1
|
Yang J, Sun W, Cui G. Roles of the NR2F Family in the Development, Disease, and Cancer of the Lung. J Dev Biol 2024; 12:24. [PMID: 39311119 PMCID: PMC11417824 DOI: 10.3390/jdb12030024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
The NR2F family, including NR2F1, NR2F2, and NR2F6, belongs to the nuclear receptor superfamily. NR2F family members function as transcription factors and play essential roles in the development of multiple organs or tissues in mammals, including the central nervous system, veins and arteries, kidneys, uterus, and vasculature. In the central nervous system, NR2F1/2 coordinate with each other to regulate the development of specific brain subregions or cell types. In addition, NR2F family members are associated with various cancers, such as prostate cancer, breast cancer, and esophageal cancer. Nonetheless, the roles of the NR2F family in the development and diseases of the lung have not been systematically summarized. In this review, we mainly focus on the lung, including recent findings regarding the roles of the NR2F family in development, physiological function, and cancer.
Collapse
Affiliation(s)
- Jiaxin Yang
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou 510005, China;
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China;
| | - Wenjing Sun
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China;
| | - Guizhong Cui
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou 510005, China;
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China;
| |
Collapse
|
2
|
Zhou WY, Liu P, Xia YF, Shi YJ, Xu HY, Ding M, Yang QQ, Qian SW, Tang Y, Lu Y, Tang QQ, Liu Y. NR2F6 is essential for brown adipocyte differentiation and systemic metabolic homeostasis. Mol Metab 2024; 81:101891. [PMID: 38307386 PMCID: PMC10864868 DOI: 10.1016/j.molmet.2024.101891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024] Open
Abstract
OBJECTIVE Brown adipose tissue (BAT) development and function are essential for maintaining energy balance. However, the key factors that specifically regulate brown adipogenesis require further identification. Here, we demonstrated that the nuclear receptor subfamily 2 group F member 6 (NR2F6) played a pivotal role in brown adipogenesis and energy homeostasis. METHODS We examined the differentiation of immortalized brown adipocytes and primary brown adipocytes when NR2F6 were deleted, and explored the mechanism through which NR2F6 regulated adipogenesis using ChIP-qPCR in vitro. Male wild type (WT) and Pdgfra-Cre-mediated deletion of Nr2f6 in preadipocytes (NR2F6-PKO) mice were fed with high fat diet (HFD) for 12 weeks, and adiposity, glucose intolerance, insulin resistance and inflammation were assessed. RESULTS NR2F6 exhibited abundant expression in BAT, while its expression was minimal in white adipose tissue (WAT). Within BAT, NR2F6 was highly expressed in preadipocytes, experienced a transient increase in the early stage of brown adipocyte differentiation, and significantly decreased in the mature adipocytes. Depletion of NR2F6 in preadipocytes inhibited brown adipogenesis, caused hypertrophy of brown adipocytes, and impaired thermogenic function of BAT, but without affecting WAT development. NR2F6 transcriptionally regulated PPARγ expression to promote adipogenic process in brown adipocytes. Loss of NR2F6 in preadipocytes led to increased susceptibility to diet-induced metabolic disorders. CONCLUSIONS Our findings unveiled NR2F6 as a novel key regulator of brown adipogenesis, potentially opening up new avenues for maintaining metabolic homeostasis by targeting NR2F6.
Collapse
Affiliation(s)
- Wei-Yu Zhou
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Pei Liu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi-Fan Xia
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi-Jie Shi
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hong-Yu Xu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Meng Ding
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qi-Qi Yang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shu-Wen Qian
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan Lu
- Institute of Metabolism and Regenerative Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Yang Liu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
3
|
Montero-Calle A, Jiménez de Ocaña S, Benavente-Naranjo R, Rejas-González R, Bartolomé RA, Martínez-Useros J, Sanz R, Dziaková J, Fernández-Aceñero MJ, Mendiola M, Casal JI, Peláez-García A, Barderas R. Functional Proteomics Characterization of the Role of SPRYD7 in Colorectal Cancer Progression and Metastasis. Cells 2023; 12:2548. [PMID: 37947626 PMCID: PMC10648221 DOI: 10.3390/cells12212548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
SPRY domain-containing protein 7 (SPRYD7) is a barely known protein identified via spatial proteomics as being upregulated in highly metastatic-to-liver KM12SM colorectal cancer (CRC) cells in comparison to its isogenic poorly metastatic KM12C CRC cells. Here, we aimed to analyze SPRYD7's role in CRC via functional proteomics. Through immunohistochemistry, the overexpression of SPRYD7 was observed to be associated with the poor survival of CRC patients and with an aggressive and metastatic phenotype. Stable SPRYD7 overexpression was performed in KM12C and SW480 poorly metastatic CRC cells and in their isogenic highly metastatic-to-liver-KM12SM-and-to-lymph-nodes SW620 CRC cells, respectively. Upon upregulation of SPRYD7, in vitro and in vivo functional assays confirmed a key role of SPRYD7 in the invasion and migration of CRC cells and in liver homing and tumor growth. Additionally, transient siRNA SPRYD7 silencing allowed us to confirm in vitro functional results. Furthermore, SPRYD7 was observed as an inductor of angiogenesis. In addition, the dysregulated SPRYD7-associated proteome and SPRYD7 interactors were elucidated via 10-plex TMT quantitative proteins, immunoproteomics, and bioinformatics. After WB validation, the biological pathways associated with the stable overexpression of SPRYD7 were visualized. In conclusion, it was demonstrated here that SPRYD7 is a novel protein associated with CRC progression and metastasis. Thus, SPRYD7 and its interactors might be of relevance in identifying novel therapeutic targets for advanced CRC.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.J.d.O.); (R.B.-N.); (R.R.-G.)
| | - Sofía Jiménez de Ocaña
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.J.d.O.); (R.B.-N.); (R.R.-G.)
| | - Ruth Benavente-Naranjo
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.J.d.O.); (R.B.-N.); (R.R.-G.)
| | - Raquel Rejas-González
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.J.d.O.); (R.B.-N.); (R.R.-G.)
| | - Rubén A. Bartolomé
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040 Madrid, Spain;
| | - Javier Martínez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-University Hospital Fundación Jiménez Díaz-Universidad Autónoma de Madrid, 28040 Madrid, Spain;
| | - Rodrigo Sanz
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, 28040 Madrid, Spain; (R.S.); (J.D.)
| | - Jana Dziaková
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, 28040 Madrid, Spain; (R.S.); (J.D.)
| | | | - Marta Mendiola
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain; (M.M.); (A.P.-G.)
| | - José Ignacio Casal
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040 Madrid, Spain;
| | - Alberto Peláez-García
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain; (M.M.); (A.P.-G.)
| | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (S.J.d.O.); (R.B.-N.); (R.R.-G.)
| |
Collapse
|
4
|
Montero-Calle A, Garranzo-Asensio M, Rejas-González R, Feliu J, Mendiola M, Peláez-García A, Barderas R. Benefits of FAIMS to Improve the Proteome Coverage of Deteriorated and/or Cross-Linked TMT 10-Plex FFPE Tissue and Plasma-Derived Exosomes Samples. Proteomes 2023; 11:35. [PMID: 37987315 PMCID: PMC10661291 DOI: 10.3390/proteomes11040035] [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: 07/19/2023] [Revised: 09/20/2023] [Accepted: 10/20/2023] [Indexed: 11/22/2023] Open
Abstract
The proteome characterization of complex, deteriorated, or cross-linked protein mixtures as paired clinical FFPE or exosome samples isolated from low plasma volumes (250 µL) might be a challenge. In this work, we aimed at investigating the benefits of FAIMS technology coupled to the Orbitrap Exploris 480 mass spectrometer for the TMT quantitative proteomics analyses of these complex samples in comparison to the analysis of protein extracts from cells, frozen tissue, and exosomes isolated from large volume plasma samples (3 mL). TMT experiments were performed using a two-hour gradient LC-MS/MS with or without FAIMS and two compensation voltages (CV = -45 and CV = -60). In the TMT experiments of cells, frozen tissue, or exosomes isolated from large plasma volumes (3 mL) with FAIMS, a limited increase in the number of identified and quantified proteins accompanied by a decrease in the number of peptides identified and quantified was observed. However, we demonstrated here a noticeable improvement (>100%) in the number of peptide and protein identifications and quantifications for the plasma exosomes isolated from low plasma volumes (250 µL) and FFPE tissue samples in TMT experiments with FAIMS in comparison to the LC-MS/MS analysis without FAIMS. Our results highlight the potential of mass spectrometry analyses with FAIMS to increase the depth into the proteome of complex samples derived from deteriorated, cross-linked samples and/or those where the material was scarce, such as FFPE and plasma-derived exosomes from low plasma volumes (250 µL), which might aid in the characterization of their proteome and proteoforms and in the identification of dysregulated proteins that could be used as biomarkers.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain; (M.G.-A.); (R.R.-G.)
