24
|
Clayton R, Göbel K, Niessen C, Paus R, Steensel M, Lim X. Homeostasis of the sebaceous gland and mechanisms of acne pathogenesis. Br J Dermatol 2019; 181:677-690. [DOI: 10.1111/bjd.17981] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2019] [Indexed: 12/13/2022]
Affiliation(s)
- R.W. Clayton
- Skin Research Institute of Singapore Agency for Science, Technology and Research (A*STAR) Singapore
- Centre for Dermatology Research University of Manchester, and NIHR Manchester Biomedical Research Centre Manchester U.K
| | - K. Göbel
- Skin Research Institute of Singapore Agency for Science, Technology and Research (A*STAR) Singapore
- Department of Dermatology Cologne Excellence Cluster on Stress Responses in Aging Associated Diseases (CECAD), and Centre for Molecular Medicine Cologne The University of Cologne Germany
| | - C.M. Niessen
- Department of Dermatology Cologne Excellence Cluster on Stress Responses in Aging Associated Diseases (CECAD), and Centre for Molecular Medicine Cologne The University of Cologne Germany
| | - R. Paus
- Centre for Dermatology Research University of Manchester, and NIHR Manchester Biomedical Research Centre Manchester U.K
- Department of Dermatology and Cutaneous Surgery University of Miami Miller School of Medicine Miami FL U.S.A
| | - M.A.M. Steensel
- Skin Research Institute of Singapore Agency for Science, Technology and Research (A*STAR) Singapore
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore
| | - X. Lim
- Skin Research Institute of Singapore Agency for Science, Technology and Research (A*STAR) Singapore
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore
| |
Collapse
|
25
|
Bioactivity Determination of a Therapeutic Recombinant Human Keratinocyte Growth Factor by a Validated Cell-based Bioassay. Molecules 2019; 24:molecules24040699. [PMID: 30769959 PMCID: PMC6412437 DOI: 10.3390/molecules24040699] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 01/21/2023] Open
Abstract
The therapeutic recombinant human keratinocyte growth factor 1 (rhKGF-1) was approved by the FDA for oral mucositis resulting from hematopoietic stem cell transplantation for hematological malignancies in 2004. However, no recommended bioassay for rhKGF-1 bioactivity has been recorded in the U.S. Pharmacopoeia. In this study, we developed an rhKGF-1-dependent bioassay for determining rhKGF-1 bioactivity based on HEK293 and HaCat cell lines that stably expressed the luciferase reporter driven by the serum response element (SRE) and human fibroblast growth factor receptor (FGFR2) IIIb. A good responsiveness to rhKGF-1 and rhKGF-2 shared by target HEK293/HaCat cell lines was demonstrated. Our stringent validation was completely focused on specificity, linearity, accuracy, precision, and robustness according to the International Council for Harmonization (ICH) Q2 (R1) guidelines, AAPS/FDA Bioanalytical Workshop and the Chinese Pharmacopoeia. We confirmed the reliability of the method in determining rhKGF bioactivity. The validated method is highly timesaving, sensitive, and simple, and is especially valuable for providing information for quality control during the manufacture, research, and development of therapeutic rhKGF.
Collapse
|
26
|
Zinkle A, Mohammadi M. Structural Biology of the FGF7 Subfamily. Front Genet 2019; 10:102. [PMID: 30809251 PMCID: PMC6379346 DOI: 10.3389/fgene.2019.00102] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/29/2019] [Indexed: 11/13/2022] Open
Abstract
Mammalian fibroblast growth factor (FGF) signaling is intricately regulated via selective binding interactions between 18 FGF ligands and four FGF receptors (FGFR1–4), three of which (FGFR1–3) are expressed as either epithelial (“b”) or mesenchymal (“c”) splice isoforms. The FGF7 subfamily, consisting of FGF3, FGF7, FGF10, and FGF22, is unique among FGFs in that its members are secreted exclusively by the mesenchyme, and specifically activate the “b” isoforms of FGFR1 (FGFR1b) and FGFR2 (FGFR2b) present in the overlying epithelium. This unidirectional mesenchyme-to-epithelium signaling contributes to the development of essentially all organs, glands, and limbs. Structural analysis has shown that members of the FGF7 subfamily achieve their restricted specificity for FGFR1b/FGFR2b by engaging in specific contacts with two alternatively spliced loop regions in the immunoglobulin-like domain 3 (D3) of these receptors. Weak basal receptor-binding affinity further constrains the FGF7 subfamily’s specificity for FGFR1b/2b. In this review, we elaborate on the structural determinants of FGF7 subfamily receptor-binding specificity, and discuss how affinity differences among the four members for the heparin sulfate (HS) co-receptor contribute to their disparate biological activities.
Collapse
Affiliation(s)
- Allen Zinkle
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, United States
| | - Moosa Mohammadi
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, United States
| |
Collapse
|
27
|
Kumtornrut C, Yamauchi T, Koike S, Aiba S, Yamasaki K. Androgens modulate keratinocyte differentiation indirectly through enhancing growth factor production from dermal fibroblasts. J Dermatol Sci 2019; 93:150-158. [PMID: 30792099 DOI: 10.1016/j.jdermsci.2019.01.007] [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: 10/29/2018] [Revised: 12/28/2018] [Accepted: 01/21/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The main pathogenesis of acne vulgaris is increase in sebum production and abnormal keratinization of the hair infundibulum. The androgens are involved in acne pathogenesis by modulating sebaceous glands to enhance sebum production. However, the molecular mechanisms of abnormal keratinization of the hair infundibulum are not fully elucidated. OBJECTIVE We hypothesized that the androgens affect the dermal fibroblasts, another androgen receptor-positive cells in the skin, resulting in abnormal keratinization through keratinocyte-fibroblast interaction. METHODS We investigated effects of androgens and estrogens on growth factors expressions by RT-PCR and western blot analysis in human fibroblast (hFB), human keratinocyte (hKC), and fibroblast-keratinocyte co-culture. In vivo, we examined the growth factor expression in acne lesions compared to normal hair follicles by laser-assisted confocal microscope. RESULTS In vitro, androgens but not estrogens significantly increased amphiregulin (AREG), epiregulin (EREG), fibroblast growth factor (FGF) 10, and insulin-like growth factor binding protein (IGFBP) 5 mRNA and protein expressions in human fibroblasts but not in keratinocytes. In vivo, AREG, EREG, FGF10, and IGFBP5 were more abundant in acne lesion compared to normal facial skin. FGF10 suppressed cytokeratin 1 and cytokeratin 10 expression in hKC, which was along with the decreased ratio of cytokeratin 10 against cytokeratin 14 in acne lesions compared to normal facial skin. Also, DHT suppressed cytokeratin 1 and cytokeratin 10, in fibroblast-keratinocyte co-culture similarly to the effect of FGF10 to hKC. CONCLUSION These observations suggested that androgens enhance growth factors production from dermal fibroblasts, and growth factors from fibroblasts alter keratinocyte differentiation in acne lesion.
