1
|
Gonzalez-Acera M, Patankar JV, Erkert L, Cineus R, Gamez-Belmonte R, Leupold T, Bubeck M, Bao LL, Dinkel M, Wang R, Schickedanz L, Limberger H, Stolzer I, Gerlach K, Diemand L, Mascia F, Gupta P, Naschberger E, Koop K, Plattner C, Sturm G, Weigmann B, Günther C, Wirtz S, Stürzl M, Hildner K, Kühl AA, Siegmund B, Gießl A, Atreya R, Hegazy AN, Trajanoski Z, Neurath MF, Becker C. Integrated multimodel analysis of intestinal inflammation exposes key molecular features of preclinical and clinical IBD. Gut 2025:gutjnl-2024-333729. [PMID: 40301114 DOI: 10.1136/gutjnl-2024-333729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 04/16/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND IBD is a chronic inflammatory condition driven by complex genetic and immune interactions, yet preclinical models often fail to fully recapitulate all aspects of the human disease. A systematic comparison of commonly used IBD models is essential to identify conserved molecular mechanisms and improve translational relevance. OBJECTIVE We performed a multimodel transcriptomic analysis of 13 widely used IBD mouse models to uncover coregulatory gene networks conserved between preclinical colitis/ileitis and human IBD and to define model-specific and conserved cellular, subcellular and molecular signatures. DESIGN We employed comparative transcriptomic analyses with curated and a priori statistical correlative methods between mouse models versus IBD patient datasets at both bulk and single-cell levels. RESULTS We identify IBD-related pathways, ontologies and cellular compositions that are translatable between mouse models and patient cohorts. We further describe a conserved core inflammatory signature of IBD-associated genes governing T-cell homing, innate immunity and epithelial barrier that translates into the new mouse gut Molecular Inflammation Score (mMIS). Moreover, specific mouse IBD models have distinct signatures for B-cell, T-cell and enteric neurons. We discover that transcriptomic relatedness of models is a function of the mode of induction, not the canonical immunotype (Th1/Th2/Th17). Moreover, the model compendium database is made available as a web explorer (http://trr241.hosting.rrze.uni-erlangen.de/SEPIA/). CONCLUSION This integrated multimodel approach provides a framework for systematically assessing the molecular landscape of intestinal inflammation. Our findings reveal conserved inflammatory circuits, refine model selection, offering a valuable resource for the IBD research community.
Collapse
Affiliation(s)
- Miguel Gonzalez-Acera
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Jay V Patankar
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Lena Erkert
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Roodline Cineus
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Reyes Gamez-Belmonte
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Tamara Leupold
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Marvin Bubeck
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Li-Li Bao
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Martin Dinkel
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Ru Wang
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Laura Schickedanz
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Heidi Limberger
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Iris Stolzer
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Katharina Gerlach
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Leonard Diemand
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Fabrizio Mascia
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Pooja Gupta
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Bayern, Germany
| | - Elisabeth Naschberger
- Department of Surgery, Universitätsklinikum, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Kristina Koop
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Christina Plattner
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Gregor Sturm
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Benno Weigmann
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Claudia Günther
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Michael Stürzl
- Department of Surgery, Universitätsklinikum, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Kai Hildner
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Anja A Kühl
- iPATH.Berlin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Gießl
- Department of Ophthalmology, Universitätsklinikum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Ahmed N Hegazy
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Zlatko Trajanoski
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| |
Collapse
|
2
|
Chandwaskar R, Dalal R, Gupta S, Sharma A, Parashar D, Kashyap VK, Sohal JS, Tripathi SK. Dysregulation of T cell response in the pathogenesis of inflammatory bowel disease. Scand J Immunol 2024; 100:e13412. [PMID: 39394898 DOI: 10.1111/sji.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024]
Abstract
Inflammatory bowel disease (IBD), comprised of Crohn's disease (CD) and ulcerative colitis (UC), are gut inflammatory diseases that were earlier prevalent in the Western Hemisphere but now are on the rise in the East, with India standing second highest in the incidence rate in the world. Inflammation in IBD is a cause of dysregulated immune response, wherein helper T (Th) cell subsets and their cytokines play a major role in the pathogenesis of IBD. In addition, gut microbiota, environmental factors such as dietary factors and host genetics influence the outcome and severity of IBD. Dysregulation between effector and regulatory T cells drives gut inflammation, as effector T cells like Th1, Th17 and Th9 subsets Th cell lineages were found to be increased in IBD patients. In this review, we attempted to discuss the role of different Th cell subsets together with other T cells like CD8+ T cells, NKT and γδT cells in the outcome of gut inflammation in IBD. We also highlighted the potential therapeutic candidates for IBD.