| | - María Garranzo-Asensio
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain; (M.G.-A.); (R.R.-G.)
| | - Raquel Rejas-González
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain; (M.G.-A.); (R.R.-G.)
| | - Jaime Feliu
- Translational Oncology Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain;
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28046 Madrid, Spain;
| | - Marta Mendiola
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28046 Madrid, Spain;
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain;
| | - Alberto Peláez-García
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain;
| | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain; (M.G.-A.); (R.R.-G.)
| |
Collapse
|
5
|
Jiang Y, Zhang Z. OVOL2: an epithelial lineage determiner with emerging roles in energy homeostasis. Trends Cell Biol 2023; 33:824-833. [PMID: 37336658 PMCID: PMC10524639 DOI: 10.1016/j.tcb.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/21/2023]
Abstract
Ovo like zinc finger 2 (OVOL2) is an evolutionarily conserved regulator of epithelial lineage determination and differentiation during embryogenesis. OVOL2 binds to DNA using zinc-finger domains to suppress epithelial-mesenchymal transition (EMT), which is critical for tumor metastasis. However, recent studies have suggested some noncanonical roles of OVOL2 that do not rely on the DNA binding of zinc-finger domains or regulation of EMT. OVOL2 and EMT regulators have emerging roles in adipogenesis, thermogenesis, and lipid metabolism. Here, we review different roles of OVOL2 from embryo development to adult tissue homeostasis, and discuss how OVOL2 and other EMT regulators orchestrate a regulatory network to control energy homeostasis. Last, we propose potential applications of targeting OVOL2 to reduce human obesity.
Collapse
Affiliation(s)
- Yiao Jiang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhao Zhang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
6
|
Cui W, Zhang Q, Wang H, Zhang X, Tian M, Liu D, Yang X. Effects of HOXC8 on the Proliferation and Differentiation of Porcine Preadipocytes. Animals (Basel) 2023; 13:2615. [PMID: 37627406 PMCID: PMC10451666 DOI: 10.3390/ani13162615] [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: 07/14/2023] [Revised: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Transcription factor Homeobox C8 (HOXC8) is identified as a white adipose gene as revealed by expression profile analysis in fat tissues. However, the specific role of HOXC8 in fat accumulation remains to be identified. This study was designed to reveal the effects of HOXC8 on preadipocyte proliferation and differentiation. We first make clear that the expression of HOXC8 is associated with fat contents in muscles, highlighting a role of HOXC8 in fat accumulation. Next, it is demonstrated that HOXC8 promotes the proliferation and differentiation of preadipocytes through gain- and loss-of-function assays in primary cultured porcine preadipocytes. Then, mechanisms underlying the regulation of HOXC8 on preadipocyte proliferation and differentiation are identified with RNA sequencing, and a number of differentially expressed genes (DEGs) in response to HOXC8 knockdown are identified. The top GO (Gene Ontology) terms enriched by DEGs involved in proliferation and differentiation, respectively, are identical. IL-17 signaling pathway is the common one significantly enriched by DEGs involved in preadipocyte proliferation and differentiation, respectively, indicating its importance in mediating fat accumulation regulated by HOXC8. Additionally, we find that the inhibition of proliferation is one of the main processes during preadipocyte differentiation. The results will contribue to further revealing the mechanisms underlying fat accumulation regulated by HOXC8.
Collapse
Affiliation(s)
- Weiguo Cui
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 166319, China
| | - Qian Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Hanqiong Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xiaohan Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Ming Tian
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Di Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Xiuqin Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
7
|
Montero-Calle A, López-Janeiro Á, Mendes ML, Perez-Hernandez D, Echevarría I, Ruz-Caracuel I, Heredia-Soto V, Mendiola M, Hardisson D, Argüeso P, Peláez-García A, Guzman-Aranguez A, Barderas R. In-depth quantitative proteomics analysis revealed C1GALT1 depletion in ECC-1 cells mimics an aggressive endometrial cancer phenotype observed in cancer patients with low C1GALT1 expression. Cell Oncol (Dordr) 2023; 46:697-715. [PMID: 36745330 PMCID: PMC10205863 DOI: 10.1007/s13402-023-00778-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Endometrial cancer (EC) is the most common cancer of the female reproductive organs. Despite the good overall prognosis of most low-grade ECs, FIGO I and FIGO II patients might experience tumor recurrence and worse prognosis. The study of alterations related to EC pathogenesis might help to get insights into underlying mechanisms involved in EC development and progression. METHODS Core tumoral samples were used to investigate the role of C1GALT1 in EC by immunohistochemistry (IHC). ECC-1 cells were used as endometrioid EC model to investigate the effect of C1GALT1 depletion using C1GALT1 specific shRNAs. SILAC quantitative proteomics analyses and cell-based assays, PCR, qPCR, WB, dot-blot and IHC analyses were used to identify, quantify and validate dysregulation of proteins. RESULTS Low C1GALT1 protein expression levels associate to a more aggressive phenotype of EC. Out of 5208 proteins identified and quantified by LC-MS/MS, 100 proteins showed dysregulation (log2fold-change ≥ 0.58 or ≤-0.58) in the cell protein extracts and 144 in the secretome of C1GALT1 depleted ECC-1 cells. Nine dysregulated proteins were validated. Bioinformatics analyses pointed out to an increase in pathways associated with an aggressive phenotype. This finding was corroborated by loss-of-function cell-based assays demonstrating higher proliferation, invasion, migration, colony formation and angiogenesis capacity in C1GALT1 depleted cells. These effects were associated to the overexpression of ANXA1, as demonstrated by ANXA1 transient silencing cell-based assays, and thus, correlating C1GALT and ANXA1 protein expression and biological effects. Finally, the negative protein expression correlation found by proteomics between C1GALT1 and LGALS3 was confirmed by IHC. CONCLUSION C1GALT1 stably depleted ECC-1 cells mimic an EC aggressive phenotype observed in patients and might be useful for the identification and validation of EC markers of progression.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain
| | | | - Marta L Mendes
- Department of Infection and Immunity, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
| | - Daniel Perez-Hernandez
- Department of Infection and Immunity, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
| | - Irene Echevarría
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain
- Biochemistry and Molecular Biology Department, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037, Madrid, Spain
| | | | - Victoria Heredia-Soto
- Translational Oncology, La Paz University Hospital (IdiPAZ), 28046, Madrid, Spain
- Center for Biomedical Research in the Cancer Network (Centro de Investigación Biomédica en Red de Cáncer, CIBERONC), Instituto de Salud Carlos III, 28046, Madrid, Spain
| | - Marta Mendiola
- Center for Biomedical Research in the Cancer Network (Centro de Investigación Biomédica en Red de Cáncer, CIBERONC), Instituto de Salud Carlos III, 28046, Madrid, Spain
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046, Madrid, Spain
| | - David Hardisson
- Department of Pathology, Hospital Universitario La Paz, 28046, Madrid, Spain
- Center for Biomedical Research in the Cancer Network (Centro de Investigación Biomédica en Red de Cáncer, CIBERONC), Instituto de Salud Carlos III, 28046, Madrid, Spain
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046, Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid, 28029, Madrid, Spain
| | - Pablo Argüeso
- Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Alberto Peláez-García
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046, Madrid, Spain.
| | - Ana Guzman-Aranguez
- Biochemistry and Molecular Biology Department, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037, Madrid, Spain.
| | - Rodrigo Barderas
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain.
