1
|
Hong Y, Xu L, Yu X, He Y, Du Y. DLK1 regulates periodontal inflammation by inhibiting NF-κB p65 and JNK signaling pathways. Odontology 2024:10.1007/s10266-024-00979-1. [PMID: 38995322 DOI: 10.1007/s10266-024-00979-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024]
Abstract
The roles and molecular mechanisms of Delta-like 1 (DLK1) in periodontitis remain largely unknown. Here, we investigated the expression of DLK1 and NF-κB p65 in Porphyromonas gingivalis (Pg.)-induced periodontitis in vivo. Periodontal inflammation and alveolar bone resorption were analyzed using western blotting, micro-computed tomography, TRAP staining, immunohistochemistry, and immunofluorescence. Raw246.7 cells were stimulated with 1 μg/ml Porphyromonas gingivalis lipopolysaccharide (Pg.LPS) to assess DLK1 expression in vitro. DLK1 overexpression was achieved, and transfection efficiency was confirmed using western blotting and immunofluorescence. The NF-κB and MAPK pathways were activated by treating cells with 1 μg/ml Pg.LPS to explore related mechanisms. Compared with normal tissues, both DLK1 and NF-κB p65 expression increased in periodontitis gingival tissues. DLK1-positive expression was observed in inflammatory infiltrating cells and osteoclasts in the marginal lacunae of the alveolar bone. DLK1 expression in CD68-positive macrophages was detected by immunofluorescence. However, DLK1 expression in Raw246.7 cells decreased after Pg.LPS stimulation and during osteoclast differentiation. DLK1 levels negatively correlated with TNF-α, IL-1β, and NFATC1. Increased DLK1 in Raw246.7 cells further inhibited COX2 and iNOS expressions. Mechanistically, DLK1 overexpression down-regulated NF-κB p65 and JNK levels. In summary, these findings suggest that DLK1 overexpression inhibits periodontal inflammation through the NF-κB p65 and JNK pathways. Interventions targeting increased DLK1 levels may have therapeutic implications for periodontitis.
Collapse
Affiliation(s)
- Yanqing Hong
- Denture Machining Center, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Shandong Provincial Key Medical and Health Discipline of Oral Medicine, Jinan Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan Key Laboratory of Oral Diseases and Tissue Regeneration, Jinan Stomatological Hospital, Jinan, 250001, Shandong Province, China
| | - Linlin Xu
- Central Laboratory, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Shandong Provincial Key Medical and Health Discipline of Oral Medicine, Jinan Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan Key Laboratory of Oral Diseases and Tissue Regeneration, Jinan Stomatological Hospital, Jinan, 250001, Shandong Province, China
| | - Xijiao Yu
- Department of Endodontics, Central Laboratory, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Shandong Provincial Key Medical and Health Discipline of Oral Medicine, Jinan Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan Key Laboratory of Oral Diseases and Tissue Regeneration, Jinan Stomatological Hospital, Jinan, 250001, Shandong Province, China
| | - Yanyan He
- Department of Endodontics, Central Laboratory, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Shandong Provincial Key Medical and Health Discipline of Oral Medicine, Jinan Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan Key Laboratory of Oral Diseases and Tissue Regeneration, Jinan Stomatological Hospital, Jinan, 250001, Shandong Province, China.
| | - Yanmei Du
- Oral Implantology, Central Laboratory, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Shandong Provincial Key Medical and Health Discipline of Oral Medicine, Jinan Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan Key Laboratory of Oral Diseases and Tissue Regeneration, Jinan Stomatological Hospital, Jinan, 250001, Shandong Province, China.
| |
Collapse
|
2
|
Rueda AD, Salvador-Martínez I, Sospedra-Arrufat I, Alcaina-Caro A, Fernández-Miñán A, Burgos-Ruiz AM, Cases I, Mohedano A, Tena JJ, Heyn H, Lopez-Rios J, Nusspaumer G. The cellular landscape of the endochondral bone during the transition to extrauterine life. Immunol Cell Biol 2024; 102:131-148. [PMID: 38184783 DOI: 10.1111/imcb.12718] [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: 11/22/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 01/08/2024]
Abstract
The cellular complexity of the endochondral bone underlies its essential and pleiotropic roles during organismal life. While the adult bone has received significant attention, we still lack a deep understanding of the perinatal bone cellulome. Here, we have profiled the full composition of the murine endochondral bone at the single-cell level during the transition from fetal to newborn life and in comparison with the adult tissue, with particular emphasis on the mesenchymal compartment. The perinatal bone contains different fibroblastic clusters with blastema-like characteristics in organizing and supporting skeletogenesis, angiogenesis and hematopoiesis. Our data also suggest dynamic inter- and intra-compartment interactions, as well as a bone marrow milieu that seems prone to anti-inflammation, which we hypothesize is necessary to ensure the proper program of lymphopoiesis and the establishment of central and peripheral tolerance in early life. Our study provides an integrative roadmap for the future design of genetic and cellular functional assays to validate cellular interactions and lineage relationships within the perinatal bone.