Collapse
Affiliation(s)
- Rucha Chandwaskar
- Amity Institute of Microbial Technology (AIMT), Amity University Jaipur, Rajasthan, India
| | - Rajdeep Dalal
- Infection and Immunology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, India
| | - Saurabh Gupta
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Aishwarya Sharma
- Sri Siddhartha Medical College and Research Center, Tumkur, Karnataka, India
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vivek K Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Jagdip Singh Sohal
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Subhash K Tripathi
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
3
|
Jin L, Macoritto M, Wang J, Bi Y, Wang F, Suarez-Fueyo A, Paez-Cortez J, Hu C, Knight H, Mascanfroni I, Staron MM, Schwartz Sterman A, Houghton JM, Westmoreland S, Tian Y. Multi-Omics Characterization of Colon Mucosa and Submucosa/Wall from Crohn's Disease Patients. Int J Mol Sci 2024; 25:5108. [PMID: 38791146 PMCID: PMC11121447 DOI: 10.3390/ijms25105108] [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: 02/02/2024] [Revised: 03/17/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Crohn's disease (CD) is a subtype of inflammatory bowel disease (IBD) characterized by transmural disease. The concept of transmural healing (TH) has been proposed as an indicator of deep clinical remission of CD and as a predictor of favorable treatment endpoints. Understanding the pathophysiology involved in transmural disease is critical to achieving these endpoints. However, most studies have focused on the intestinal mucosa, overlooking the contribution of the intestinal wall in Crohn's disease. Multi-omics approaches have provided new avenues for exploring the pathogenesis of Crohn's disease and identifying potential biomarkers. We aimed to use transcriptomic and proteomic technologies to compare immune and mesenchymal cell profiles and pathways in the mucosal and submucosa/wall compartments to better understand chronic refractory disease elements to achieve transmural healing. The results revealed similarities and differences in gene and protein expression profiles, metabolic mechanisms, and immune and non-immune pathways between these two compartments. Additionally, the identification of protein isoforms highlights the complex molecular mechanisms underlying this disease, such as decreased RTN4 isoforms (RTN4B2 and RTN4C) in the submucosa/wall, which may be related to the dysregulation of enteric neural processes. These findings have the potential to inform the development of novel therapeutic strategies to achieve TH.
Collapse
Affiliation(s)
- Liang Jin
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (L.J.)
| | | | - Jing Wang
- Immunology Research, AbbVie, Cambridge, MA 02139, USA (A.S.-F.)
| | - Yingtao Bi
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (L.J.)
| | - Fei Wang
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (L.J.)
| | | | | | - Chenqi Hu
- Alnylam Pharmaceuticals, Cambridge, MA 02139, USA
| | - Heather Knight
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (L.J.)
| | | | | | | | - Jean Marie Houghton
- Division of Gastroenterology, Department of Medicine, UMass Chan Medical School, Worcester, MA 01655, USA;
| | | | - Yu Tian
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (L.J.)
| |
Collapse
|
4
|
Uchida AM, Garber JJ, Pyne A, Peterson K, Roelstraete B, Olén O, Halfvarson J, Ludvigsson JF. Eosinophilic esophagitis is associated with increased risk of later inflammatory bowel disease in a nationwide Swedish population cohort. United European Gastroenterol J 2024; 12:34-43. [PMID: 38058270 PMCID: PMC10859712 DOI: 10.1002/ueg2.12493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/20/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Earlier studies on the possible association between eosinophilic esophagitis (EoE) and inflammatory bowel disease (IBD) have been contradictory. METHODS Patients with biopsy-verified EoE diagnosed between 1990 and 2017 in Sweden (n = 1587) were age- and sex-matched with up to five general population reference individuals (n = 7808). EoE was defined using pathology reports from all 28 pathology centers in Sweden (the ESPRESSO study). Multivariate Cox regression then estimated hazard ratios for future IBD. IBD was defined based on the international classification of disease codes and histopathology codes. In secondary analyses, sibling comparators were used to further reduce potential familial confounding. Additionally, we performed logistic regression examining earlier IBD in EoE. RESULTS During follow-up until 2020, 16 (0.01%) EoE patients and 21 (0.003%) general population reference individuals diagnosed with IBD, corresponding to a 3.5-fold increased risk of future IBD (aHR = 3.56; 95% CI 1.79-7.11). EoE was linked to Crohn's disease (aHR = 3.39 [95% CI 1.02-9.60]) but not to ulcerative colitis (aHR = 1.37; 95% CI 0.38-4.86). Compared to their siblings, patients with EoE were at a 2.48-fold increased risk of IBD (aHR = 2.48; 95% CI 0.92-6.70). Earlier IBD was 15 times more likely in EoE patients than in matched reference individuals (odds ratio, 15.39; 95% CI 7.68-33.59). CONCLUSION In this nationwide cohort study, EoE was associated with a 3.5-fold increased risk of later IBD diagnosis. This risk increase may be due to shared genetic or early environmental risk factors, but also surveillance bias could play a role.
