1
|
Keane K, Stephens M, Roizes S, Xue J, Liao S, von der Weid PY. The spatiotemporal development of mesenteric lymphatic changes in the TNFΔARE/+ mouse model of terminal ileitis. Am J Physiol Gastrointest Liver Physiol 2025; 328:G624-G643. [PMID: 40062472 DOI: 10.1152/ajpgi.00334.2024] [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: 10/16/2024] [Revised: 11/13/2024] [Accepted: 02/13/2025] [Indexed: 05/06/2025]
Abstract
Crohn's disease (CD) is a chronic inflammatory bowel disease, which also encompasses significant alterations of the mesenteric lymphatic system. Whether these changes are a mere consequence of or directly contribute to the inflammation is unknown. Here, we characterized the spatial and temporal development of these events in the TNFΔARE/+ mouse, which develops CD-like ileitis and significant mesenteric lymphatic alterations. At 8, 12, 20, and 28 wk of age, specific pathogen-free (SPF), germ-free (GF) TNFΔARE/+ and wild-type (WT) mice were assessed for ileitis via myeloperoxidase (MPO) activity while mesenteric lymphatic alterations were assessed by confocal immunofluorescence imaging. Lymphatic alterations in the SPF TNFΔARE/+ occurred in a stepwise manner between 8 and 28 wk of age beginning with the development of mesenteric lymphadenopathy at 8 wk despite no significant ileitis. By 12-wk ileal MPO significantly elevates concomitantly with lymphangiectasia of the mesenteric collecting lymphatic vessels (CLVs) and clustering of CD45+ immune cells around them. At 20 wk, significant lymphangiogenesis of the initials (initial lymphatic vessel) and tertiary lymphoid organs aligned along lymphatic collectors (CA-TLOs) had developed. At 28 wk, lymphangiectasia, lymphangiogenesis, and CA-TLOs increased. However, 28-wk-old GF TNFΔARE/+, while displaying no ileitis, presented with mesenteric lymphadenopathy, lymphangiectasia, and lymphangiogenesis but no immune cell clustering nor CA-TLOs. The TNFΔARE/+ mice develop terminal ileitis and lymphatic alterations in a stepwise manner beginning with mesenteric lymph node lymphadenopathy and ileal inflammation, followed by CLV dilation and lymphangiogenesis. These lymphatic alterations are exacerbated by the gut microbiome, with immune cell clustering and tertiary lymphoid organ formation being entirely dependent of its presence.NEW & NOTEWORTHY The mesenteric lymphatic system displays striking morphological alterations in Crohn's disease. To assess the importance of these changes in the perpetuation of the disease, we established the timeframe of their occurrence with respect to the development of ileitis in a mouse model of Crohn's disease and in the same model derived germ-free where intestinal inflammation does not occur. Although immune-related alterations seem to depend on microbiome, changes specifically affecting lymphatic vessels persist in its absence.
Collapse
Affiliation(s)
- Keith Keane
- Department of Physiology and Pharmacology, Inflammation Research Network, The Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Matthew Stephens
- Department of Physiology and Pharmacology, Inflammation Research Network, The Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Simon Roizes
- Department of Physiology and Pharmacology, Inflammation Research Network, The Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jingna Xue
- Department of Microbiology, Immunology and Infectious Diseases, Inflammation Research Network, The Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Shan Liao
- Department of Microbiology, Immunology and Infectious Diseases, Inflammation Research Network, The Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Pierre-Yves von der Weid
- Department of Physiology and Pharmacology, Inflammation Research Network, The Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
2
|
Li X, Liu X, Wei H, Liu Y, Xu G. Neutrophils in tumor- and inflammation-induced lymphangiogenesis. Int J Biol Sci 2025; 21:2223-2234. [PMID: 40083688 PMCID: PMC11900808 DOI: 10.7150/ijbs.103458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
Lymphangiogenesis is the formation of new lymphatic vessels from preexisting vessels and occurs during embryonic lymphatic development and under pathological conditions induced by internal or external stimuli. Emerging evidence suggests that neutrophils contribute to the construction and remodeling of new lymphatic vessels. Neutrophils migrate to lymph nodes through the lymphatic vessels or high endothelial venules, and neutrophil migration may depend on the phenotype of the neutrophil. The presence of unique neutrophil phenotypes in individuals with lymphangiogenesis has been reported. Neutrophils promote lymphangiogenesis mainly by secreting lymphotropic factors or increasing their bioavailability and by collaborating with various immune cells. Neutrophils mediate lymphangiogenesis and exert complex effects on tumors and inflammation. The selective inhibition of specific neutrophil and neutrophil lymphangiogenic molecules may provide a novel approach for the prevention and treatment of associated diseases.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, P. R. China
| | - Xiaoxin Liu
- Department of Nephrology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430077 Wuhan, Hubei, P. R. China
| | - Haotian Wei
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, P. R. China
| | - Yanyan Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, P. R. China
| | - Gang Xu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, P. R. China
| |
Collapse
|
3
|
Zhou YW, Ren Y, Lu MM, Xu LL, Cheng WX, Zhang MM, Ding LP, Chen D, Gao JG, Du J, Jin CL, Chen CX, Li YF, Cheng T, Jiang PL, Yang YD, Qian PX, Xu PF, Jin X. Crohn's disease as the intestinal manifestation of pan-lymphatic dysfunction: An exploratory proposal based on basic and clinical data. World J Gastroenterol 2024; 30:34-49. [PMID: 38293325 PMCID: PMC10823898 DOI: 10.3748/wjg.v30.i1.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/08/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024] Open