Collapse
Affiliation(s)
- Chanat Kumtornrut
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan; Division of Dermatology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Takeshi Yamauchi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Saaya Koike
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Kenshi Yamasaki
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
28
|
Collins LE, Troeberg L. Heparan sulfate as a regulator of inflammation and immunity. J Leukoc Biol 2018; 105:81-92. [PMID: 30376187 DOI: 10.1002/jlb.3ru0618-246r] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/30/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022] Open
Abstract
Heparan sulfate is found on the surface of most cell types, as well as in basement membranes and extracellular matrices. Its strong anionic properties and highly variable structure enable this glycosaminoglycan to provide binding sites for numerous protein ligands, including many soluble mediators of the immune system, and may promote or inhibit their activity. The formation of ligand binding sites on heparan sulfate (HS) occurs in a tissue- and context-specific fashion through the action of several families of enzymes, most of which have multiple isoforms with subtly different specificities. Changes in the expression levels of these biosynthetic enzymes occur in response to inflammatory stimuli, resulting in structurally different HS and acquisition or loss of binding sites for immune mediators. In this review, we discuss the multiple roles for HS in regulating immune responses, and the evidence for inflammation-associated changes to HS structure.
Collapse
Affiliation(s)
- Laura E Collins
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Linda Troeberg
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
29
|
Kawane T, Qin X, Jiang Q, Miyazaki T, Komori H, Yoshida CA, Matsuura-Kawata VKDS, Sakane C, Matsuo Y, Nagai K, Maeno T, Date Y, Nishimura R, Komori T. Runx2 is required for the proliferation of osteoblast progenitors and induces proliferation by regulating Fgfr2 and Fgfr3. Sci Rep 2018; 8:13551. [PMID: 30202094 PMCID: PMC6131145 DOI: 10.1038/s41598-018-31853-0] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/28/2018] [Indexed: 01/18/2023] Open
Abstract
Runx2 and Sp7 are essential transcription factors for osteoblast differentiation. However, the molecular mechanisms responsible for the proliferation of osteoblast progenitors remain unclear. The early onset of Runx2 expression caused limb defects through the Fgfr1–3 regulation by Runx2. To investigate the physiological role of Runx2 in the regulation of Fgfr1–3, we compared osteoblast progenitors in Sp7−/− and Runx2−/− mice. Osteoblast progenitors accumulated and actively proliferated in calvariae and mandibles of Sp7−/− but not of Runx2−/− mice, and the number of osteoblast progenitors and their proliferation were dependent on the gene dosage of Runx2 in Sp7−/− background. The expression of Fgfr2 and Fgfr3, which were responsible for the proliferation of osteoblast progenitors, was severely reduced in Runx2−/− but not in Sp7−/− calvariae. Runx2 directly regulated Fgfr2 and Fgfr3, increased the proliferation of osteoblast progenitors, and augmented the FGF2-induced proliferation. The proliferation of Sp7−/− osteoblast progenitors was enhanced and strongly augmented by FGF2, and Runx2 knockdown reduced the FGF2-induced proliferation. Fgfr inhibitor AZD4547 abrogated all of the enhanced proliferation. These results indicate that Runx2 is required for the proliferation of osteoblast progenitors and induces proliferation, at least partly, by regulating Fgfr2 and Fgfr3 expression.
Collapse
Affiliation(s)
- Tetsuya Kawane
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Xin Qin
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Qing Jiang
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan.,Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Toshihiro Miyazaki
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Hisato Komori
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Carolina Andrea Yoshida
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | | | - Chiharu Sakane
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Yuki Matsuo
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Kazuhiro Nagai
- Transfusion and Cell Therapy Unit, Nagasaki University Hospital, Nagasaki, 852-8501, Japan
| | - Takafumi Maeno
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan.,Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, 545-8585, Japan
| | - Yuki Date
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan.,Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Riko Nishimura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, 565-0871, Japan
| | - Toshihisa Komori
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan. .,Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan.
| |
Collapse
|
30
|
Geng L, Hou N, Zhang M, Xu Y, Zhang Q, Wang J, Zhang L, Zhang Q. Comparative study of the effect of different fucoidans from Sargassum maclurei and Saccharina japonica on FGFs/FGFR signaling activation in BaF3 cells. Int J Biol Macromol 2018; 107:2429-2435. [PMID: 29055704 DOI: 10.1016/j.ijbiomac.2017.10.122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/03/2017] [Accepted: 10/18/2017] [Indexed: 01/21/2023]
Abstract
Different sulfated polysaccharides have distinct abilities to activate specific fibroblast growth factor (FGF) signaling pathways in FGFR1c-expressing BaF3 cells. In the current study, we first isolated and characterized different fucoidan fractions from Sargassum maclurei and Saccharina japonica. All of the fucoidan fractions were incubated with BaF3 cells in the presence of FGF-1, -2, -7, -8, -9, and -10, respectively, to evaluate their FGFs/FGFR1c signal-activating ability. Our data showed that low molecular weight fucoidan fraction from S. japonica with highest sulfate content (LMWF-2M) had the most potent activity among all of the six tested FGFs. Low sulfated heteropolysacchairde fraction LMWF-0.5M, along with SMP-1, SMP-D-1, and SMP-A-1, only activated the FGF-2/FGFR1c pair (P<0.05). In contrast, SMP, SMP-A, and SMP-D stimulated BaF3 cell proliferation except for FGF-8. Both LMWF-1M and LMWF-2M facilitated FGF-1-, -2-, -8-, and -9-based signaling. The sulfate content was the major contributing factor to the observed activity followed by the molecular weight. The monosaccharide composition also affected the activity, in that SMP and its derivatives with varied monosaccharide composition could not induce BaF3 cell proliferation in the presence of FGF-8. The structure-activity relationships revealed in current study provided useful information for the potential application of fucoidans in FGF/FGFR signaling regulation.