- Functional Proteomics Unit, UFIEC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain.
| |
Collapse
|
8
|
Besrour-Aouam N, de Los Rios V, Hernández-Alcántara AM, Mohedano ML, Najjari A, López P, Ouzari HI. Proteomic and in silico analyses of dextran synthesis influence on Leuconostoc lactis AV1n adaptation to temperature change. Front Microbiol 2023; 13:1077375. [PMID: 36713162 PMCID: PMC9875047 DOI: 10.3389/fmicb.2022.1077375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
Leuconostoc lactis is found in vegetables, fruits, and meat and is used by the food industry in the preparation of dairy products, wines, and sugars. We have previously demonstrated that the dextransucrase of Lc. lactis (DsrLL) AV1n produces a high-molecular-weight dextran from sucrose, indicating its potential use as a dextran-forming starter culture. We have also shown that this bacterium was able to produce 10-fold higher levels of dextran at 20°C than at 37°C, at the former temperature accompanied by an increase in dsrLL gene expression. However, the general physiological response of Lc. lactis AV1n to cold temperature in the presence of sucrose, leading to increased production of dextran, has not been yet investigated. Therefore, we have used a quantitative proteomics approach to investigate the cold temperature-induced changes in the proteomic profile of this strain in comparison to its proteomic response at 37°C. In total, 337 proteins were found to be differentially expressed at the applied significance criteria (adjusted p-value ≤ 0.05, FDR 5%, and with a fold-change ≥ 1.5 or ≤ 0.67) with 204 proteins overexpressed, among which 13% were involved in protein as well as cell wall, and envelope component biosynthesis including DsrLL. Proteins implicated in cold stress were expressed at a high level at 20°C and possibly play a role in the upregulation of DsrLL, allowing the efficient synthesis of the protein essential for its adaptation to cold. Post-transcriptional regulation of DsrLL expression also seems to take place through the interplay of exonucleases and endonucleases overexpressed at 20°C, which would influence the half-life of the dsrLL transcript. Furthermore, the mechanism of cold resistance of Lc. lactis AV1n seems to be also based on energy saving through a decrease in growth rate mediated by a decrease in carbohydrate metabolism and its orientation toward the production pathways for storage molecules. Thus, this better understanding of the responses to low temperature and mechanisms for environmental adaptation of Lc. lactis could be exploited for industrial use of strains belonging to this species.
Collapse
Affiliation(s)
- Norhane Besrour-Aouam
- Centro de Investigaciones Biológicas Margarita Salas, CIB-CSIC, Madrid, Spain
- Laboratoire Microorganismes et Biomolécules Actives (LR03ES03), Faculté des Sciences de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Vivian de Los Rios
- Centro de Investigaciones Biológicas Margarita Salas, CIB-CSIC, Madrid, Spain
| | | | - Mᵃ Luz Mohedano
- Centro de Investigaciones Biológicas Margarita Salas, CIB-CSIC, Madrid, Spain
| | - Afef Najjari
- Laboratoire Microorganismes et Biomolécules Actives (LR03ES03), Faculté des Sciences de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Paloma López
- Centro de Investigaciones Biológicas Margarita Salas, CIB-CSIC, Madrid, Spain
| | - Hadda-Imene Ouzari
- Laboratoire Microorganismes et Biomolécules Actives (LR03ES03), Faculté des Sciences de Tunis, Université Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
9
|
Montero-Calle A, Gómez de Cedrón M, Quijada-Freire A, Solís-Fernández G, López-Alonso V, Espinosa-Salinas I, Peláez-García A, Fernández-Aceñero MJ, Ramírez de Molina A, Barderas R. Metabolic Reprogramming Helps to Define Different Metastatic Tropisms in Colorectal Cancer. Front Oncol 2022; 12:903033. [PMID: 35957902 PMCID: PMC9358964 DOI: 10.3389/fonc.2022.903033] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/09/2022] [Indexed: 12/02/2022] Open
Abstract
Approximately 25% of colorectal cancer (CRC) patients experience systemic metastases, with the most frequent target organs being the liver and lung. Metabolic reprogramming has been recognized as one of the hallmarks of cancer. Here, metabolic and functional differences between two CRC cells with different metastatic organotropisms (metastatic KM12SM CRC cells to the liver and KM12L4a to the lung when injected in the spleen and in the tail vein of mice) were analysed in comparison to their parental non-metastatic isogenic KM12C cells, for a subsequent investigation of identified metabolic targets in CRC patients. Meta-analysis from proteomic and transcriptomic data deposited in databases, qPCR, WB, in vitro cell-based assays, and in vivo experiments were used to survey for metabolic alterations contributing to their different organotropism and for the subsequent analysis of identified metabolic markers in CRC patients. Although no changes in cell proliferation were observed between metastatic cells, KM12SM cells were highly dependent on oxidative phosphorylation at mitochondria, whereas KM12L4a cells were characterized by being more energetically efficient with lower basal respiration levels and a better redox management. Lipid metabolism-related targets were found altered in both cell lines, including LDLR, CD36, FABP4, SCD, AGPAT1, and FASN, which were also associated with the prognosis of CRC patients. Moreover, CD36 association with lung metastatic tropism of CRC cells was validated in vivo. Altogether, our results suggest that LDLR, CD36, FABP4, SCD, FASN, LPL, and APOA1 metabolic targets are associated with CRC metastatic tropism to the liver or lung. These features exemplify specific metabolic adaptations for invasive cancer cells which stem at the primary tumour.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Functional Proteomics Unit, Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Gómez de Cedrón
- Precision Nutrition and Cancer Program, Molecular Oncology Group, IMDEA Food Institute, Campus of International Excellence (CEI) University Autonomous of Madrid (UAM) + Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Adriana Quijada-Freire
- Precision Nutrition and Cancer Program, Molecular Oncology Group, IMDEA Food Institute, Campus of International Excellence (CEI) University Autonomous of Madrid (UAM) + Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Guillermo Solís-Fernández
- Molecular Imaging and Photonics Division, Chemistry Department, Faculty of Sciences, KU Leuven, Leuven, Belgium
| | - Victoria López-Alonso
- Unidad de Biología Computacional, Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Espinosa-Salinas
- Platform for Clinical Trials in Nutrition and Health (GENYAL), IMDEA Food Institute, Campus of International Excellence (CEI) University Autonomous of Madrid (UAM) + Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Alberto Peláez-García
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), Madrid, Spain
| | - María Jesús Fernández-Aceñero
- Servicio de Anatomía Patológica Hospital Clínico San Carlos, Departamento de Anatomía Patológica, Facultad de Medicina, Complutense University of Madrid, Madrid, Spain
| | - Ana Ramírez de Molina
- Precision Nutrition and Cancer Program, Molecular Oncology Group, IMDEA Food Institute, Campus of International Excellence (CEI) University Autonomous of Madrid (UAM) + Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Rodrigo Barderas
- Functional Proteomics Unit, Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
10
|
Garranzo‐Asensio M, Rodríguez‐Cobos J, San Millán C, Poves C, Fernández‐Aceñero MJ, Pastor‐Morate D, Viñal D, Montero‐Calle A, Solís‐Fernández G, Ceron M, Gámez‐Chiachio M, Rodríguez N, Guzmán‐Aránguez A, Barderas R, Domínguez G. In-depth proteomics characterization of ∆Np73 effectors identifies key proteins with diagnostic potential implicated in lymphangiogenesis, vasculogenesis and metastasis in colorectal cancer. Mol Oncol 2022; 16:2672-2692. [PMID: 35586989 PMCID: PMC9298678 DOI: 10.1002/1878-0261.13228] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 03/17/2022] [Accepted: 05/17/2022] [Indexed: 11/18/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer-related death worldwide. Alterations in proteins of the p53-family are a common event in CRC. ΔNp73, a p53-family member, shows oncogenic properties and its effectors are largely unknown. We performed an in-depth proteomics characterization of transcriptional control by ∆Np73 of the secretome of human colon cancer cells and validated its clinical potential. The secretome was analyzed using high-density antibody microarrays and stable isotopic metabolic labeling. Validation was performed by semiquantitative PCR, ELISA, dot-blot and western blot analysis. Evaluation of selected effectors was carried out using 60 plasma samples from CRC patients, individuals carrying premalignant colorectal lesions and colonoscopy-negative controls. In total, 51 dysregulated proteins were observed showing at least 1.5-foldchange in expression. We found an important association between the overexpression of ∆Np73 and effectors related to lymphangiogenesis, vasculogenesis and metastasis, such as brain-derived neurotrophic factor (BDNF) and the putative aminoacyl tRNA synthase complex-interacting multifunctional protein 1 (EMAP-II)-vascular endothelial growth factor C-vascular endothelial growth factor receptor 3 axis. We further demonstrated the usefulness of BDNF as a potential CRC biomarker able to discriminate between CRC patients and premalignant individuals from controls with high sensitivity and specificity.