Collapse
Affiliation(s)
- Amiko M Uchida
- Division of Gastroenterology, Hepatology & Nutrition, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - John J Garber
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ashley Pyne
- Division of Gastroenterology, Hepatology & Nutrition, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Kathryn Peterson
- Division of Gastroenterology, Hepatology & Nutrition, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Bjorn Roelstraete
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ola Olén
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Sachs' Children and Youth Hospital, Stockholm South General Hospital, Stockholm, Sweden
- Department of Clinical Science and Education Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Halfvarson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatrics, Örebro University, Örebro, Sweden
- Department of Medicine, Celiac Disease Center, Columbia University College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
5
|
Qian J, Lu J, Huang Y, Wang M, Chen B, Bao J, Wang L, Cui D, Luo B, Yan F. Periodontitis Salivary Microbiota Worsens Colitis. J Dent Res 2021; 101:559-568. [PMID: 34796773 DOI: 10.1177/00220345211049781] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Evidence suggests that periodontitis contributes to the pathogenesis of inflammatory bowel disease, including Crohn's disease and ulcerative colitis. However, few studies have examined the role of swallowing and saliva in the pathogenesis of gastrointestinal diseases. Saliva contains an enormous number of oral bacteria and is swallowed directly into the intestine. Here, we explored the influence of periodontitis salivary microbiota on colonic inflammation and possible mechanisms in dextran sulfate sodium (DSS)-induced colitis. The salivary microbiota was collected from healthy individuals and those with periodontitis and gavaged to C57BL/6 mice. Periodontitis colitis was induced by DSS for 5 d and ligature for 1 wk. The degree of colon inflammation was evaluated through hematoxylin and eosin staining, ELISA, and quantitative real-time polymerase chain reaction. Immune parameters were measured with quantitative real-time polymerase chain reaction, flow cytometry, and immunofluorescence. The gut microbiota and metabolome analyses were performed via 16S rRNA gene sequencing and liquid chromatography-mass spectrometry. Although no significant colitis-associated phenotypic changes were found under physiologic conditions, periodontitis salivary microbiota exacerbated colitis in a periodontitis colitis model after DSS induction. The immune response more closely resembled the pathology of ulcerative colitis, including aggravated macrophage M2 polarization and Th2 cell induction (T helper 2). Inflammatory bowel disease-associated microbiota, such as Blautia, Helicobacter, and Ruminococcus, were changed in DSS-induced colitis after periodontitis salivary microbiota gavage. Periodontitis salivary microbiota decreased unsaturated fatty acid levels and increased arachidonic acid metabolism in DSS-induced colitis, which was positively correlated with Aerococcus and Ruminococcus, suggesting the key role of these metabolic events and microbes in the exacerbating effect of periodontitis salivary microbiota on experimental colitis. Our study demonstrated that periodontitis contributes to the pathogenesis of colitis through the swallowing of salivary microbiota, confirming the role of periodontitis in systemic disease and providing new insights into the etiology of gastrointestinal inflammatory diseases.
Collapse
Affiliation(s)
- J Qian
- Affiliated Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - J Lu
- Affiliated Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Y Huang
- Affiliated Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - M Wang
- Affiliated Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - B Chen
- Affiliated Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - J Bao
- Affiliated Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - L Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - D Cui
- Affiliated Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - B Luo
- Affiliated Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - F Yan
- Affiliated Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|