Collapse
Affiliation(s)
- Lihua Geng
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech., Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ningning Hou
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech., Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meng Zhang
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yingjie Xu
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Qi Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech., Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lijuan Zhang
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Quanbin Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
31
|
Abashev TM, Metzler MA, Wright DM, Sandell LL. Retinoic acid signaling regulates Krt5 and Krt14 independently of stem cell markers in submandibular salivary gland epithelium. Dev Dyn 2018; 246:135-147. [PMID: 27884045 DOI: 10.1002/dvdy.24476] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Retinoic acid (RA), the active metabolite of vitamin A, has been demonstrated to be important for growth and branching morphogenesis of mammalian embryonic salivary gland epithelium. However, it is not known whether RA functions directly within epithelial cells or in associated tissues that influence morphogenesis of salivary epithelium. Moreover, downstream targets of RA regulation have not been identified. RESULTS Here, we show that canonical RA signaling occurs in multiple tissues of embryonic mouse salivary glands, including epithelium, associated parasympathetic ganglion neurons, and nonneuronal mesenchyme. By culturing epithelium explants in isolation from other tissues, we demonstrate that RA influences epithelium morphogenesis by direct action in that tissue. Moreover, we demonstrate that inhibition of RA signaling represses cell proliferation and expression of FGF10 signaling targets, and upregulates expression of basal epithelial keratins Krt5 and Krt14. Importantly, we show that the stem cell gene Kit is regulated inversely from Krt5/Krt14 by RA signaling. CONCLUSIONS RA regulates Krt5 and Krt14 expression independently of stem cell character in developing salivary epithelium. RA, or chemical inhibitors of RA signaling, could potentially be used for modulating growth and differentiation of epithelial stem cells for the purpose of re-populating damaged glands or generating bioengineered organs. Developmental Dynamics 246:135-147, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Timur M Abashev
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| | - Melissa A Metzler
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| | - Diana M Wright
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| | - Lisa L Sandell
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| |
Collapse
|
32
|
He MY, Wang G, Han SS, Li K, Jin Y, Liu M, Si ZP, Wang J, Liu GS, Yang X. Negative impact of hyperglycaemia on mouse alveolar development. Cell Cycle 2017; 17:80-91. [PMID: 29143549 DOI: 10.1080/15384101.2017.1403683] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Diabetes mellitus in pregnancy has been known to affect the embryonic development of various systems, including cardiovascular and nervous systems. However, whether this disease could have a negative impact on embryonic respiratory system remains controversial. In this study, we demonstrated that pregestational diabetes mellitus (PGDM)-induced defects in lung development in mice are mainly characterized by the changes in the morphological structure of the lung. Immunostaining and Western blotting showed that proliferation increased and apoptosis decreased in PGDM. Hyperglycaemia caused pulmonary tissue fibrationas manifested by an increase in Masson staining and decorin expression in PGDM lungs, and the immunofluorescent pro-SPC+ type II pulmonary epithelial cell number was decreased. The alteration of pulmonary epithelial cell differentiation might be due to hyperglycaemia-activated Wnt signalling and suppressed GATA6 expression in PGDM mouse lung tissues and MLE-12 cells. The treatment of MLE-12 cells with high glucose in the presence/absence of XAV939 or su5402 further proved that hyperglycaemia suppressed the expression of GATA6 and pro-SPC by activating Wnt signalling and induced the expression of decorin, α-SMA and TGF-β by activating Fgf signalling. Therefore, in this study, we revealed that hyperglycemia induced dysfunctional pulmonary cell apoptosis and proliferation, as well as pulmonary myofibroblast hyperplasia, which contributed to the formation of aberrant structure of alveolar walls. Furthermore, the hyperglycaemia also inhibited the differentiation of pulmonary epithelial cells through the canonical Wnt and Fgf signalling, and the alteration of Fgf and Wnt signalling activated TGF-β, which would promote the AECII EMT process.