Collapse
Affiliation(s)
| | - Javier Rodríguez‐Cobos
- Departamento de Bioquímica, Facultad de MedicinaInstituto de Investigaciones Biomédicas “Alberto Sols”, CSIC‐UAM, IdiPAZMadridSpain
| | - Coral San Millán
- Departamento de Bioquímica, Facultad de MedicinaInstituto de Investigaciones Biomédicas “Alberto Sols”, CSIC‐UAM, IdiPAZMadridSpain
| | - Carmen Poves
- Gastroenterology UnitHospital Universitario Clínico San CarlosMadridSpain
| | | | - Daniel Pastor‐Morate
- Departamento de Bioquímica, Facultad de MedicinaInstituto de Investigaciones Biomédicas “Alberto Sols”, CSIC‐UAM, IdiPAZMadridSpain
| | - David Viñal
- Medical Oncology DepartmentHospital Universitario La PazMadridSpain
| | - Ana Montero‐Calle
- Chronic Disease Programme (UFIEC)Instituto de Salud Carlos IIIMadridSpain
| | | | - María‐Ángeles Ceron
- Surgical Pathology DepartmentHospital Universitario Clínico San CarlosMadridSpain
| | - Manuel Gámez‐Chiachio
- Departamento de Bioquímica, Facultad de MedicinaInstituto de Investigaciones Biomédicas “Alberto Sols”, CSIC‐UAM, IdiPAZMadridSpain
| | - Nuria Rodríguez
- Medical Oncology DepartmentHospital Universitario La PazMadridSpain
| | - Ana Guzmán‐Aránguez
- Departamento de Bioquímica y Biología Molecular, Facultad de Óptica y OptometríaUniversidad Complutense de MadridSpain
| | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC)Instituto de Salud Carlos IIIMadridSpain
| | - Gemma Domínguez
- Departamento de Bioquímica, Facultad de MedicinaInstituto de Investigaciones Biomédicas “Alberto Sols”, CSIC‐UAM, IdiPAZMadridSpain
| |
Collapse
|
11
|
Zhao F, Ke J, Pan W, Pan H, Shen M. Synergistic effects of ISL1 and KDM6B on non-alcoholic fatty liver disease through the regulation of SNAI1. Mol Med 2022; 28:12. [PMID: 35100965 PMCID: PMC8802528 DOI: 10.1186/s10020-021-00428-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 12/20/2021] [Indexed: 11/15/2022] Open
Abstract
Background The increasing incidence of non-alcoholic fatty liver disease (NAFLD) has been reported worldwide, which urges understanding of its pathogenesis and development of more effective therapeutical methods for this chronic disease. In this study, we aimed to investigate the effects of a LIM homeodomain transcription factor, islet1 (ISL1) on NAFLD. Methods Male C57BL/6J mice were fed with a diet high in fat content to produce NAFLD models. These models were then treated with overexpressed ISL1 (oe-ISL1), oe-Lysine-specific demethylase 6B (KDM6B), oe-SNAI1, or short hairpin RNA against SNAI1. We assessed triglyceride and cholesterol contents in the plasma and liver tissues and determined the expressions of ISL1, KDM6B and SNAI1 in liver tissues. Moreover, the in vitro model of lipid accumulation was constructed using fatty acids to explore the in vitro effect of ISL1/KDM6B/SNAI1 in NAFLD. Results The results showed that the expressions of ISL1, KDM6B, and SNAI1 where decreased, but contents of triglyceride and cholesterol increased in mice exposed to high-fat diet. ISL1 inhibited lipogenesis and promoted lipolysis and exhibited a synergizing effect with KDM6B to upregulate the expression of SNAI1. Moreover, both KDM6B and SNAI1 could inhibit lipogenesis and induce lipolysis. Importantly, the therapeutic effects of ISL1 on in vitro model of lipid accumulations was also confirmed through the modulation of KDM6B and SNAI1. Conclusions Taken together, these findings highlighted that ISL1 effectively ameliorated NAFLD by inducing the expressions of KDM6B and SNAI1, which might be a promising drug for the treatment of NAFLD. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00428-7.
Collapse
Affiliation(s)
- Fei Zhao
- Health Management Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Jinjing Ke
- Health Management Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Wensheng Pan
- Health Management Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Hanghai Pan
- Health Management Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Miao Shen
- Health Management Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China.
| |
Collapse
|
12
|
Zhang J, Guo F, Zhou R, Xiang C, Zhang Y, Gao J, Cao G, Yang H. Proteomics and transcriptome reveal the key transcription factors mediating the protection of Panax notoginseng saponins (PNS) against cerebral ischemia/reperfusion injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153613. [PMID: 34500302 DOI: 10.1016/j.phymed.2021.153613] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND AND PURPOSE Transcription factors (TFs) play a critical role in the cerebral ischemia/reperfusion injury (IRI). Panax notoginseng saponins (PNS) are extensively used in the treatment of acute cerebral ischemia in China, but the mechanism of their effects, especially at the TF level, remains unclear. In this study, a combination of transcriptomics, proteomics and network pharmacology analysis was used to identify the key TFs involved in the protection of PNS against middle cerebral artery occlusion (MCAO)-induced IRI. METHODS AND RESULTS Sprague-Dawley rats which were subjected to 1.5 hours of MCAO-induced occlusionand then followed by reperfusion, were treated with PNS at a concentration of 36 mg/kg or 72 mg/kg daily for 7 days. PNS significantly decreased neurological deficient scores and infarction rate; prevented cerebral tissue damage; and reduced CASP3 activity, levels of TNF, IL1B and CCL2 after IRI. Through a combination of transcriptomics and proteomics, 9 critical TFs were identified, including Excision repair cross-complementing group 2 (ERCC2), Nuclear receptor subfamily 4 group A member 3 (NR4A3) and 7 other TFs. The targets of ERCC2 and NR4A3, such as Ubxn11, Ush2a, Numr2, Oxt, Ubxn11, Scrt2, Ttc34 and Lrrc23, were verified by using real-time PCR analysis. RNA-seq analyses indicated that PNS regulated nerve system development and inflammation, and the majority of the identified TFs were also involved in these processes. By using network pharmacology analysis, 73 chemical components in PNS were predicted to affect ERCC2, NR4A3 and 3 other identified TFs. CONCLUSION ERCC2, NR4A3 and 7 other TFs were of importance in the protection of PNS against IRI. This study promoted the understanding of protective mechanism of PNS against cerebral IRI and facilitated the identification of possible targets of PNS.
Collapse
Affiliation(s)
- Jingjing Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Chinese Institute for Brain Research, Beijing, 102206, China
| | - Feifei Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Rui Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Changpei Xiang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jinhuan Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Guangzhao Cao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hongjun Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
13
|
Razmara E, Bitaraf A, Karimi B, Babashah S. Functions of the SNAI family in chondrocyte-to-osteocyte development. Ann N Y Acad Sci 2021; 1503:5-22. [PMID: 34403146 DOI: 10.1111/nyas.14668] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/22/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Different cellular mechanisms contribute to osteocyte development. And while critical roles for members of the zinc finger protein SNAI family (SNAIs) have been discussed in cancer-related models, there are few reviews summarizing their importance for chondrocyte-to-osteocyte development. To help fill this gap, we review the roles of SNAIs in the development of mature osteocytes from chondrocytes, including the regulation of chondro- and osteogenesis through different signaling pathways and in programmed cell death. We also discuss how epigenetic factors-including DNA methylation, histone methylation and acetylation, and noncoding RNAs-contribute differently to both chondrocyte and osteocyte development. To better grasp the important roles of SNAIs in bone development, we also review genotype-phenotype correlations in different animal models. We end with comments about the possible importance of the SNAI family in cartilage/bone development and the potential applications for therapeutic goals.