Collapse
Affiliation(s)
- Alejandro Díaz Rueda
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Irepan Salvador-Martínez
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Ismael Sospedra-Arrufat
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Ana Alcaina-Caro
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Ana Fernández-Miñán
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Ana M Burgos-Ruiz
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Ildefonso Cases
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Alberto Mohedano
- Intensive Care Unit, Severo Ochoa University Hospital Leganés, Madrid, Spain
| | - Juan J Tena
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Javier Lopez-Rios
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
- Universidad Loyola Andalucía, School of Health Sciences, Dos Hermanas, Seville, Spain
| | - Gretel Nusspaumer
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| |
Collapse
|
3
|
Peng Y, Yin H, Li S, Yang H. Transcriptome of pituitary function changes in rat model of high altitude cerebral edema. Genomics 2022; 114:110519. [PMID: 36347325 DOI: 10.1016/j.ygeno.2022.110519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/22/2022] [Accepted: 11/04/2022] [Indexed: 11/06/2022]
Abstract
High altitude cerebral edema (HACE) is a serious subtype of acute mountain sickness (AMS). Studies have suggested that increased expression of corticotropin releasing hormone receptor 1 (CRFR1) in pituitary is related to the development of HACE, but no study has revealed the molecular landscape of pituitary function changes in this process. Rat model of HACE was established by simulating the high-altitude hypobaric hypoxia environment. Then RNA-sequencing was performed of rat pituitary gland (PG) in HACE and non-HACE groups. The function annotations, enrichment analysis, protein-protein interaction (PPI) network, chromosome location and drug repositioning of differentially expressed genes (DEGs) were explored based on the transcriptomic data. And we found pituitary secretion function was disordered in HACE, which was partly due to activated inflammation and oxidative stress. In addition, we identified potential biomarkers for early recognition of pituitary dysfunction and potential protective drugs for pituitary function in HACE.
Collapse
Affiliation(s)
- Yuyang Peng
- Multidisciplinary Center for Pituitary Adenomas of Chongqing, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Huachun Yin
- Multidisciplinary Center for Pituitary Adenomas of Chongqing, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Song Li
- Multidisciplinary Center for Pituitary Adenomas of Chongqing, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China; Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Hui Yang
- Multidisciplinary Center for Pituitary Adenomas of Chongqing, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China; Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China.
| |
Collapse
|
4
|
Du X, Zhu M, Zhang T, Wang C, Tao J, Yang S, Zhu Y, Zhao W. The Recombinant Eg.P29-Mediated miR-126a-5p Promotes the Differentiation of Mouse Naive CD4 + T Cells via DLK1-Mediated Notch1 Signal Pathway. Front Immunol 2022; 13:773276. [PMID: 35211114 PMCID: PMC8861942 DOI: 10.3389/fimmu.2022.773276] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
Cystic echinococcosis (CE) is a zoonotic parasitic disease spread worldwide caused by Echinococcus granulosus (Eg), which sometimes causes serious damage; however, in many cases, people are not aware that they are infected. A number of recombinant vaccines based on Eg are used to evaluate their effectiveness against the infection. Our previous report showed that recombinant Eg.P29 (rEg.P29) has a marvelous immunoprotection and can induce Th1 immune response. Furthermore, data of miRNA microarray in mice spleen CD4+ T cells showed that miR-126a-5p was significantly elevated 1 week after immunization by using rEg.P29. Therefore, in this perspective, we discussed the role of miR-126a-5p in the differentiation of naive CD4+ T cells into Th1/Th2 under rEg.P29 immunization and determined the mechanisms associated with delta-like 1 homolog (DLK1) and Notch1 signaling pathway. One week after P29 immunization of mice, we found that miR-126a-5p was significantly increased and DLK1 expression was decreased, while Notch1 pathway activation was enhanced and Th1 response was significantly stronger. The identical conclusion was obtained by overexpression of mmu-miR-126a-5p in primary naive CD4+ T cells in mice. Intriguingly, mmu-miR-126a-5p was significantly raised in serum from mice infected with protoscolex in the early stages of infection and markedly declined in the late stages of infection, while has-miR-126-5p expression was dramatically reduced in serum from CE patients. Taken together, we show that miR-126a-5p functions as a positive regulator of Notch1-mediated differentiation of CD4+ T cells into Th1 through downregulating DLK1 in vivo and in vitro. Hsa-miR-126-5p is potentially a very promising diagnostic biomarker for CE.
Collapse
Affiliation(s)
- Xiancai Du
- School of Basic Medical Science of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Mingxing Zhu
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China.,Center of Scientific Technology of Ningxia Medical University, Yinchuan, China
| | - Tingrui Zhang
- School of Basic Medical Science of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Chan Wang
- School of Basic Medical Science of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Jia Tao
- School of Basic Medical Science of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Songhao Yang
- School of Basic Medical Science of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Yazhou Zhu
- School of Basic Medical Science of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Wei Zhao
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China.,Center of Scientific Technology of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
5
|
Miyauchi K, Ki S, Ukai M, Suzuki Y, Inoue K, Suda W, Matsui T, Ito Y, Honda K, Koseki H, Ohara O, Tanaka RJ, Okada-Hatakeyama M, Kubo M. Essential Role of STAT3 Signaling in Hair Follicle Homeostasis. Front Immunol 2021; 12:663177. [PMID: 34867936 PMCID: PMC8635990 DOI: 10.3389/fimmu.2021.663177] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
Dominant-negative mutations associated with signal transducer and activator of transcription 3 (STAT3) signaling, which controls epithelial proliferation in various tissues, lead to atopic dermatitis in hyper IgE syndrome. This dermatitis is thought to be attributed to defects in STAT3 signaling in type 17 helper T cell specification. However, the role of STAT3 signaling in skin epithelial cells remains unclear. We found that STAT3 signaling in keratinocytes is required to maintain skin homeostasis by negatively controlling the expression of hair follicle-specific keratin genes. These expression patterns correlated with the onset of dermatitis, which was observed in specific pathogen-free conditions but not in germ-free conditions, suggesting the involvement of Toll-like receptor-mediated inflammatory responses. Thus, our study suggests that STAT3-dependent gene expression in keratinocytes plays a critical role in maintaining the homeostasis of skin, which is constantly exposed to microorganisms.