Collapse
Affiliation(s)
- Mei-Yao He
- a Department of Pediatrics and Neonatology , Institute of Fetal-Preterm Labor Medicine ; The First Affiliated Hospital , Jinan University , Guangzhou 510630 , China
| | - Guang Wang
- b Division of Histology & Embryology , Key Laboratory for Regenerative Medicine of the Ministry of Education , Medical College , Jinan University , Guangzhou 510632 , China
| | - Sha-Sha Han
- a Department of Pediatrics and Neonatology , Institute of Fetal-Preterm Labor Medicine ; The First Affiliated Hospital , Jinan University , Guangzhou 510630 , China
| | - Ke Li
- b Division of Histology & Embryology , Key Laboratory for Regenerative Medicine of the Ministry of Education , Medical College , Jinan University , Guangzhou 510632 , China
| | - Ya Jin
- a Department of Pediatrics and Neonatology , Institute of Fetal-Preterm Labor Medicine ; The First Affiliated Hospital , Jinan University , Guangzhou 510630 , China
| | - Meng Liu
- b Division of Histology & Embryology , Key Laboratory for Regenerative Medicine of the Ministry of Education , Medical College , Jinan University , Guangzhou 510632 , China
| | - Zhen-Peng Si
- a Department of Pediatrics and Neonatology , Institute of Fetal-Preterm Labor Medicine ; The First Affiliated Hospital , Jinan University , Guangzhou 510630 , China
| | - Ju Wang
- c Institute of Biomedicine , National Engineering Research Center of Genetic Medicine , Jinan University , Guangzhou 510632 , China
| | - Guo-Sheng Liu
- a Department of Pediatrics and Neonatology , Institute of Fetal-Preterm Labor Medicine ; The First Affiliated Hospital , Jinan University , Guangzhou 510630 , China
| | - Xuesong Yang
- b Division of Histology & Embryology , Key Laboratory for Regenerative Medicine of the Ministry of Education , Medical College , Jinan University , Guangzhou 510632 , China
| |
Collapse
|
33
|
Kotnala S, Tyagi A, Muyal JP. rHuKGF ameliorates protease/anti-protease imbalance in emphysematous mice. Pulm Pharmacol Ther 2017; 45:124-135. [DOI: 10.1016/j.pupt.2017.05.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/02/2017] [Accepted: 05/24/2017] [Indexed: 10/19/2022]
|
34
|
Sathi GA, Farahat M, Hara ES, Taketa H, Nagatsuka H, Kuboki T, Matsumoto T. MCSF orchestrates branching morphogenesis in developing submandibular gland tissue. J Cell Sci 2017; 130:1559-1569. [DOI: 10.1242/jcs.196907] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/21/2017] [Indexed: 12/16/2022] Open
Abstract
The importance of macrophages in tissue development and regeneration have been strongly emphasized. However, the specific roles of macrophage colony-stimulating factor (MCSF), the key regulator of macrophage differentiation, in glandular tissue development have been unexplored. Here, we disclose new macrophage-independent roles of MCSF in tissue development. We initially found that MCSF is markedly upregulated at embryonic day E13.5, at a stage preceding the colonization of macrophages (at E15.5) in mouse submandibular gland (SMG) tissue. Surprisingly, MCSF-induced branching morphogenesis was based on a direct effect on epithelial cells, as well as indirectly, by modulating the expression of major growth factors of SMG growth, FGF7 and FGF10, via the phosphoinositide 3-kinase (PI3K) pathway. Additionally, given the importance of neurons in SMG organogenesis, MCSF-induced SMG growth was associated with regulation of neurturin expression and neuronal network development during early SMG development in an in vitro organogenesis model as well as in vivo. These results indicate that MCSF plays pleiotropic roles and is an important regulator of early SMG morphogenesis.
Collapse
Affiliation(s)
- Gulsan Ara Sathi
- Department of Biomaterials, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Mahmoud Farahat
- Department of Biomaterials, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Emilio Satoshi Hara
- Department of Biomaterials, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Hiroaki Taketa
- Center for the Development of Medical and Health Care Education, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Hitoshi Nagatsuka
- Department of Oral Pathology and Medicine, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Takuya Matsumoto
- Department of Biomaterials, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| |
Collapse
|
35
|
Mescher AL, Neff AW, King MW. Inflammation and immunity in organ regeneration. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 66:98-110. [PMID: 26891614 DOI: 10.1016/j.dci.2016.02.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/19/2016] [Accepted: 02/09/2016] [Indexed: 06/05/2023]
Abstract
The ability of vertebrates to regenerate amputated appendages is increasingly well-understood at the cellular level. Cells mediating an innate immune response and inflammation in the injured tissues are a prominent feature of the limb prior to formation of a regeneration blastema, with macrophage activity necessary for blastema growth and successful development of the new limb. Studies involving either anti-inflammatory or pro-inflammatory agents suggest that the local inflammation produced by injury and its timely resolution are both important for regeneration, with blastema patterning inhibited in the presence of unresolved inflammation. Various experiments with Xenopus larvae at stages where regenerative competence is declining show improved digit formation after treatment with certain immunosuppressive, anti-inflammatory, or antioxidant agents. Similar work with the larval Xenopus tail has implicated adaptive immunity with regenerative competence and suggests a requirement for regulatory T cells in regeneration, which also occurs in many systems of tissue regeneration. Recent analyses of the human nail organ indicate a capacity for local immune tolerance, suggesting roles for adaptive immunity in the capacity for mammalian appendage regeneration. New information and better understanding regarding the neuroendocrine-immune axis in the response to stressors, including amputation, suggest additional approaches useful for investigating effects of the immune system during repair and regeneration.
Collapse
Affiliation(s)
- Anthony L Mescher
- Center for Developmental and Regenerative Biology; Indiana University School of Medicine - Bloomington, USA.
| | - Anton W Neff
- Center for Developmental and Regenerative Biology; Indiana University School of Medicine - Bloomington, USA.
| | - Michael W King
- Center for Developmental and Regenerative Biology; Indiana University School of Medicine - Terre Haute, USA.
| |
Collapse
|
36
|
Chen X, Hou XM, Fan YF, Jin YT, Wang YL. Sonic hedgehog protein regulates fibroblast growth factor 8 expression in metanephric explant culture from BALB/c mice: Possible mechanisms associated with renal morphogenesis. Mol Med Rep 2016; 14:2929-36. [PMID: 27510750 PMCID: PMC5042753 DOI: 10.3892/mmr.2016.5614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 06/17/2016] [Indexed: 11/06/2022] Open
Abstract
The sonic hedgehog (SHH) morphogen regulates cell differentiation and controls a number of genes during renal morphogenesis. To date, the effects of SHH on fibroblast growth factors (Fgfs) in embryonic kidney development remain unclear. In the present study, explants of BALB/c mouse embryonic kidney tissues were used to investigate the role of exogenous SHH on Fgf8 and Fgf10 expression levels ex vivo. Ureteric bud branches and epithelial metanephric derivatives were used to determine the renal morphogenesis with Dolichos biflorus agglutinin or hematoxylin‑eosin staining. mRNA expression levels were determined using reverse transcription‑quantitative polymerase chain reaction, while the protein expression levels were examined using immunohistochemistry and western blot analysis. During the initial stages of metanephric development, low levels of SHH, Fgf8, and Fgf10 expression were observed, which were found to increase significantly during more advanced stages of metanephric development. In addition, exogenous SHH protein treatment increased the number of ureteric bud branches and enhanced the formation of nephrons. Exogenous SHH reduced the Fgf8 mRNA and protein expression levels, whereas cyclopamine (an SHH‑smoothened receptor inhibitor) interfered with SHH‑mediated downregulation of Fgf8 expression. By contrast, exogenous SHH protein was not found to modulate Fgf10 mRNA and protein expression levels. In conclusion, these results indicate that the modulatory effects of SHH on BALB/c mouse metanephric explant cultures may involve the regulation of Fgf8 expression but not Fgf10 expression, which provides evidence for the functional role of Fgf proteins in renal morphogenesis.