Collapse
Affiliation(s)
- Ehsan Razmara
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Behnaz Karimi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
14
|
An YZ, Cho E, Ling J, Zhang X. The Axin2-snail axis promotes bone invasion by activating cancer-associated fibroblasts in oral squamous cell carcinoma. BMC Cancer 2020; 20:987. [PMID: 33046030 PMCID: PMC7552517 DOI: 10.1186/s12885-020-07495-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
Background In bone-invasive oral squamous cell carcinoma (OSCC), cancer-associated fibroblasts (CAFs) infiltrate into bony tissue ahead of OSCC cells. In the present study, we aimed to investigate the role of the Axin2-Snail axis in the biological behaviour of CAFs and bone invasion in OSCC. Methods The clinicopathological significance of Axin2 and Snail expression was investigated by immunohistochemistry in an OSCC cohort containing 217 tissue samples from patients with long-term follow-up. The influence of the Axin2-Snail axis on the biological behaviour of OSCC cells and CAFs was further investigated both in vitro and in vivo. Results Axin2 expression was significantly associated with Snail expression, the desmoplasia status, and bone invasion in patients with OSCC. In multivariate analysis, lymph node metastasis, desmoplasia, Axin2 expression, and Snail expression were independent poor prognostic factors in our cohort. Consistent with these findings, OSCC cells demonstrated attenuated oncogenic activity as well as decreased expression of Snail and various cytokines after Axin2 knockdown in vitro. Among the related cytokines, C-C motif chemokine ligand 5 (CCL5) and interleukin 8 (IL8) demonstrated a strong influence on the biological behaviour of CAFs in vitro. Moreover, both the desmoplastic reaction and osteolytic lesions in the calvaria were predominantly decreased after Axin2 knockdown in OSCC cells in vivo using a BALB/c athymic nude mouse xenograft model. Conclusions Oncogenic activities of the Axin2-Snail axis are not limited to the cancer cells themselves but rather extend to CAFs via regulation of the cytokine-mediated cancer-stromal interaction, with further implications for bone invasion as well as a poor prognosis in OSCC.
Collapse
Affiliation(s)
- Yin-Zhe An
- Key laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Eunae Cho
- Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, Republic of Korea.,BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, South Korea.,Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Junqi Ling
- Key laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China. .,Department of Endodontics, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, 56 Lingyuanxi Road, Guangzhou, 510055, Guangdong, China.
| | - Xianglan Zhang
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Republic of Korea. .,Department of Pathology, Yanbian University Hospital, Yanji City, 133000, Jilin Province, China.
| |
Collapse
|
15
|
Garranzo-Asensio M, San Segundo-Acosta P, Povés C, Fernández-Aceñero MJ, Martínez-Useros J, Montero-Calle A, Solís-Fernández G, Sanchez-Martinez M, Rodríguez N, Cerón MÁ, Fernandez-Diez S, Domínguez G, de Los Ríos V, Peláez-García A, Guzmán-Aránguez A, Barderas R. Identification of tumor-associated antigens with diagnostic ability of colorectal cancer by in-depth immunomic and seroproteomic analysis. J Proteomics 2020; 214:103635. [PMID: 31918032 DOI: 10.1016/j.jprot.2020.103635] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/02/2020] [Accepted: 01/05/2020] [Indexed: 12/18/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer related death worldwide. Its diagnosis at early stages would significantly improve the survival of CRC patients. The humoral immune response has been demonstrated useful for cancer diagnosis, predating clinical symptoms up to 3 years. Here, we employed an in-depth seroproteomic approach to identify proteins that elicit a humoral immune response in CRC patients. The seroproteomic approach relied on the immunoprecipitation with patient-derived autoantibodies of proteins from CRC cell lines with different metastatic properties followed by LC-MS/MS. After bioinformatics, we focused on 31 targets of CRC autoantibodies. After WB and IHC validation, ERP44 and TALDO1 showed potential to discriminate disease-free and metastatic CRC patients, and time to recurrence of CRC patients in stage II. Using plasma samples of 30 healthy individuals, 28 premalignant individuals, and 32 CRC patients, nine out of 13 selected targets for seroreactive analysis showed significant diagnostic ability to discriminate either CRC patients or premalignant subjects from controls. Our results suggest that the here defined panel of CRC autoantibodies and their target proteins should be included in CRC blood-based biomarker panels to get a clinically useful blood-based diagnostic signature for CRC detection. SIGNIFICANCE: Colorectal cancer is one of the deadliest cancer types mainly due to its late diagnosis. Its early diagnosis, therefore, is of great importance since it would significantly improve the survival of CRC patients. In our work, the in-depth seroproteomic analysis of colorectal cancer using isolated IgGs from colorectal cancer patients and controls and protein extract of colorectal cancer cells provide the identification of valuable biomarkers with diagnostic and prognostic ability of the disease.
Collapse
Affiliation(s)
- María Garranzo-Asensio
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, Majadahonda E-28220, Madrid, Spain
| | - Pablo San Segundo-Acosta
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, Majadahonda E-28220, Madrid, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Carmen Povés
- Gastroenterology Unit, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | | | - Javier Martínez-Useros
- Translational Oncology Division, OncoHealth Institute, Fundacion Jimenez Diaz University Hospital, E-28040 Madrid, Spain
| | - Ana Montero-Calle
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, Majadahonda E-28220, Madrid, Spain
| | | | | | - Nuria Rodríguez
- Medical Oncology Department, Hospital Universitario La Paz, E-28046 Madrid, Spain
| | - María Ángeles Cerón
- Surgical Pathology Department, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | | | - Gemma Domínguez
- Departamento de Medicina, Facultad de Medicina, Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, E-28029 Madrid, Spain
| | | | | | - Ana Guzmán-Aránguez
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Rodrigo Barderas
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, Majadahonda E-28220, Madrid, Spain.
| |
Collapse
|
16
|
Valverde A, ben Hassine A, Serafín V, Muñoz‐San Martín C, Pedrero M, Garranzo‐Asensio M, Gamella M, Raouafi N, Barderas R, Yáñez‐Sedeño P, Campuzano S, Pingarrón JM. Dual Amperometric Immunosensor for Improving Cancer Metastasis Detection by the Simultaneous Determination of Extracellular and Soluble Circulating Fraction of Emerging Metastatic Biomarkers. ELECTROANAL 2019. [DOI: 10.1002/elan.201900506] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alejandro Valverde
- Analytical Chemistry Dept., Faculty of ChemistryComplutense University of Madrid. E-28040 Madrid Spain
| | - Amira ben Hassine
- Analytical Chemistry Dept., Faculty of ChemistryComplutense University of Madrid. E-28040 Madrid Spain
- University of Tunis El ManarTunis Faculty of Science, Sensors and Biosensors Group, Laboratory of Analytical Chemistry and Electrochemistry 2092 Tunis El Manar Tunisia
| | - Verónica Serafín
- Analytical Chemistry Dept., Faculty of ChemistryComplutense University of Madrid. E-28040 Madrid Spain
| | - Cristina Muñoz‐San Martín
- Analytical Chemistry Dept., Faculty of ChemistryComplutense University of Madrid. E-28040 Madrid Spain
| | - María Pedrero
- Analytical Chemistry Dept., Faculty of ChemistryComplutense University of Madrid. E-28040 Madrid Spain
| | | | - Maria Gamella
- Analytical Chemistry Dept., Faculty of ChemistryComplutense University of Madrid. E-28040 Madrid Spain
| | - Noureddine Raouafi
- University of Tunis El ManarTunis Faculty of Science, Sensors and Biosensors Group, Laboratory of Analytical Chemistry and Electrochemistry 2092 Tunis El Manar Tunisia
| | - Rodrigo Barderas
- UFIECInstitute of Health Carlos III. E-28220 Majadahonda, Madrid Spain
| | - Paloma Yáñez‐Sedeño
- Analytical Chemistry Dept., Faculty of ChemistryComplutense University of Madrid. E-28040 Madrid Spain
| | - Susana Campuzano
- Analytical Chemistry Dept., Faculty of ChemistryComplutense University of Madrid. E-28040 Madrid Spain
| | - José M. Pingarrón
- Analytical Chemistry Dept., Faculty of ChemistryComplutense University of Madrid. E-28040 Madrid Spain
| |
Collapse
|
17
|
San Segundo-Acosta P, Oeo-Santos C, Benedé S, de Los Ríos V, Navas A, Ruiz-Leon B, Moreno C, Pastor-Vargas C, Jurado A, Villalba M, Barderas R. Delineation of the Olive Pollen Proteome and Its Allergenome Unmasks Cyclophilin as a Relevant Cross-Reactive Allergen. J Proteome Res 2019; 18:3052-3066. [PMID: 31192604 DOI: 10.1021/acs.jproteome.9b00167] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Olive pollen is a major allergenic source worldwide due to its extensive cultivation. We have combined available genomics data with a comprehensive proteomics approach to get the annotated olive tree (Olea europaea L.) pollen proteome and define its complex allergenome. A total of 1907 proteins were identified by LC-MS/MS using predicted protein sequences from its genome. Most proteins (60%) were predicted to possess catalytic activity and be involved in metabolic processes. In total, 203 proteins belonging to 47 allergen families were found in olive pollen. A peptidyl-prolyl cis-trans isomerase, cyclophilin, produced in Escherichia coli, was found as a new olive pollen allergen (Ole e 15). Most Ole e 15-sensitized patients were children (63%) and showed strong IgE recognition to the allergen. Ole e 15 shared high sequence identity with other plant, animal, and fungal cyclophilins and presented high IgE cross-reactivity with pollen, plant food, and animal extracts.