Collapse
Affiliation(s)
- Kosuke Miyauchi
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
| | - Sewon Ki
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
| | - Masao Ukai
- Laboratory for Integrated Cellular Systems, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
- Graduate School of Medical Life Sciences, Yokohama City University, Yokohama, Japan
| | - Yoshie Suzuki
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
| | - Kentaro Inoue
- Laboratory for Integrated Cellular Systems, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
- Department of Computer Science and Systems Engineering, Faculty of Engineering, University of Miyazaki, Miyazaki-shi, Japan
| | - Wataru Suda
- Laboratory for Microbiome science, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Takeshi Matsui
- Laboratory for Evolutionary Cell Biology of the Skin, School of Bioscience and Biotechnology, Tokyo University of Technology, Hachioji, Japan
| | - Yoshihiro Ito
- Laboratory for Gut Homeostasis, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
| | - Kenya Honda
- Laboratory for Gut Homeostasis, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Haruhiko Koseki
- Disease Biology Group, RIKEN Medical Sciences Innovation Hub Program, Kanagawa, Japan
- Laboratory for Developmental Genetics, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
| | - Osamu Ohara
- Laboratory for Integrative Genomics, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Reiko J. Tanaka
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Mariko Okada-Hatakeyama
- Laboratory for Integrated Cellular Systems, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
- Graduate School of Medical Life Sciences, Yokohama City University, Yokohama, Japan
- Institute for Protein Research, Osaka University, Suita-shi, Japan
| | - Masato Kubo
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Japan
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Noda-shi, Japan
- *Correspondence: Masato Kubo,
| |
Collapse
|
6
|
Weickert MT, Hecker JS, Buck MC, Schreck C, Rivière J, Schiemann M, Schallmoser K, Bassermann F, Strunk D, Oostendorp RAJ, Götze KS. Bone marrow stromal cells from MDS and AML patients show increased adipogenic potential with reduced Delta-like-1 expression. Sci Rep 2021; 11:5944. [PMID: 33723276 PMCID: PMC7961144 DOI: 10.1038/s41598-021-85122-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 02/24/2021] [Indexed: 12/29/2022] Open
Abstract
Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are clonal hematopoietic stem cell disorders with a poor prognosis, especially for elderly patients. Increasing evidence suggests that alterations in the non-hematopoietic microenvironment (bone marrow niche) can contribute to or initiate malignant transformation and promote disease progression. One of the key components of the bone marrow (BM) niche are BM stromal cells (BMSC) that give rise to osteoblasts and adipocytes. It has been shown that the balance between these two cell types plays an important role in the regulation of hematopoiesis. However, data on the number of BMSC and the regulation of their differentiation balance in the context of hematopoietic malignancies is scarce. We established a stringent flow cytometric protocol for the prospective isolation of a CD73+ CD105+ CD271+ BMSC subpopulation from uncultivated cryopreserved BM of MDS and AML patients as well as age-matched healthy donors. BMSC from MDS and AML patients showed a strongly reduced frequency of CFU-F (colony forming unit-fibroblast). Moreover, we found an altered phenotype and reduced replating efficiency upon passaging of BMSC from MDS and AML samples. Expression analysis of genes involved in adipo- and osteogenic differentiation as well as Wnt- and Notch-signalling pathways showed significantly reduced levels of DLK1, an early adipogenic cell fate inhibitor in MDS and AML BMSC. Matching this observation, functional analysis showed significantly increased in vitro adipogenic differentiation potential in BMSC from MDS and AML patients. Overall, our data show BMSC with a reduced CFU-F capacity, and an altered molecular and functional profile from MDS and AML patients in culture, indicating an increased adipogenic lineage potential that is likely to provide a disease-promoting microenvironment.
Collapse
Affiliation(s)
- Marie-Theresa Weickert
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Judith S Hecker
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Michèle C Buck
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Christina Schreck
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Jennifer Rivière
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Matthias Schiemann
- Flow Cytometry Unit (CyTUM-MIH), Institute of Microbiology, Immunology, and Hygiene, Technical University of Munich, Munich, Germany
| | - Katharina Schallmoser
- Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria.,Department for Blood Group Serology and Transfusion Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Florian Bassermann
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Partner Site, Munich, Germany
| | - Dirk Strunk
- Experimental and Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - Robert A J Oostendorp
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Katharina S Götze
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany. .,German Cancer Consortium (DKTK), Heidelberg, Partner Site, Munich, Germany.
| |
Collapse
|
7
|
Baulina N, Kiselev I, Favorova O. Imprinted Genes and Multiple Sclerosis: What Do We Know? Int J Mol Sci 2021; 22:1346. [PMID: 33572862 PMCID: PMC7866243 DOI: 10.3390/ijms22031346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune neurodegenerative disease of the central nervous system that arises from interplay between non-genetic and genetic risk factors. The epigenetics functions as a link between these factors, affecting gene expression in response to external influence, and therefore should be extensively studied to improve the knowledge of MS molecular mechanisms. Among others, the epigenetic mechanisms underlie the establishment of parent-of-origin effects that appear as phenotypic differences depending on whether the allele was inherited from the mother or father. The most well described manifestation of parent-of-origin effects is genomic imprinting that causes monoallelic gene expression. It becomes more obvious that disturbances in imprinted genes at the least affecting their expression do occur in MS and may be involved in its pathogenesis. In this review we will focus on the potential role of imprinted genes in MS pathogenesis.