Collapse
Affiliation(s)
- Xing Chen
- Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xiao-Ming Hou
- Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - You-Fei Fan
- Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yu-Ting Jin
- Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yu-Lin Wang
- Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
37
|
Akizawa H, Nagatomo H, Odagiri H, Kohri N, Yamauchi N, Yanagawa Y, Nagano M, Takahashi M, Kawahara M. Conserved roles of fibroblast growth factor receptor 2 signaling in the regulation of inner cell mass development in bovine blastocysts. Mol Reprod Dev 2016; 83:516-25. [DOI: 10.1002/mrd.22646] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/30/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Hiroki Akizawa
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Hiroaki Nagatomo
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Haruka Odagiri
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Nanami Kohri
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Nobuhiko Yamauchi
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Yojiro Yanagawa
- Laboratory of Theriogenology, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine; Hokkaido University; Sapporo Japan
| | - Masashi Nagano
- Laboratory of Theriogenology, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine; Hokkaido University; Sapporo Japan
| | - Masashi Takahashi
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Manabu Kawahara
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| |
Collapse
|
38
|
Abstract
The fibroblast growth factor (Fgf) family of ligands and receptor tyrosine kinases is required throughout embryonic and postnatal development and also regulates multiple homeostatic functions in the adult. Aberrant Fgf signaling causes many congenital disorders and underlies multiple forms of cancer. Understanding the mechanisms that govern Fgf signaling is therefore important to appreciate many aspects of Fgf biology and disease. Here we review the mechanisms of Fgf signaling by focusing on genetic strategies that enable in vivo analysis. These studies support an important role for Erk1/2 as a mediator of Fgf signaling in many biological processes but have also provided strong evidence for additional signaling pathways in transmitting Fgf signaling in vivo.
Collapse
Affiliation(s)
- J Richard Brewer
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Pierre Mazot
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| |
Collapse
|
39
|
Fibroblast Growth Factor-10 (FGF-10) Mobilizes Lung-resident Mesenchymal Stem Cells and Protects Against Acute Lung Injury. Sci Rep 2016; 6:21642. [PMID: 26869337 PMCID: PMC4751498 DOI: 10.1038/srep21642] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 01/25/2016] [Indexed: 01/07/2023] Open
Abstract
FGF-10 can prevent or reduce lung specific inflammation due to traumatic or infectious lung injury. However, the exact mechanisms are poorly characterized. Additionally, the effect of FGF-10 on lung-resident mesenchymal stem cells (LR-MSCs) has not been studied. To better characterize the effect of FGF-10 on LR-MSCs, FGF-10 was intratracheally delivered into the lungs of rats. Three days after instillation, bronchoalveolar lavage was performed and plastic-adherent cells were cultured, characterized and then delivered therapeutically to rats after LPS intratracheal instillation. Immunophenotyping analysis of FGF-10 mobilized and cultured cells revealed expression of the MSC markers CD29, CD73, CD90, and CD105, and the absence of the hematopoietic lineage markers CD34 and CD45. Multipotency of these cells was demonstrated by their capacity to differentiate into osteocytes, adipocytes, and chondrocytes. Delivery of LR-MSCs into the lungs after LPS injury reduced the inflammatory response as evidenced by decreased wet-to-dry ratio, reduced neutrophil and leukocyte recruitment and decreased inflammatory cytokines compared to control rats. Lastly, direct delivery of FGF-10 in the lungs of rats led to an increase of LR-MSCs in the treated lungs, suggesting that the protective effect of FGF-10 might be mediated, in part, by the mobilization of LR-MSCs in lungs.
Collapse
|
40
|
Abstract
Fibroblast growth factors (FGFs) have been shown to alter growth and differentiation of reproductive tissues in a variety of species. Within the female reproductive tract, the effects of FGFs have been focused on the ovary, and the most studied one is FGF2, which stimulates granulosa cell proliferation and decreases differentiation (decreased steroidogenesis). Other FGFs have also been implicated in ovarian function, and this review summarizes the effects of members of two subfamilies on ovarian function; the FGF7 subfamily that also contains FGF10, and the FGF8 subfamily that also contains FGF18. There are data to suggest that FGF8 and FGF18 have distinct actions on granulosa cells, despite their apparent similar receptor binding properties. Studies of non-reproductive developmental biology also indicate that FGF8 is distinct from FGF18, and that FGF7 is also distinct from FGF10 despite similar receptor binding properties. In this review, the potential mechanisms of differential action of FGF7/FGF10 and FGF8/FGF18 during organogenesis will be reviewed and placed in the context of follicle development. A model is proposed in which FGF8 and FGF18 differentially activate receptors depending on the properties of the extracellular matrix in the follicle.