Abstract
Crohn's disease (CD) is caused by immune, environmental, and genetic factors. It can involve the entire gastrointestinal tract, and although its prevalence is rapidly increasing its etiology remains unclear. Emerging biological and small-molecule drugs have advanced the treatment of CD; however, a considerable proportion of patients are non-responsive to all known drugs. To achieve a breakthrough in this field, innovations that could guide the further development of effective therapies are of utmost urgency. In this review, we first propose the innovative concept of pan-lymphatic dysfunction for the general distribution of lymphatic dysfunction in various diseases, and suggest that CD is the intestinal manifestation of pan-lymphatic dysfunction based on basic and clinical preliminary data. The supporting evidence is fully summarized, including the existence of lymphatic system dysfunction, recognition of the inside-out model, disorders of immune cells, changes in cell plasticity, partial overlap of the underlying mechanisms, and common gut-derived fatty and bile acid metabolism. Another benefit of this novel concept is that it proposes adopting the zebrafish model for studying intestinal diseases, especially CD, as this model is good at presenting and mimicking lymphatic dysfunction. More importantly, the ensuing focus on improving lymphatic function may lead to novel and promising therapeutic strategies for CD.
Collapse
Affiliation(s)
- Yu-Wei Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yue Ren
- Department of Gastroenterology, The Second Hospital of Jiaxing, Jiaxing 314000, Zhejiang Province, China
| | - Miao-Miao Lu
- Endoscopy Center, Children’s Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Ling-Ling Xu
- Department of Gastroenterology, The Second People’s Hospital of Yuhang District, Hangzhou 310000, Zhejiang Province, China
| | - Wei-Xin Cheng
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Meng-Meng Zhang
- Department of Gastroenterology, Hangzhou Shangcheng District People’s Hospital, Hangzhou 310003, Zhejiang Province, China
| | - Lin-Ping Ding
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Dong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Jian-Guo Gao
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Juan Du
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Ci-Liang Jin
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Chun-Xiao Chen
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yun-Fei Li
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Tao Cheng
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Peng-Lei Jiang
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yi-Da Yang
- Department of Infectious Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Peng-Xu Qian
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Peng-Fei Xu
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Xi Jin
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
4
|
Nikolakis D, de Voogd FAE, Pruijt MJ, Grootjans J, van de Sande MG, D’Haens GR. The Role of the Lymphatic System in the Pathogenesis and Treatment of Inflammatory Bowel Disease. Int J Mol Sci 2022; 23:ijms23031854. [PMID: 35163775 PMCID: PMC8836364 DOI: 10.3390/ijms23031854] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 02/04/2023] Open
Abstract
Although the number of therapeutic options for the treatment of inflammatory bowel disease (IBD) has increased in recent years, patients suffer from decreased quality of life due to non-response or loss of response to the currently available treatments. An increased understanding of the disease’s etiology could provide novel insights for treatment strategies in IBD. Lymphatic system components are generally linked to immune responses and presumably related to inflammatory diseases pathophysiology. This review aims to summarize findings on immune-mediated mechanisms in lymphoid tissues linked with IBD pathogenesis and (potential) novel treatments. Enhanced innate and adaptive immune responses were observed in mesenteric lymph nodes (MLNs) and other lymphoid structures, such as Peyer’s patches, in patients with IBD and in animal models. Furthermore, the phenomenon of lymphatic obstruction in the form of granulomas in MLNs and lymphatic vessels correlates with disease activity. There is also evidence that abnormalities in the lymphatic stromal components and lymph node microbiome are common in IBD and could be exploited therapeutically. Finally, novel agents targeting lymphocyte trafficking have been added to the treatment armamentarium in the field of IBD. Overall, gut-associated lymphoid tissue plays a key role in IBD immunopathogenesis, which could offer novel therapeutic targets.
Collapse
Affiliation(s)
- Dimitrios Nikolakis
- Department of Gastroenterology, Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Academic Medical Center, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (D.N.); (F.A.E.d.V.); (M.J.P.); (J.G.)
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Onassis Foundation, 4 Aeschinou Street, 10558 Athens, Greece
| | - Floris A. E. de Voogd
- Department of Gastroenterology, Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Academic Medical Center, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (D.N.); (F.A.E.d.V.); (M.J.P.); (J.G.)
| | - Maarten J. Pruijt
- Department of Gastroenterology, Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Academic Medical Center, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (D.N.); (F.A.E.d.V.); (M.J.P.); (J.G.)
| | - Joep Grootjans
- Department of Gastroenterology, Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Academic Medical Center, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (D.N.); (F.A.E.d.V.); (M.J.P.); (J.G.)
| | - Marleen G. van de Sande
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Geert R. D’Haens
- Department of Gastroenterology, Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Academic Medical Center, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (D.N.); (F.A.E.d.V.); (M.J.P.); (J.G.)