Collapse
Affiliation(s)
- Natalia Baulina
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (I.K.); (O.F.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Ivan Kiselev
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (I.K.); (O.F.)
| | - Olga Favorova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (I.K.); (O.F.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
8
|
López-López S, Monsalve EM, Romero de Ávila MJ, González-Gómez J, Hernández de León N, Ruiz-Marcos F, Baladrón V, Nueda ML, García-León MJ, Screpanti I, Felli MP, Laborda J, García-Ramírez JJ, Díaz-Guerra MJM. NOTCH3 signaling is essential for NF-κB activation in TLR-activated macrophages. Sci Rep 2020; 10:14839. [PMID: 32908186 PMCID: PMC7481794 DOI: 10.1038/s41598-020-71810-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
Macrophage activation by Toll receptors is an essential event in the development of the response against pathogens. NOTCH signaling pathway is involved in the control of macrophage activation and the inflammatory processes. In this work, we have characterized NOTCH signaling in macrophages activated by Toll-like receptor (TLR) triggering and determined that DLL1 and DLL4 are the main ligands responsible for NOTCH signaling. We have identified ADAM10 as the main protease implicated in NOTCH processing and activation. We have also observed that furin, which processes NOTCH receptors, is induced by TLR signaling in a NOTCH-dependent manner. NOTCH3 is the only NOTCH receptor expressed in resting macrophages. Its expression increased rapidly in the first hours after TLR4 activation, followed by a gradual decrease, which was coincident with an elevation of the expression of the other NOTCH receptors. All NOTCH1, 2 and 3 contribute to the increased NOTCH signaling detected in activated macrophages. We also observed a crosstalk between NOTCH3 and NOTCH1 during macrophage activation. Finally, our results highlight the relevance of NOTCH3 in the activation of NF-κB, increasing p65 phosphorylation by p38 MAP kinase. Our data identify, for the first time, NOTCH3 as a relevant player in the control of inflammation.
Collapse
Affiliation(s)
- Susana López-López
- Universidad de Castilla-La Mancha, Medical School/CRIB, Laboratory of Biochemistry and Molecular Biology, Department of Inorganic and Organic Chemistry and Biochemistry, UCLM, C/Almansa 14, 02008, Albacete, Spain.,Unidad Asociada de Biomedicina UCLM, Unidad Asociada CSIC, Albacete, Spain
| | - Eva María Monsalve
- Universidad de Castilla-La Mancha, Medical School/CRIB, Laboratory of Biochemistry and Molecular Biology, Department of Inorganic and Organic Chemistry and Biochemistry, UCLM, C/Almansa 14, 02008, Albacete, Spain.,Unidad Asociada de Biomedicina UCLM, Unidad Asociada CSIC, Albacete, Spain
| | - María José Romero de Ávila
- Universidad de Castilla-La Mancha, Medical School/CRIB, Laboratory of Biochemistry and Molecular Biology, Department of Inorganic and Organic Chemistry and Biochemistry, UCLM, C/Almansa 14, 02008, Albacete, Spain.,Unidad Asociada de Biomedicina UCLM, Unidad Asociada CSIC, Albacete, Spain
| | - Julia González-Gómez
- Universidad de Castilla-La Mancha, CRIB/Biomedicine Unit, Pharmacy School, UCLM/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | | | | | - Victoriano Baladrón
- Universidad de Castilla-La Mancha, Medical School/CRIB, Laboratory of Biochemistry and Molecular Biology, Department of Inorganic and Organic Chemistry and Biochemistry, UCLM, C/Almansa 14, 02008, Albacete, Spain.,Unidad Asociada de Biomedicina UCLM, Unidad Asociada CSIC, Albacete, Spain
| | - María Luisa Nueda
- Universidad de Castilla-La Mancha, CRIB/Biomedicine Unit, Pharmacy School, UCLM/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - María Jesús García-León
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa (CSIC), Universidad Autónoma de Madrid, 28049, Madrid, Spain.,INSERM UMR_S1109, Tumor Biomechanics, 67000, Strasbourg, France.,Université de Strasbourg, 67000, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000, Strasbourg, France
| | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161, Roma, Italy
| | - María Pía Felli
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Jorge Laborda
- Universidad de Castilla-La Mancha, CRIB/Biomedicine Unit, Pharmacy School, UCLM/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - José Javier García-Ramírez
- Universidad de Castilla-La Mancha, Medical School/CRIB, Laboratory of Biochemistry and Molecular Biology, Department of Inorganic and Organic Chemistry and Biochemistry, UCLM, C/Almansa 14, 02008, Albacete, Spain. .,Unidad Asociada de Biomedicina UCLM, Unidad Asociada CSIC, Albacete, Spain.
| | - María José M Díaz-Guerra
- Universidad de Castilla-La Mancha, Medical School/CRIB, Laboratory of Biochemistry and Molecular Biology, Department of Inorganic and Organic Chemistry and Biochemistry, UCLM, C/Almansa 14, 02008, Albacete, Spain. .,Unidad Asociada de Biomedicina UCLM, Unidad Asociada CSIC, Albacete, Spain.