Collapse
Affiliation(s)
- Christopher A Price
- Faculty of Veterinary MedicineCentre de recherche en reproduction animale, University of Montreal, 3200 rue Sicotte, St-Hyacinthe, Quebec, Canada J2S 7C6
| |
Collapse
|
41
|
Keratinocyte growth factor-2 inhibits bacterial infection with Pseudomonas aeruginosa pneumonia in a mouse model. J Infect Chemother 2015; 22:44-52. [PMID: 26617350 DOI: 10.1016/j.jiac.2015.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 09/24/2015] [Accepted: 10/14/2015] [Indexed: 11/22/2022]
Abstract
To determine protective effects of concurrent administration of Keratinocyte growth factor-2 (KGF-2) with Pseudomonas aeruginosa (P. aeruginosa) inoculation on the induced pneumonia. KGF-2 (5 mg/kg) was concurrently administered into the left lobe of 55 mice with P. aeruginosa PAO1 (5 × 10(6) CFU, half-lethal dose); 55 mice in the control group were concurrently administered PBS with the PAO1. We detected and analyzed: body temperature; amount of P. aeruginosa in homogenates; count of total number of nucleated cells and of mononuclear macrophages; protein concentration in bronchoalveolar lavage fluid (BALF); lung wet-to-dry weight ratio; cytokines in BALF and blood; and lung morphology. To study survival rate, concurrent administration of KGF-2 (experimental group) versus PBS (control) with a lethal dose of PAO1 (1 × 10(7) CFU was performed, and survivorship was documented for 7 days post-inoculation. The bacterial CFU in lung homogenates was significantly decreased in the KGF-2 group compared to the control group. There were significantly more mononuclear macrophages in the BALF from the KGF-2 group than from the control group (p < 0.05). KGF-2 increased the surfactant protein and GM-CSF mRNA in lung at 6 h and 72 h after inoculation. Significant reduction of lung injury scores, protein concentrations, lung wet-to-dry weight ratio, and IL-6 and TNF-α levels was noted in the KGF-2 treated rats at 72 h after inoculation (p < 0.05). The 7-day survival rate of the KGF-2 group was significantly higher than that of the control group (p < 0.05). Concurrent administration of KGF-2 facilitates the clearance of P. aeruginosa from the lungs, attenuates P. aeruginosa-induced lung injury, and extends the 7-day survival rate in mice model with P. aeruginosa pneumonia.
Collapse
|
42
|
FGF10: A multifunctional mesenchymal-epithelial signaling growth factor in development, health, and disease. Cytokine Growth Factor Rev 2015; 28:63-9. [PMID: 26559461 DOI: 10.1016/j.cytogfr.2015.10.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 10/19/2015] [Indexed: 12/15/2022]
Abstract
The FGF family comprises 22 members with diverse functions in development and health. FGF10 specifically activates FGFR2b in a paracrine manner with heparan sulfate as a co-factor. FGF10and FGFR2b are preferentially expressed in the mesenchyme and epithelium, respectively. FGF10 is a mesenchymal signaling molecule in the epithelium. FGF10 knockout mice die shortly after birth due to the complete absence of lungs as well as fore- and hindlimbs. FGF10 is also essential for the development of multiple organs. The phenotypes of Fgf10 knockout mice are very similar to those of FGFR2b knockout mice, indicating that FGF10 acts as a ligand that is specific to FGFR2b in mouse multi-organ development. FGF10 also plays roles in epithelial-mesenchymal transition, the repair of tissue injury, and embryonic stem cell differentiation. In humans, FGF10 loss-of-function mutations result in inherited diseases including aplasia of lacrimal and salivary gland, lacrimo-auriculo-dento-digital syndrome, and chronic obstructive pulmonary disease. FGF10 is also involved in the oncogenicity of pancreatic and breast cancers. Single nucleotide polymorphisms in FGF10 are also potential risk factors for limb deficiencies, cleft lip and palate, and extreme myopia. These findings indicate that FGF10 is a crucial paracrine signal from the mesenchyme to epithelium for development, health, and disease.
Collapse
|
43
|
Human adipose-derived stromal cells as a feeder layer to improve keratinocyte expansion for clinical applications. Tissue Eng Regen Med 2015. [DOI: 10.1007/s13770-015-0007-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
44
|
Shimizu O, Yasumitsu T, Shiratsuchi H, Oka S, Watanabe T, Saito T, Yonehara Y. Immunolocalization of FGF-2, -7, -8, -10 and FGFR-1-4 during regeneration of the rat submandibular gland. J Mol Histol 2015; 46:421-9. [PMID: 26173945 DOI: 10.1007/s10735-015-9631-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 07/07/2015] [Indexed: 01/13/2023]
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) play important roles in the development of the submandibular gland. Although regeneration of submandibular glands follows a similar process to their development, it is unknown how FGFs and FGFRs are distributed during regeneration of submandibular gland. The aim of this study was to determine the localization of FGFs and FGFRs during such regenerative processes. After 7 days' obstruction, the submandibular glands were collected at days 0, 1, 3, 7, 11 and 14 after duct release to study regeneration. The regenerative processes of the submandibular gland were investigated by immunohistochemistry for FGF-2, 7, 8, 10 and FGFR-1-4. Immunohistochemical staining revealed that FGF-2 was moderately expressed in the epithelial cells of duct-like structures (DLS) and newly formed acinar cells (NFAC) at days 0-7, and strongly in intercalated duct (ICD) at control gland and Day 7-14. FGF-7 was localized moderately in NFAC and DLS. FGF-8 was localized moderately in the epithelial cells of DLS during regeneration. Strong positive immunoreactions for FGF-10 were found in NFAC and the epithelial cells of DLS during regeneration, as well as the ICD and lateral surfaces of the maturing acinar cells (MAC). FGFR-1 was expressed moderately in the ICD, and weakly in the NFAC and MAC. Positive immunoreactions for FGFR-2 were not observed during regeneration. Additionally, FGFR-4 was detected strongly in the ICD and slightly in NFAC. These findings suggest that FGF-2, -7, -8 and -10 play important roles in NFAC, MAC, and DLS through FGFR-1 and -4 during regeneration of submandibular gland.