Collapse
Affiliation(s)
- Pablo San Segundo-Acosta
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas , Universidad Complutense de Madrid , E-28040 Madrid , Spain
| | - Carmen Oeo-Santos
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas , Universidad Complutense de Madrid , E-28040 Madrid , Spain
| | - Sara Benedé
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas , Universidad Complutense de Madrid , E-28040 Madrid , Spain
| | | | - Ana Navas
- Hospital Universitario Reina Sofía de Córdoba , E-14004 Córdoba , Spain
| | - Berta Ruiz-Leon
- Hospital Universitario Reina Sofía de Córdoba , E-14004 Córdoba , Spain
| | - Carmen Moreno
- Hospital Universitario Reina Sofía de Córdoba , E-14004 Córdoba , Spain
| | - Carlos Pastor-Vargas
- Department of Immunology , Instituto de Investigación Sanitaria Hospital Universitario Fundación Jiménez Díaz (IIS-FJD, UAM) , E-28040 Madrid , Spain
| | - Aurora Jurado
- Hospital Universitario Reina Sofía de Córdoba , E-14004 Córdoba , Spain
| | - Mayte Villalba
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas , Universidad Complutense de Madrid , E-28040 Madrid , Spain
| | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC) , Instituto de Salud Carlos III , Majadahonda, E-28220 Madrid , Spain
| |
Collapse
|
18
|
Banfai K, Ernszt D, Pap A, Bai P, Garai K, Belharazem D, Pongracz JE, Kvell K. "Beige" Cross Talk Between the Immune System and Metabolism. Front Endocrinol (Lausanne) 2019; 10:369. [PMID: 31275241 PMCID: PMC6591453 DOI: 10.3389/fendo.2019.00369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/24/2019] [Indexed: 12/25/2022] Open
Abstract
With thymic senescence the epithelial network shrinks to be replaced by adipose tissue. Transcription factor TBX-1 controls thymus organogenesis, however, the same TBX-1 has also been reported to orchestrate beige adipose tissue development. Given these different roles of TBX-1, we have assessed if thymic TBX-1 expression persists and demonstrates this dualism during adulthood. We have also checked whether thymic adipose involution could yield beige adipose tissue. We have used adult mouse and human thymus tissue from various ages to evaluate the kinetics of TBX-1 expression, as well as mouse (TEP1) and human (1889c) thymic epithelial cells (TECs) for our studies. Electron micrographs show multi-locular lipid deposits typical of beige adipose cells. Histology staining shows the accumulation of neutral lipid deposits. qPCR measurements show persistent and/or elevating levels of beige-specific and beige-indicative markers (TBX-1, EAR-2, UCP-1, PPAR-gamma). We have performed miRNome profiling using qPCR-based QuantStudio platform and amplification-free NanoString platform. We have observed characteristic alterations, including increased miR21 level (promoting adipose tissue development) and decreased miR34a level (bias toward beige adipose tissue differentiation). Finally, using the Seahorse metabolic platform we have recorded a metabolic profile (OCR/ECAR ratio) indicative of beige adipose tissue. In summary, our results support that thymic adipose tissue emerging with senescence is bona fide beige adipose tissue. Our data show how the borders blur between a key immune tissue (the thymus) and a key metabolic tissue (beige adipose tissue) with senescence. Our work contributes to the understanding of cross talk between the immune system and metabolism.
Collapse
Affiliation(s)
- Krisztina Banfai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
| | - David Ernszt
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
- Department of Physiology, Medical School, University of Pécs, Pécs, Hungary
| | - Attila Pap
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Bai
- Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, Debrecen, Hungary
- MTA-DE Lendulet Laboratory of Cellular Metabolism, Debrecen, Hungary
- Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Kitti Garai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
| | - Djeda Belharazem
- Department of Pathology, University Hospital of Mannheim, Mannheim, Germany
| | - Judit E. Pongracz
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
| | - Krisztian Kvell
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
- *Correspondence: Krisztian Kvell
| |
Collapse
|
19
|
A recombinant isoform of the Ole e 7 olive pollen allergen assembled by de novo mass spectrometry retains the allergenic ability of the natural allergen. J Proteomics 2018; 187:39-46. [DOI: 10.1016/j.jprot.2018.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/29/2018] [Accepted: 06/03/2018] [Indexed: 01/24/2023]
|
20
|
Samblas M, Milagro FI, Mansego ML, Marti A, Martinez JA. PTPRS and PER3 methylation levels are associated with childhood obesity: results from a genome-wide methylation analysis. Pediatr Obes 2018; 13:149-158. [PMID: 28614626 DOI: 10.1111/ijpo.12224] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 04/07/2017] [Accepted: 05/01/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND The global prevalence of childhood overweight and obesity has increased in the last years. Epigenetic dysregulation affecting gene expression could be a determinant in early-life obesity onset and accompanying complications. OBJECTIVE The aim of the present investigation was to analyse the putative association between DNA methylation and childhood obesity. METHODS DNA was isolated from white blood cells of 24 children obtained from the GENOI study and was hybridized in a 450K methylation array. Two CpG sites associated with obesity were validated in 91 children by MassArray® EpiTyper™ technology. RESULTS Genome-wide analysis identified 734 CpGs (783 genes) differentially methylated between cases (n = 12) and controls (n = 12). Ingenuity Pathway Analysis showed that these genes were involved in oxidative stress and circadian rhythm signalling pathways. Moreover, the DNA methylation levels of VIPR2, GRIN2D, ADCYAP1R1, PER3 and PTPRS regions correlated with the obesity trait. EpiTyper™ validation also identified significant correlations between methylation levels of CpG sites on PTPRS and PER3 with BMI z-score. CONCLUSIONS This study identified several CpG sites and specifically several CpGs in the PTPRS and PER3 genes differentially methylated between obese and non-obese children, suggesting a role for DNA methylation concerning development of childhood obesity.
Collapse
Affiliation(s)
- M Samblas
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - F I Milagro
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain.,CIBERobn, Physiopathology of Obesity, Carlos III Institute, Madrid, Spain
| | - M L Mansego
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - A Marti
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,CIBERobn, Physiopathology of Obesity, Carlos III Institute, Madrid, Spain.,IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
| | - J A Martinez
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain.,CIBERobn, Physiopathology of Obesity, Carlos III Institute, Madrid, Spain.,IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
| | | |
Collapse
|
21
|
Garranzo-Asensio M, San Segundo-Acosta P, Martínez-Useros J, Montero-Calle A, Fernández-Aceñero MJ, Häggmark-Månberg A, Pelaez-Garcia A, Villalba M, Rabano A, Nilsson P, Barderas R. Identification of prefrontal cortex protein alterations in Alzheimer's disease. Oncotarget 2018; 9:10847-10867. [PMID: 29541381 PMCID: PMC5834268 DOI: 10.18632/oncotarget.24303] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 01/13/2018] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia in developed countries. A better understanding of the events taking place at the molecular level would help to identify novel protein alterations, which might be used in diagnosis or for treatment development. In this study, we have performed the high-throughput analysis of 706 molecules mostly implicated in cell-cell communication and cell signaling processes by using two antibody microarray platforms. We screened three AD pathological groups -each one containing four pooled samples- from Braak stages IV, V and VI, and three control groups from two healthy subjects, five frontotemporal and two vascular dementia patients onto Panorama and L-Series antibody microarrays to identify AD-specific alterations not common to other dementias. Forty altered proteins between control and AD groups were detected, and validated by i) meta-analysis of mRNA alterations, ii) WB, and iii) FISH and IHC using an AD-specific tissue microarray containing 44 samples from AD patients at different Braak stages, and frontotemporal and vascular dementia patients and healthy individuals as controls. We identified altered proteins in AD not common to other dementias like the E3 ubiquitin-protein ligase TOPORS, Layilin and MICB, and validated the association to AD of the previously controverted proteins DDIT3 and the E3 ubiquitin-protein ligase XIAP. These altered proteins constitute interesting targets for further immunological analyses using sera, plasma and CSF to identify AD blood- or cerebrospinal fluid-biomarkers and to perform functional analysis to determine their specific role in AD, and their usefulness as potential therapeutic targets of intervention.