| |
Collapse
|
9
|
Rodríguez-Cano MM, González-Gómez MJ, Sánchez-Solana B, Monsalve EM, Díaz-Guerra MJM, Laborda J, Nueda ML, Baladrón V. NOTCH Receptors and DLK Proteins Enhance Brown Adipogenesis in Mesenchymal C3H10T1/2 Cells. Cells 2020; 9:cells9092032. [PMID: 32899774 PMCID: PMC7565505 DOI: 10.3390/cells9092032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 12/26/2022] Open
Abstract
The NOTCH family of receptors and ligands is involved in numerous cell differentiation processes, including adipogenesis. We recently showed that overexpression of each of the four NOTCH receptors in 3T3-L1 preadipocytes enhances adipogenesis and modulates the acquisition of the mature adipocyte phenotype. We also revealed that DLK proteins modulate the adipogenesis of 3T3-L1 preadipocytes and mesenchymal C3H10T1/2 cells in an opposite way, despite their function as non-canonical inhibitory ligands of NOTCH receptors. In this work, we used multipotent C3H10T1/2 cells as an adipogenic model. We used standard adipogenic procedures and analyzed different parameters by using quantitative-polymerase chain reaction (qPCR), quantitative reverse transcription-polymerase chain reaction (qRT-PCR), luciferase, Western blot, and metabolic assays. We revealed that C3H10T1/2 multipotent cells show higher levels of NOTCH receptors expression and activity and lower Dlk gene expression levels than 3T3-L1 preadipocytes. We found that the overexpression of NOTCH receptors enhanced C3H10T1/2 adipogenesis levels, and the overexpression of NOTCH receptors and DLK (DELTA-like homolog) proteins modulated the conversion of cells towards a brown-like adipocyte phenotype. These and our prior results with 3T3-L1 preadipocytes strengthen the idea that, depending on the cellular context, a precise and highly regulated level of global NOTCH signaling is necessary to allow adipogenesis and determine the mature adipocyte phenotype.
Collapse
Affiliation(s)
- María-Milagros Rodríguez-Cano
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (M.-M.R.-C.); (M.-J.G.-G.)
| | - María-Julia González-Gómez
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (M.-M.R.-C.); (M.-J.G.-G.)
| | - Beatriz Sánchez-Solana
- National Institutes of Health, Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA;
| | - Eva-María Monsalve
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (E.-M.M.); (M.-J.M.D.-G.)
| | - María-José M. Díaz-Guerra
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (E.-M.M.); (M.-J.M.D.-G.)
| | - Jorge Laborda
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (M.-M.R.-C.); (M.-J.G.-G.)
- Correspondence: (J.L.); (M.-L.N.); (V.B.); Tel.: +34-967-599-200 (ext. 2926) (V.B.); Fax: +34-967-599-327 (V.B.)
| | - María-Luisa Nueda
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (M.-M.R.-C.); (M.-J.G.-G.)
- Correspondence: (J.L.); (M.-L.N.); (V.B.); Tel.: +34-967-599-200 (ext. 2926) (V.B.); Fax: +34-967-599-327 (V.B.)
| | - Victoriano Baladrón
- Departamento de Química Inorgánica, Laboratorio de Bioquímica y Biología Molecular, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Orgánica y Bioquímica, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008 Albacete, Spain; (E.-M.M.); (M.-J.M.D.-G.)
- Correspondence: (J.L.); (M.-L.N.); (V.B.); Tel.: +34-967-599-200 (ext. 2926) (V.B.); Fax: +34-967-599-327 (V.B.)
| |
Collapse
|
10
|
Zou L, Wang X, Guo Z, Sun H, Shao C, Yang Y, Sun W. Differential urinary proteomics analysis of myocardial infarction using iTRAQ quantification. Mol Med Rep 2019; 19:3972-3988. [PMID: 30942401 PMCID: PMC6471447 DOI: 10.3892/mmr.2019.10088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 02/06/2019] [Indexed: 11/06/2022] Open
Abstract
Myocardial infarction (MI) is a disease characterized by high morbidity and mortality rates. MI biomarkers are frequently used in clinical diagnosis; however, their specificity and sensitivity remain unsatisfactory. Urinary proteome is an easy, efficient and noninvasive source to examine biomarkers associated with various diseases. The present study, to the best of the authors' knowledge, is the first to examine the urinary proteome using the isobaric tags for relative and absolute quantitation (iTRAQ) technology to identify potential diagnostic biomarkers of MI. The urinary proteome was analyzed within 12 h following the first symptoms of early‑onset MI and at day 7 following percutaneous coronary intervention via iTRAQ labeling and two‑dimensional liquid chromatography‑tandem mass spectrometry. Candidate biomarkers were validated by multiple reaction monitoring (MRM) analysis. A total of 233 urinary proteins were differentially expressed. Gene enrichment analysis identified that the urinary proteome in patients with MI was associated with atherosclerosis, abnormal coagulation and abnormal cell metabolism. In total, 12 differentially expressed urinary proteins were validated by MRM analysis, five of which were associated with MI for the first time in the present study. Binary logistic regression analysis suggested that the combination of five urinary proteins (antithrombin‑III, complement C3, α‑1‑acid glycoprotein 1, serotransferrin and cathepsin Z) may be used to diagnose MI with 94% sensitivity and 93% specificity. In addition, the protein expression levels of three proteins were significantly restored to normal levels following surgical treatment. The verified candidate biomarkers may be used for the diagnosis of MI, and for monitoring the disease status and the effects of treatments for MI. The present results may facilitate future clinical applications of the urinary proteome to diagnose MI.