Collapse
Affiliation(s)
- Osamu Shimizu
- Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan,
| | | | | | | | | | | | | |
Collapse
|
45
|
MacKenzie B, Korfei M, Henneke I, Sibinska Z, Tian X, Hezel S, Dilai S, Wasnick R, Schneider B, Wilhelm J, El Agha E, Klepetko W, Seeger W, Schermuly R, Günther A, Bellusci S. Increased FGF1-FGFRc expression in idiopathic pulmonary fibrosis. Respir Res 2015; 16:83. [PMID: 26138239 PMCID: PMC4495640 DOI: 10.1186/s12931-015-0242-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 06/24/2015] [Indexed: 01/04/2023] Open
Abstract
Background Recent clinical studies show that tyrosine kinase inhibitors slow the rate of lung function decline and decrease the number of acute exacerbations in patients with Idiopathic Pulmonary Fibrosis (IPF). However, in the murine bleomycin model of fibrosis, not all tyrosine kinase signaling is detrimental. Exogenous ligands Fibroblast Growth Factor (FGF) 7 and 10 improve murine lung repair and increase survival after injury via tyrosine kinase FGF receptor 2b-signaling. Therefore, the level and location of FGF/FGFR expression as well as the exogenous effect of the most highly expressed FGFR2b ligand, FGF1, was analyzed on human lung fibroblasts. Methods FGF ligand and receptor expression was evaluated in donor and IPF whole lung homogenates using western blotting and qPCR. Immunohistochemistry for FGF1 and FGFR1/2/3/4 were performed on human lung tissue. Lastly, the effects of FGF1, a potent, multi-FGFR ligand, were studied on primary cultures of IPF and non-IPF donor fibroblasts. Western blots for pro-fibrotic markers, proliferation, FACS for apoptosis, transwell assays and MetaMorph analyses on cell cultures were performed. Results Whole lung homogenate analyses revealed decreased FGFR b-isoform expression, and an increase in FGFR c-isoform expression. Of the FGFR2b-ligands, FGF1 was the most significantly increased in IPF patients; downstream targets of FGF-signaling, p-ERK1/2 and p-AKT were also increased. Immunohistochemistry revealed FGF1 co-localization within basal cell sheets, myofibroblast foci, and Surfactant protein-C positive alveolar epithelial type-II cells as well as co-localization with FGFR1, FGFR2, FGFR3, FGFR4 and myofibroblasts expressing the migratory marker Fascin. Both alone and in the presence of heparin, FGF1 led to increased MAPK-signaling in primary lung fibroblasts. While smooth muscle actin was unchanged, heparin + FGF1 decreased collagen production in IPF fibroblasts. In addition, FGF1 + heparin increased apoptosis and cell migration. The FGFR inhibitor (PD173074) attenuated these effects. Conclusions Strong expression of FGF1/FGFRs in pathogenic regions of IPF suggest that aberrant FGF1-FGFR signaling is increased in IPF patients and may contribute to the pathogenesis of lung fibrosis by supporting fibroblast migration and increased MAPK-signaling. Electronic supplementary material The online version of this article (doi:10.1186/s12931-015-0242-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- BreAnne MacKenzie
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany
| | - Martina Korfei
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany
| | - Ingrid Henneke
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany
| | - Zaneta Sibinska
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany
| | - Xia Tian
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany
| | - Stefanie Hezel
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany
| | - Salma Dilai
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany
| | - Roxana Wasnick
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany
| | - Beate Schneider
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany
| | - Jochen Wilhelm
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany
| | - Elie El Agha
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany
| | - Walter Klepetko
- Department of Thoracic Surgery, General Hospital University Vienna, Vienna, Austria
| | - Werner Seeger
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany.,German Center for Lung Research, Greifenstein, Germany
| | - Ralph Schermuly
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany.,German Center for Lung Research, Greifenstein, Germany
| | - Andreas Günther
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany.,German Center for Lung Research, Greifenstein, Germany.,AGAPLESION Lung Clinic Waldhof-Elgershausen, Greifenstein, Germany
| | - Saverio Bellusci
- German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, Giessen, Hessen, Germany. .,German Center for Lung Research, Greifenstein, Germany. .,Developmental Biology Program, Division of Surgery, Saban Research Institute of Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, USA. .,Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya Street, Kazan, 420008, Russian Federation.
| |
Collapse
|
46
|
Saito S, Morishima K, Ui T, Hoshino H, Matsubara D, Ishikawa S, Aburatani H, Fukayama M, Hosoya Y, Sata N, Lefor AK, Yasuda Y, Niki T. The role of HGF/MET and FGF/FGFR in fibroblast-derived growth stimulation and lapatinib-resistance of esophageal squamous cell carcinoma. BMC Cancer 2015; 15:82. [PMID: 25884729 PMCID: PMC4345039 DOI: 10.1186/s12885-015-1065-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 02/02/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Although advanced esophageal squamous-cell carcinoma (ESCC) is treated using a multidisciplinary approach, outcomes remain unsatisfactory. The microenvironment of cancer cells has recently been shown to strongly influence the biologic properties of malignancies. We explored the effect of supernatant from esophageal fibroblasts on the cell growth and chemo-resistance of ESCC cell lines. METHODS We used 22 ESCC cell lines, isolated primary human esophageal fibroblasts and immortalized fibroblasts. We first examined cell proliferation induced by fibroblast supernatant. The effect of supernatant was evaluated to determine whether paracrine signaling induced by fibroblasts can influence the proliferation of cancer cells. Next, we examined the effects of adding growth factors HGF, FGF1, FGF7, and FGF10, to the culture medium of cancer cells. These growth factors are assumed to be present in the culture supernatants of fibroblasts and may exert a paracrine effect on the proliferation of cancer cells. We also examined the intrinsic role of HGF/MET and FGFs/FGFR in ESCC proliferation. In addition, we examined the inhibitory effect of lapatinib on ESCC cell lines and studied whether the fibroblast supernatants affect the inhibitory effect of lapatinib on ESCC cell proliferation. Finally, we tested whether the FGFR inhibitor PD-173074 could eliminate the rescue effect against lapatinib that was induced by fibroblast supernatants. RESULTS The addition of fibroblast supernatant induces cell proliferation in the majority of cell lines tested. The results of experiments to evaluate the effects of adding growth factors and kinase inhibitors suggests that the stimulating effect of fibroblasts was attributable in part to HGF/MET or FGF/FGFR. The results also indicate diversity in the degree of dependence on HGF/MET and FGF/FGFR among the cell lines. Though lapanitib at 1 μM inhibits cell proliferation by more than 50% in the majority of the ESCC cell lines, fibroblast supernatant can rescue the growth inhibition of ESCC cells. However, the rescue effect is abrogated by co-treatment with FGFR inhibitor. CONCLUSION These results demonstrate that cell growth of ESCC depends on diverse receptor tyrosine kinase signaling, in both cell-autonomous and cell-non-autonomous manners. The combined inhibition of these signals may hold promise for the treatment of ESCC.