Collapse
Affiliation(s)
- Maria Garranzo-Asensio
- Biochemistry and Molecular Biology Department I, Chemistry Faculty, Complutense University of Madrid, Madrid, Spain
| | - Pablo San Segundo-Acosta
- Biochemistry and Molecular Biology Department I, Chemistry Faculty, Complutense University of Madrid, Madrid, Spain
| | - Javier Martínez-Useros
- Translational Oncology Division, OncoHealth Institute, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
| | - Ana Montero-Calle
- Biochemistry and Molecular Biology Department I, Chemistry Faculty, Complutense University of Madrid, Madrid, Spain
| | - María Jesús Fernández-Aceñero
- Servicio de Anatomía Patológica Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Departamento de Anatomía Patològica, Facultad de Medicina, Complutense University of Madrid, Madrid, Spain
| | - Anna Häggmark-Månberg
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH – Royal Institute of Technology, Stockholm, Sweden
| | | | - Mayte Villalba
- Biochemistry and Molecular Biology Department I, Chemistry Faculty, Complutense University of Madrid, Madrid, Spain
| | - Alberto Rabano
- Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| | - Peter Nilsson
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Rodrigo Barderas
- Biochemistry and Molecular Biology Department I, Chemistry Faculty, Complutense University of Madrid, Madrid, Spain
- UFIEC, National Institute of Health Carlos III, Majadahonda, Madrid, Spain
| |
Collapse
|
22
|
Fang S, Huang Y, Zhong S, Li Y, Zhang Y, Li Y, Sun J, Liu X, Wang Y, Zhang S, Xu T, Sun X, Gu P, Li D, Zhou H, Li B, Fan X. Regulation of Orbital Fibrosis and Adipogenesis by Pathogenic Th17 Cells in Graves Orbitopathy. J Clin Endocrinol Metab 2017; 102:4273-4283. [PMID: 28938397 DOI: 10.1210/jc.2017-01349] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022]
Abstract
CONTEXT T helper (Th)17 cells are correlated with many human autoimmune disorders, including Graves disease, and may play key roles in the pathogenesis of Graves orbitopathy (GO). OBJECTIVE To study the phenotype of Th17 cells in patients with GO and healthy subjects, investigate the fibrosis and adipogenesis in orbital fibroblasts (OFs) modulated by interleukin (IL)-17A, and determine the interaction between Th17 cells and OFs. DESIGN/SETTING/PARTICIPANTS Blood samples and orbital tissues from GO patients and healthy controls were collected. MAIN OUTCOME MEASURES We conducted multicolor flow cytometry, immunohistochemical and immunofluorescent stainings, Western blotting, a PathScan intracellular signaling assay, Luminex and enzyme-linked immunosorbent assays, and protein mass spectrum. RESULTS Interferon-γ- and IL-22-expressing Th17 cells are increased in GO patients, which are positively related to clinical activity score. Costimulatory molecules are highly expressed in GO orbits and most GO OFs are CD90+. IL-17A promotes TGF-β-induced fibrosis in CD90+ OFs but impedes 15-deoxy-Δ12,14-prostaglandin J2-induced adipogenesis in CD90- OFs. Th17 cells promote proinflammatory cytokine secretion in both CD90+ and CD90- OFs. Meanwhile, both CD90+ and CD90- OFs contribute to Th17 cell differentiation through prostaglandin E2 production, which can be attenuated by indomethacin. Furthermore, Th17 cells upregulate costimulatory molecule expression on OFs. CONCLUSION Our findings unravel the pathogenicity of IL-17A in the initiation and progression of GO. In-depth interpretation of the molecular basis of OFs delineated by CD90 and Th17-OF interaction will help to afford a novel approach to better therapeutic strategies for GO.
Collapse
Affiliation(s)
- Sijie Fang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Yazhuo Huang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Sisi Zhong
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Yangyang Li
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Yidan Zhang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Yinwei Li
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Jing Sun
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Xingtong Liu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Yang Wang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Shuo Zhang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Tianle Xu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai First People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Ping Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Dan Li
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Huifang Zhou
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
23
|
Mendes M, Peláez-García A, López-Lucendo M, Bartolomé RA, Calviño E, Barderas R, Casal JI. Mapping the Spatial Proteome of Metastatic Cells in Colorectal Cancer. Proteomics 2017; 17. [DOI: 10.1002/pmic.201700094] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 07/28/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Marta Mendes
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| | - Alberto Peláez-García
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| | - María López-Lucendo
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| | - Rubén A. Bartolomé
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| | - Eva Calviño
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| | - Rodrigo Barderas
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
- Instituto de Salud Carlos III.; Majadahonda Spain
| | - J. Ignacio Casal
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| |
Collapse
|
24
|
Mas S, Oeo-Santos C, Cuesta-Herranz J, Díaz-Perales A, Colás C, Fernández J, Barber D, Rodríguez R, de los Ríos V, Barderas R, Villalba M. A relevant IgE-reactive 28 kDa protein identified from Salsola kali pollen extract by proteomics is a natural degradation product of an integral 47 kDa polygalaturonase. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:1067-1076. [DOI: 10.1016/j.bbapap.2017.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 04/07/2017] [Accepted: 05/08/2017] [Indexed: 12/29/2022]
|
25
|
Hernandez JL, Davda D, Majmudar JD, Won SJ, Prakash A, Choi AI, Martin BR. Correlated S-palmitoylation profiling of Snail-induced epithelial to mesenchymal transition. MOLECULAR BIOSYSTEMS 2017; 12:1799-808. [PMID: 27030425 DOI: 10.1039/c6mb00019c] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Epithelial cells form spatially-organized adhesion complexes that establish polarity gradients, regulate cell proliferation, and direct wound healing. As cells accumulate oncogenic mutations, these key tumor suppression mechanisms are disrupted, eliminating many adhesion complexes and bypassing contact inhibition. The transcription factor Snail is often expressed in malignant cancers, where it promotes transcriptional reprogramming to drive epithelial-mesenchymal transition (EMT) and establishes a more invasive state. S-Palmitoylation describes the fatty-acyl post-translational modification of cysteine residues in proteins, and is required for membrane anchoring, trafficking, localization and function of hundreds of proteins involved in cell growth, polarity, and signaling. Since Snail-expression disrupts apico-basolateral cell polarity, we asked if Snail-dependent transformation induces proteome-wide changes in S-palmitoylation. MCF10A breast cancer cells were retrovirally transduced with Snail and correlated proteome-wide changes in protein abundance and S-palmitoylation were profiled by using stable isotope labeling in cell culture with amino acid (SILAC) mass spectrometry. This analysis identified increased levels of proteins involved in migration, glycolysis, and cell junction remodeling, and decreased levels of proteins involved in cell adhesion. Overall, protein S-palmitoylation is highly correlated with protein abundance, yet for a subset of proteins, this correlation is uncoupled. These findings suggest that Snail-overexpression affects the S-palmitoylation cycle of some proteins, which may participate in cell polarity and tumor suppression.
Collapse
Affiliation(s)
- Jeannie L Hernandez
- Department of Chemistry, University of Michigan, 930 N. University Ave., Ann Arbor, MI 48109, USA.
| | - Dahvid Davda
- Department of Chemistry, University of Michigan, 930 N. University Ave., Ann Arbor, MI 48109, USA. and Program in Chemical Biology, University of Michigan, 930 N. University Ave., Ann Arbor, MI 48109, USA
| | - Jaimeen D Majmudar
- Department of Chemistry, University of Michigan, 930 N. University Ave., Ann Arbor, MI 48109, USA.
| | - Sang Joon Won
- Program in Chemical Biology, University of Michigan, 930 N. University Ave., Ann Arbor, MI 48109, USA
| | - Ashesh Prakash
- Department of Chemistry, University of Michigan, 930 N. University Ave., Ann Arbor, MI 48109, USA.
| | - Alexandria I Choi
- Department of Chemistry, University of Michigan, 930 N. University Ave., Ann Arbor, MI 48109, USA.