Collapse
Affiliation(s)
- Lili Zou
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Xubo Wang
- Department of Cardiology, The Fourth Hospital of Jilin University, Changchun, Jilin 130011, P.R. China
| | - Zhengguang Guo
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Haidan Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Chen Shao
- National Key Laboratory of Medical Molecular Biology, Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Yehong Yang
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Wei Sun
- Core Instrument Facility, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| |
Collapse
|
11
|
Davis-Knowlton J, Turner JE, Turner A, Damian-Loring S, Hagler N, Henderson T, Emery IF, Bond K, Duarte CW, Vary CPH, Eldrup-Jorgensen J, Liaw L. Characterization of smooth muscle cells from human atherosclerotic lesions and their responses to Notch signaling. J Transl Med 2019; 99:290-304. [PMID: 29795127 PMCID: PMC6309523 DOI: 10.1038/s41374-018-0072-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis is the most common cause of heart disease and stroke. The use of animal models has advanced our understanding of the molecular signaling that contributes to atherosclerosis. Further understanding of this degenerative process in humans will require human tissue. Plaque removed during endarterectomy procedures to relieve arterial obstructions is usually discarded, but can be an important source of diseased cells. Resected tissue from carotid and femoral endarterectomy procedures were compared with carotid arteries from donors with no known cardiovascular disease. Vascular smooth muscle cells (SMC) contribute to plaque formation and may determine susceptibility to rupture. Notch signaling is implicated in the progression of atherosclerosis, and plays a receptor-specific regulatory role in SMC. We defined protein localization of Notch2 and Notch3 within medial and plaque SMC using immunostaining, and compared Notch2 and Notch3 levels in total plaques with whole normal arteries using immunoblot. We successfully derived SMC populations from multiple endarterectomy specimens for molecular analysis. To better define the protein signature of diseased SMC, we utilized sequential window acquisition of all theoretical spectra (SWATH) proteomic analysis to compare normal carotid artery SMC with endarterectomy-derived SMC. Similarities in protein profile and differentiation markers validated the SMC identity of our explants. We identified a subset of differentially expressed proteins that are candidates as functional markers of diseased SMC. To understand how Notch signaling may affect diseased SMC, we performed Jagged1 stimulation of primary cultures. In populations that displayed significant growth, Jagged1 signaling through Notch2 suppressed proliferation; cultures with low growth potential were non-responsive to Jagged1. In addition, Jagged1 did not promote contractile smooth muscle actin nor have a significant effect on the mature differentiated phenotype. Thus, SMC derived from atherosclerotic lesions show distinct proteomic profiles and have altered Notch signaling in response to Jagged1 as a differentiation stimulus, compared with normal SMC.
Collapse
Affiliation(s)
- Jessica Davis-Knowlton
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA,Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA, 02111, USA
| | - Jacqueline E. Turner
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Anna Turner
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Sydney Damian-Loring
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Nicholas Hagler
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Terry Henderson
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Ivette F. Emery
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Kyle Bond
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Christine W. Duarte
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA,Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA, 02111, USA
| | - Calvin P. H. Vary
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA,Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA, 02111, USA
| | - Jens Eldrup-Jorgensen
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Lucy Liaw
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA. .,Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA, 02111, USA.
| |
Collapse
|
12
|
Nueda ML, González-Gómez MJ, Rodríguez-Cano MM, Monsalve EM, Díaz-Guerra MJM, Sánchez-Solana B, Laborda J, Baladrón V. DLK proteins modulate NOTCH signaling to influence a brown or white 3T3-L1 adipocyte fate. Sci Rep 2018; 8:16923. [PMID: 30446682 PMCID: PMC6240076 DOI: 10.1038/s41598-018-35252-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 11/01/2018] [Indexed: 02/05/2023] Open
Abstract
The role of NOTCH signaling in adipogenesis is highly controversial, with data indicating null, positive or negative effects on this differentiation process. We hypothesize that these contradictory results could be due to the different global NOTCH signaling levels obtained in different experimental settings, because of a specific modulation of NOTCH receptors’ activity by their ligands. We have previously demonstrated that DLK1 and DLK2, two non-canonical NOTCH1 ligands that inhibit NOTCH1 signaling in a dose-dependent manner, modulate the adipogenesis process of 3T3-L1 preadipocytes. In this work, we show that over-expression of any of the four NOTCH receptors enhanced adipogenesis of 3T3-L1 preadipocytes. We also determine that DLK proteins inhibit not only the activity of NOTCH1, but also the activity of NOTCH2, 3 and 4 receptors to different degrees. Interestingly, we have observed, by different approaches, that NOTCH1 over-expression seems to stimulate the differentiation of 3T3-L1 cells towards a brown-like adipocyte phenotype, whereas cells over-expressing NOTCH2, 3 or 4 receptors or DLK proteins would rather differentiate towards a white-like adipocyte phenotype. Finally, our data also demonstrate a complex feed-back mechanism involving Notch and Dlk genes in the regulation of their expression, which suggest that a precise level of global NOTCH expression and NOTCH-dependent transcriptional activity of specific targets could be necessary to determine the final phenotype of 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- María-Luisa Nueda
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica y Bioquímica, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC. C/Almansa 14, 02008, Albacete, Spain
| | - María-Julia González-Gómez
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica y Bioquímica, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC. C/Almansa 14, 02008, Albacete, Spain
| | - María-Milagros Rodríguez-Cano
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica y Bioquímica, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC. C/Almansa 14, 02008, Albacete, Spain
| | - Eva-María Monsalve
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica, Orgánica y Bioquímica, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - María José M Díaz-Guerra
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica, Orgánica y Bioquímica, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain
| | - Beatriz Sánchez-Solana
- Laboratory of Cellular Oncology, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jorge Laborda
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica y Bioquímica, Facultad de Farmacia/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC. C/Almansa 14, 02008, Albacete, Spain
| | - Victoriano Baladrón
- Área de Bioquímica y Biología Molecular, Dpto. Química Inorgánica, Orgánica y Bioquímica, Facultad de Medicina de Albacete/CRIB/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, C/Almansa 14, 02008, Albacete, Spain.