Collapse
Affiliation(s)
- Shin Saito
- Department of Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi, 329-0498, Japan.
| | - Kazue Morishima
- Department of Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi, 329-0498, Japan.
| | - Takashi Ui
- Department of Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi, 329-0498, Japan.
| | - Hiroko Hoshino
- Department of Integrative Pathology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi, 329-0498, Japan.
| | - Daisuke Matsubara
- Department of Integrative Pathology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi, 329-0498, Japan.
| | - Shumpei Ishikawa
- Department of Genomic Pathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-0034, Japan.
| | - Hiroyuki Aburatani
- Division of Genome Science, Research Center for Advanced Science and Technology, University of Tokyo, 4-6-1 Komaba, Meguro, Tokyo, 153-8904, Japan.
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-0033, Japan.
| | - Yoshinori Hosoya
- Department of Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi, 329-0498, Japan.
| | - Naohiro Sata
- Department of Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi, 329-0498, Japan.
| | - Alan K Lefor
- Department of Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi, 329-0498, Japan.
| | - Yoshikazu Yasuda
- Department of Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi, 329-0498, Japan.
| | - Toshiro Niki
- Department of Integrative Pathology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-City, Tochigi, 329-0498, Japan.
| |
Collapse
|
47
|
Requirement of Gαi1/3–Gab1 Signaling Complex for Keratinocyte Growth Factor–Induced PI3K–AKT–mTORC1 Activation. J Invest Dermatol 2015; 135:181-191. [DOI: 10.1038/jid.2014.326] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 06/24/2014] [Accepted: 07/14/2014] [Indexed: 01/06/2023]
|
48
|
Wu X, Huang H, Wang C, Lin S, Huang Y, Wang Y, Liang G, Yan Q, Xiao J, Wu J, Yang Y, Li X. Identification of a novel peptide that blocks basic fibroblast growth factor-mediated cell proliferation. Oncotarget 2014; 4:1819-28. [PMID: 24142482 PMCID: PMC3858566 DOI: 10.18632/oncotarget.1312] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Basic fibroblast growth factor (bFGF) has been implicated in tumor growth via interactions with its receptors (FGFRs) on the cell surface and therefore, bFGF/FGFRs are considered essential targets for cancer therapy. Herein, a consensus heptapeptide (LSPPRYP) was identified for the first time from a phage display heptapeptide library after three sequential rounds of biopanning against FGFR-expressing cells with competitive displacement of phage by bFGF, followed by subtraction of non-specific binding by FGFR-deficient cells. Phage bearing LSPPRYP showed high levels of binding to Balb/c 3T3 cells expressing high-affinity bFGF-binding FGFR (bFGFR), but not to the cells that do not express bFGFR (Cos-7), or express a very low affinity bFGFR (HaCat). The selected-phage-derived peptide synthesized by solid phase method using a rapid and practical Fmoc strategy was found to specifically compete with bFGF for binding to its receptors, inhibit bFGF-stimulated cell proliferation by inducing cell cycle arrest, and block bFGF-induced activation of Erk1 and Erk2 kinase in B16-F10 melanoma cells. Importantly, treatment of melanoma-bearing mice with the synthetic peptide significantly suppressed tumor growth. The results demonstrate a strong anticancer activity of the isolated bFGFR-binding peptide (and its future derivatives), which may have great potential for cancer therapy.
Collapse
Affiliation(s)
- Xiaoping Wu
- School of Pharmaceutical Science, Key Laboratory of Biotechnology and Pharmaceutical Engineering of Zhejiang Province, Wenzhou Medical College, Wenzhou, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Tong L, Bi J, Zhu X, Wang G, Liu J, Rong L, Wang Q, Xu N, Zhong M, Zhu D, Song Y, Bai C. Keratinocyte growth factor-2 is protective in lipopolysaccharide-induced acute lung injury in rats. Respir Physiol Neurobiol 2014; 201:7-14. [PMID: 24973472 DOI: 10.1016/j.resp.2014.06.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 06/20/2014] [Accepted: 06/20/2014] [Indexed: 11/26/2022]
Abstract
Keratinocyte growth factor-2 (KGF-2) plays a key role in lung development, but its role in acute lung injury has not been well characterized. Lipopolysaccharide instillation caused acute lung injury, which significantly elevated lung wet-to-dry weight ratio, protein and neutrophils in bronchoalveolar lavage fluid (BALF), inhibited surfactant protein A and C expression in lung tissue, and increased pathological injury. Pretreatment with KGF-2 improved the above lung injury parameters, partially restored surfactant protein A and C expression, and KGF-2 given 2-3 days before LPS challenge showed maximum lung injury improvement. Pretreatment with KGF-2 also markedly reduced the levels of TNF-α, MIP-2, IL-1β and IL-6 in BALF and the levels of IL-1β and IL-6 in lung tissue. Histological analysis showed there was increased proliferation of alveolar type II epithelial cells in lung parenchyma, which reached maximal 2 days after KGF-2 instillation. Intratracheal administration of KGF-2 attenuates lung injury induced by LPS, suggesting KGF-2 may be potent in the intervention of acute lung injury.
Collapse
Affiliation(s)
- Lin Tong
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Jing Bi
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Xiaodan Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Guifang Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Jie Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Linyi Rong
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Qin Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Nuo Xu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Ming Zhong
- Division of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Duming Zhu
- Division of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China; Shanghai Public Health Clinical Center, Shanghai 201508, PR China; Zhongshan Hospital, Qingpu Branch, Fudan University, Shanghai 201700, PR China.
| | - Chunxue Bai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China.
| |
Collapse
|
50
|
Common Amino Acid Sequences Deduced from Coding Exons of the PorcineFGF4Gene in Two Breeds and Production of the Encoded Protein inEscherichia coli. Biosci Biotechnol Biochem 2014; 77:173-7. [DOI: 10.1271/bbb.120698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|