| | - Brent R Martin
- Department of Chemistry, University of Michigan, 930 N. University Ave., Ann Arbor, MI 48109, USA. and Program in Chemical Biology, University of Michigan, 930 N. University Ave., Ann Arbor, MI 48109, USA
| |
Collapse
|
26
|
IL-17 axis accelerates the inflammatory progression of obese in mice via TBK1 and IKBKE pathway. Immunol Lett 2017; 184:67-75. [PMID: 28237848 DOI: 10.1016/j.imlet.2017.02.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/04/2017] [Accepted: 02/09/2017] [Indexed: 02/06/2023]
Abstract
Obesity mediates immune inflammatory response and induces IL-17 expression. Adipgenesis can be regulated by IL-17 and it causes TBK1 activation. The inhibition of TBK1 and the inhibition of I IKBKE reduces inflammatory response and improves obesity. It is hypothesized that IL-17 deficiency inhibits obesity progression and inflammation. 3T3-L1 preadipocytes were differentiated in vitro and treated with IL-17. RAW264.7 cells and differentiated 3T3-L1 were pretreated with TBK1 inhibitor and then stimulated with IL-17. Wild-type and IL-17 knock out mice were fed with high-fat diet. IL-17 inhibits adipocyte differentiation from mouse-derived 3T3-L1 preadipocytes and reduces mRNA expression of proadipogenic transcription factors and adipokines in adipocyte cells. IL-17 also showed up-regulation of mRNA levels of inflammatory cytokines in RAW cells. The inhibitor of TBK1 and IKBKE attenuates the effect of IL-17. Loss of IL-17 deficiency improves diet-induced obesity, fatty liver, glucose and lipid metabolism in mice. The expression of TBK1 and IKBKE decreased in the spleen and liver of IL-17 deficiency mice. Moreover, the inflammatory response within the visceral adipose tissue and Th1 cells were inhibited, however, M2 macrophage and Th2 cells increased in IL-17 deficiency mice. IL-17 inhibits adipogenesis where a lack of IL-17 ameliorates glucose metabolism. As well, the inhibition of TBK1 reduces inflammation induced by IL-17. Therefore, IL-17 may be involved in the development of obesity and metabolic dysfunction in a TBK1-dependent manner.
Collapse
|
27
|
Plikus MV, Guerrero-Juarez CF, Ito M, Li YR, Dedhia PH, Zheng Y, Shao M, Gay DL, Ramos R, Hsi TC, Oh JW, Wang X, Ramirez A, Konopelski SE, Elzein A, Wang A, Supapannachart RJ, Lee HL, Lim CH, Nace A, Guo A, Treffeisen E, Andl T, Ramirez RN, Murad R, Offermanns S, Metzger D, Chambon P, Widgerow AD, Tuan TL, Mortazavi A, Gupta RK, Hamilton BA, Millar SE, Seale P, Pear WS, Lazar MA, Cotsarelis G. Regeneration of fat cells from myofibroblasts during wound healing. Science 2017; 355:748-752. [PMID: 28059714 DOI: 10.1126/science.aai8792] [Citation(s) in RCA: 423] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/19/2016] [Indexed: 12/14/2022]
Abstract
Although regeneration through the reprogramming of one cell lineage to another occurs in fish and amphibians, it has not been observed in mammals. We discovered in the mouse that during wound healing, adipocytes regenerate from myofibroblasts, a cell type thought to be differentiated and nonadipogenic. Myofibroblast reprogramming required neogenic hair follicles, which triggered bone morphogenetic protein (BMP) signaling and then activation of adipocyte transcription factors expressed during development. Overexpression of the BMP antagonist Noggin in hair follicles or deletion of the BMP receptor in myofibroblasts prevented adipocyte formation. Adipocytes formed from human keloid fibroblasts either when treated with BMP or when placed with human hair follicles in vitro. Thus, we identify the myofibroblast as a plastic cell type that may be manipulated to treat scars in humans.
Collapse
Affiliation(s)
- Maksim V Plikus
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA. .,Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Mayumi Ito
- The Ronald O. Perelman Department of Dermatology, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Yun Rose Li
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Priya H Dedhia
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying Zheng
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mengle Shao
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Denise L Gay
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,INSERM U967, Commissariat à L'énergie Atomique et aux Énergies Alternatives, Institut de Radiobiologie Cellulaire et Moléculaire 92265 Fontenay-aux-Roses Cedex, France
| | - Raul Ramos
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Tsai-Ching Hsi
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Ji Won Oh
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.,Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Xiaojie Wang
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Amanda Ramirez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Sara E Konopelski
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Arijh Elzein
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Anne Wang
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rarinthip June Supapannachart
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hye-Lim Lee
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Chae Ho Lim
- The Ronald O. Perelman Department of Dermatology, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Arben Nace
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amy Guo
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elsa Treffeisen
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 328116, USA
| | - Ricardo N Ramirez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Rabi Murad
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964, Université de Strasbourg, Illkirch 67404, France
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964, Institut d'Etudes Avancées de l'Université de Strasbourg, Collège de France, Illkirch 67404, France
| | - Alan D Widgerow
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, Irvine, CA 92868, USA
| | - Tai-Lan Tuan
- The Saban Research Institute of Children's Hospital Los Angeles and Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bruce A Hamilton
- Departments of Medicine and Cellular and Molecular Medicine, Moores Cancer Center and Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sarah E Millar
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick Seale
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Warren S Pear
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitchell A Lazar
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - George Cotsarelis
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
28
|
|
29
|
Herrera A, Herrera M, Peña C. The emerging role of Snail1 in the tumor stroma. Clin Transl Oncol 2015; 18:872-7. [PMID: 26687368 DOI: 10.1007/s12094-015-1474-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 12/08/2015] [Indexed: 01/11/2023]
Abstract
The transcription factor Snail1 leads to the epithelial-mesenchymal transition by repressing the adherent and tight junctions in epithelial cells. This process is related to an increase of cell migratory and mesenchymal properties during both embryonic development and tumor progression. Although Snail1 expression is very limited in adult animals, emerging evidence has placed Snail at the forefront of medical science. As a transcriptional repressor, Snail1 confers cancer stem cell-like traits on tumor cells and promotes drug resistance, tumor recurrence and metastasis. In this review, we summarize recent reports that suggest the pro-tumorigenic roles of Snail1 expression in tumor stroma. The crosstalk between tumor and stromal cells mediated by Snail1 regulates paracrine communication, pro-tumorigenic abilities of cancer cells, extracellular matrix characteristics and mesenchymal differentiation in cancer stem cells and cancer-associated fibroblasts. Therefore, understanding the regulation and functional roles of Snail1 in the tumor microenvironment will provide us with new therapies for treating metastatic disease.
Collapse
Affiliation(s)
- A Herrera
- "Cancer Cell Signaling" Research Group, Hospital Universitario Puerta de Hierro de Majadahonda, Majadahonda, Madrid, Spain
| | - M Herrera
- "Cancer Cell Signaling" Research Group, Hospital Universitario Puerta de Hierro de Majadahonda, Majadahonda, Madrid, Spain
| | - C Peña
- "Cancer Cell Signaling" Research Group, Hospital Universitario Puerta de Hierro de Majadahonda, Majadahonda, Madrid, Spain.
| |
Collapse
|
30
|
Peláez-García A, Barderas R, Mendes M, Lopez-Lucendo M, Sanchez JC, García de Herreros A, Casal JI. Data from proteomic characterization of the role of Snail1 in murine mesenchymal stem cells and 3T3-L1 fibroblasts differentiation. Data Brief 2015; 4:606-13. [PMID: 26322327 PMCID: PMC4543208 DOI: 10.1016/j.dib.2015.07.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 07/13/2015] [Accepted: 07/20/2015] [Indexed: 02/02/2023] Open
Abstract
The transcription factor (TF) Snail1 is a major inducer of the epithelial–mesenchymal transition (EMT) during embryonic development and cancer progression. Ectopic expression of Snail in murine mesenchymal stem cells (mMSC) abrogated their differentiation to osteoblasts or adipocytes. We used either stable isotopic metabolic labeling (SILAC) for 3T3-L1 cells or isobaric labeling with tandem mass tags (TMT) for mMSC stably transfected cells with Snail1 or control. We carried out a proteomic analysis on the nuclear fraction since Snail is a nuclear TF that mediates its effects mainly through the regulation of other TFs. Proteomics data have been deposited in ProteomeXchange via the PRIDE partner repository with the dataset identifiers PXD001529 and PXD002157 (Vizcaino et al., 2014) [1]. Data are associated with a research article published in Molecular and Cellular Proteomics (Pelaez-Garcia et al., 2015) [2].
Collapse
Affiliation(s)
- A Peláez-García
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - R Barderas
- Biochemistry and Molecular Biology I Department, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - M Mendes
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - M Lopez-Lucendo
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - J C Sanchez
- Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - J I Casal
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| |
Collapse
|