| |
Collapse
|
13
|
Brady RA, Mocca CP, Plaut RD, Takeda K, Burns DL. Comparison of the immune response during acute and chronic Staphylococcus aureus infection. PLoS One 2018; 13:e0195342. [PMID: 29596507 PMCID: PMC5875981 DOI: 10.1371/journal.pone.0195342] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/20/2018] [Indexed: 01/15/2023] Open
Abstract
Staphylococcus aureus bacteria are able to grow in a planktonic state that is associated with acute infections and in biofilms that are associated with chronic infections. Acute infections, such as skin infections, are often self-limiting. However, chronic infections, such as implant infections, can be difficult to clear and may require surgical intervention. The host immune response may contribute to the different outcomes often associated with these two disease types. We used proteomic arrays and two murine models for an initial, descriptive characterization of the contribution of the host immune response to outcomes of acute versus chronic S. aureus disease. We compared the immune responses between a model of self-limiting skin and soft tissue infection caused by the planktonic form of S. aureus versus a model of surgical mesh implant infection, which we show to be caused by a bacterial biofilm. The significantly altered host cytokines and chemokines were largely different in the two models, with responses diminished by 21 days post-implantation in surgical mesh infection. Because bacterial levels remained constant during the 21 days that the surgical mesh infection was followed, those cytokines that are significantly increased during chronic infection are not likely effective in eradicating biofilm. Comparison of the levels of cytokines and chemokines in acute versus chronic S. aureus infection can provide a starting point for evaluation of the role of specific immune factors that are present in one disease manifestation but not the other.
Collapse
Affiliation(s)
- Rebecca A. Brady
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, FDA, Silver Spring, Maryland, United States of America
- * E-mail:
| | - Christopher P. Mocca
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, FDA, Silver Spring, Maryland, United States of America
| | - Roger D. Plaut
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, FDA, Silver Spring, Maryland, United States of America
| | - Kazuyo Takeda
- Microscopy and Imaging Core Facility, Center for Biologics Evaluation and Research, FDA, Silver Spring, Maryland, United States of America
| | - Drusilla L. Burns
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, FDA, Silver Spring, Maryland, United States of America
| |
Collapse
|
14
|
Zhang X, Hu B, Sun J, Li J, Liu S, Song J. Inhibitory Effect of Low-Intensity Pulsed Ultrasound on the Expression of Lipopolysaccharide-Induced Inflammatory Factors in U937 Cells. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2017; 36:2419-2429. [PMID: 28600899 DOI: 10.1002/jum.14239] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/01/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVES Low-intensity pulsed ultrasound (US) has been reported to promote periodontal tissue regeneration and reduce inflammation in soft tissues and in bone infectious diseases. Here we investigated the effect of low-intensity pulsed US on the expression of lipopolysaccharide (LPS)-induced inflammatory factors in U937 macrophage cells. METHODS U937 cells were stimulated with different concentrations of LPS and exposed to different intensities of low-intensity pulsed US. Cell viability and apoptosis of U937 cells were determined by cell-counting kit assays and flow cytometry. A real-time polymerase chain reaction and an enzyme-linked immunosorbent assay were used to test the expression of inflammatory factors. The expression levels of toll-like receptor 4, p65, p-IκBα, and IκBα were assessed by western blots. RESULTS Tumor necrosis factor α began to increase in U937 cells on induction with 1-μg/mL LPS. Low-intensity pulsed US at the intensity of 60 mW/cm2 was more effective in reducing interleukin 8 (IL-8) expression. Furthermore, LPS inhibited the viability and increased apoptosis of U937 cells, whereas low-intensity pulsed US significantly reversed these effects (P < .05). Low-intensity pulsed US reduced the protein expression of IL-6 and IL-8 at both gene and protein levels in U937 cells. The western blot and immunofluorescence showed that low-intensity pulsed US primarily suppressed the degradation and phosphorylation of IκBα and the translocation of p65 into the nuclei. CONCLUSIONS Low-intensity pulsed US alleviated the expression of inflammatory factors induced by LPS in U937 cells. This process was modulated by suppressing the toll-like receptor 4-nuclear factor κB signaling pathway. Therefore, low-intensity pulsed US might be a potential immunomodulatory therapy for the treatment of periodontitis.
Collapse
Affiliation(s)
- Xuan Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Bo Hu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jicheng Sun
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Shan Liu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
15
|
Nguyen NM, de Oliveira Andrade F, Jin L, Zhang X, Macon M, Cruz MI, Benitez C, Wehrenberg B, Yin C, Wang X, Xuan J, de Assis S, Hilakivi-Clarke L. Maternal intake of high n-6 polyunsaturated fatty acid diet during pregnancy causes transgenerational increase in mammary cancer risk in mice. Breast Cancer Res 2017; 19:77. [PMID: 28673325 PMCID: PMC5494892 DOI: 10.1186/s13058-017-0866-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/07/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Maternal and paternal high-fat (HF) diet intake before and/or during pregnancy increases mammary cancer risk in several preclinical models. We studied if maternal consumption of a HF diet that began at a time when the fetal primordial germ cells travel to the genital ridge and start differentiating into germ cells would result in a transgenerational inheritance of increased mammary cancer risk. METHODS Pregnant C57BL/6NTac mouse dams were fed either a control AIN93G or isocaloric HF diet composed of corn oil high in n-6 polyunsaturated fatty acids between gestational days 10 and 20. Offspring in subsequent F1-F3 generations were fed only the control diet. RESULTS Mammary tumor incidence induced by 7,12-dimethylbenz[a]anthracene was significantly higher in F1 (p < 0.016) and F3 generation offspring of HF diet-fed dams (p < 0.040) than in the control offspring. Further, tumor latency was significantly shorter (p < 0.028) and burden higher (p < 0.027) in F1 generation HF offspring, and similar trends were seen in F3 generation HF offspring. RNA sequencing was done on normal mammary glands to identify signaling differences that may predispose to increased breast cancer risk by maternal HF intake. Analysis revealed 1587 and 4423 differentially expressed genes between HF and control offspring in F1 and F3 generations, respectively, of which 48 genes were similarly altered in both generations. Quantitative real-time polymerase chain reaction analysis validated 13 chosen up- and downregulated genes in F3 HF offspring, but only downregulated genes in F1 HF offspring. Ingenuity Pathway Analysis identified upregulation of Notch signaling as a key alteration in HF offspring. Further, knowledge-fused differential dependency network analysis identified ten node genes that in the HF offspring were uniquely connected to genes linked to increased cancer risk (ANKEF1, IGFBP6, SEMA5B), increased resistance to cancer treatments (SLC26A3), poor prognosis (ID4, JAM3, TBX2), and impaired anticancer immunity (EGR3, ZBP1). CONCLUSIONS We conclude that maternal HF diet intake during pregnancy induces a transgenerational increase in offspring mammary cancer risk in mice. The mechanisms of inheritance in the F3 generation may be different from the F1 generation because significantly more changes were seen in the transcriptome.
Collapse
Affiliation(s)
- Nguyen M Nguyen
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Fabia de Oliveira Andrade
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Lu Jin
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Xiyuan Zhang
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Madisa Macon
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - M Idalia Cruz
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Carlos Benitez
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Bryan Wehrenberg
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chao Yin
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Xiao Wang
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington, VA, USA
| | - Jianhua Xuan
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington, VA, USA
| | - Sonia de Assis
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Leena Hilakivi-Clarke
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA.
| |
Collapse
|
16
|
Ruiz-García A, López-López S, García-Ramírez JJ, Baladrón V, Ruiz-Hidalgo MJ, López-Sanz L, Ballesteros Á, Laborda J, Monsalve EM, Díaz-Guerra MJM. The Tetraspanin TSPAN33 Controls TLR-Triggered Macrophage Activation through Modulation of NOTCH Signaling. THE JOURNAL OF IMMUNOLOGY 2016; 197:3371-3381. [PMID: 27574297 DOI: 10.4049/jimmunol.1600421] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 08/08/2016] [Indexed: 12/15/2022]
Abstract
The involvement of NOTCH signaling in macrophage activation by Toll receptors has been clearly established, but the factors and pathways controlling NOTCH signaling during this process have not been completely delineated yet. We have characterized the role of TSPAN33, a tetraspanin implicated in a disintegrin and metalloproteinase (ADAM) 10 maturation, during macrophage proinflammatory activation. Tspan33 expression increases in response to TLR signaling, including responses triggered by TLR4, TLR3, and TLR2 activation, and it is enhanced by IFN-γ. In this study, we report that induction of Tspan33 expression by TLR and IFN-γ is largely dependent on NOTCH signaling, as its expression is clearly diminished in macrophages lacking Notch1 and Notch2 expression, but it is enhanced after overexpression of a constitutively active intracellular domain of NOTCH1. TSPAN33 is the member of the TspanC8 tetraspanin subgroup more intensely induced during macrophage activation, and its overexpression increases ADAM10, but not ADAM17, maturation. TSPAN33 favors NOTCH processing at the membrane by modulating ADAM10 and/or Presenilin1 activity, thus increasing NOTCH signaling in activated macrophages. Moreover, TSPAN33 modulates TLR-induced proinflammatory gene expression, at least in part, by increasing NF-κB-dependent transcriptional activity. Our results suggest that TSPAN33 represents a new control element in the development of inflammation by macrophages that could constitute a potential therapeutic target.
Collapse
Affiliation(s)
- Almudena Ruiz-García
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas, Unidad de Biomedicina, Universidad de Castilla-La Mancha/Consejo Superior de Investigaciones Cientificas, 02006 Albacete, Spain
| | - Susana López-López
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas, Unidad de Biomedicina, Universidad de Castilla-La Mancha/Consejo Superior de Investigaciones Cientificas, 02006 Albacete, Spain
| | - José Javier García-Ramírez
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas, Unidad de Biomedicina, Universidad de Castilla-La Mancha/Consejo Superior de Investigaciones Cientificas, 02006 Albacete, Spain
| | - Victoriano Baladrón
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas, Unidad de Biomedicina, Universidad de Castilla-La Mancha/Consejo Superior de Investigaciones Cientificas, 02006 Albacete, Spain
| | - María José Ruiz-Hidalgo
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas, Unidad de Biomedicina, Universidad de Castilla-La Mancha/Consejo Superior de Investigaciones Cientificas, 02006 Albacete, Spain
| | - Laura López-Sanz
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas, Unidad de Biomedicina, Universidad de Castilla-La Mancha/Consejo Superior de Investigaciones Cientificas, 02006 Albacete, Spain
| | - Ángela Ballesteros
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas, Unidad de Biomedicina, Universidad de Castilla-La Mancha/Consejo Superior de Investigaciones Cientificas, 02006 Albacete, Spain
| | - Jorge Laborda
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas, Unidad de Biomedicina, Universidad de Castilla-La Mancha/Consejo Superior de Investigaciones Cientificas, 02006 Albacete, Spain
| | - Eva María Monsalve
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas, Unidad de Biomedicina, Universidad de Castilla-La Mancha/Consejo Superior de Investigaciones Cientificas, 02006 Albacete, Spain
| | - María José M Díaz-Guerra
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas, Unidad de Biomedicina, Universidad de Castilla-La Mancha/Consejo Superior de Investigaciones Cientificas, 02006 Albacete, Spain
| |
Collapse
|