- Correspondence:
| |
Collapse
|
5
|
Ocansey DKW, Pei B, Xu X, Zhang L, Olovo CV, Mao F. Cellular and molecular mediators of lymphangiogenesis in inflammatory bowel disease. J Transl Med 2021; 19:254. [PMID: 34112196 PMCID: PMC8190852 DOI: 10.1186/s12967-021-02922-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Background Recent studies reporting the intricate crosstalk between cellular and molecular mediators and the lymphatic endothelium in the development of inflammatory bowel diseases (IBD) suggest altered inflammatory cell drainage and lymphatic vasculature, implicating the lymphatic system as a player in the occurrence, development, and recurrence of intestinal diseases. This article aims to review recent data on the modulatory functions of cellular and molecular components of the IBD microenvironment on the lymphatic system, particularly lymphangiogenesis. It serves as a promising therapeutic target for IBD management and treatment. The interaction with gut microbiota is also explored. Main text Evidence shows that cells of the innate and adaptive immune system and certain non-immune cells participate in the complex processes of inflammatory-induced lymphangiogenesis through the secretion of a wide spectrum of molecular factors, which vary greatly among the various cells. Lymphangiogenesis enhances lymphatic fluid drainage, hence reduced infiltration of immunomodulatory cells and associated-inflammatory cytokines. Interestingly, some of the cellular mediators, including mast cells, neutrophils, basophils, monocytes, and lymphatic endothelial cells (LECs), are a source of lymphangiogenic molecules, and a target as they express specific receptors for lymphangiogenic factors. Conclusion The effective target of lymphangiogenesis is expected to provide novel therapeutic interventions for intestinal inflammatory conditions, including IBD, through both immune and non-immune cells and based on cellular and molecular mechanisms of lymphangiogenesis that facilitate inflammation resolution.
Collapse
Affiliation(s)
- Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Bing Pei
- Department of Clinical Laboratory, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, Jiangsu, People's Republic of China
| | - Xinwei Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Lu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Chinasa Valerie Olovo
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Department of Microbiology, University of Nigeria, Nsukka, 410001, Nigeria
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
6
|
Zhang L, Ocansey DKW, Liu L, Olovo CV, Zhang X, Qian H, Xu W, Mao F. Implications of lymphatic alterations in the pathogenesis and treatment of inflammatory bowel disease. Biomed Pharmacother 2021; 140:111752. [PMID: 34044275 DOI: 10.1016/j.biopha.2021.111752] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/06/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by intense immune dysregulation, gut microbiota imbalance, and intestinal epithelium destruction. Among the factors that contribute to the pathogenesis of IBD, lymphatics have received less attention, hence less studied, characterized, and explored. However, in recent years, the role of the lymphatic system in gastrointestinal pathophysiology continues to be highlighted. This paper examines the implications of lymphatic changes in IBD pathogenesis related to immune cells, gut microbiota, intestinal and mesenteric epithelial barrier integrity, and progression to colorectal cancer (CRC). Therapeutic targets of lymphatics in IBD studies are also presented. Available studies indicate that lymph nodes and other secondary lymphatic tissues, provide highly specialized microenvironments for mounting effective immune responses and that lymphatic integrity plays a significant role in small intestine homeostasis, where the lymphatic vasculature effectively controls tissue edema, leukocyte exit, bacterial antigen, and inflammatory chemokine clearance. In IBD, there are functional and morphological alterations in intestinal and mesenteric lymphatic vessels (more profoundly in Crohn's disease [CD] compared to ulcerative colitis [UC]), including lymphangiogenesis, lymphangiectasia, lymphadenopathy, and lymphatic vasculature blockade, affecting not only immunity but gut microbiota and epithelial barrier integrity. While increased lymphangiogenesis is primarily associated with a good prognosis of IBD, increased lymphangiectasia, lymphadenopathy, and lymphatic vessel occlusion correlate with poor prognosis. IBD therapies that target the lymphatic system seek to increase lymphangiogenesis via induction of lymphangiogenic factors and inhibition of its antagonists. The resultant increased lymphatic flow coupled with other anti-inflammatory activities restores gut homeostasis.
Collapse
Affiliation(s)
- Lu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China; Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Lianqin Liu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Chinasa Valerie Olovo
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China; Department of Microbiology, University of Nigeria, Nsukka 410001, Nigeria
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Hui Qian
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Wenrong Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China.
| |
Collapse
|
7
|
Oliver G, Kipnis J, Randolph GJ, Harvey NL. The Lymphatic Vasculature in the 21 st Century: Novel Functional Roles in Homeostasis and Disease. Cell 2020; 182:270-296. [PMID: 32707093 PMCID: PMC7392116 DOI: 10.1016/j.cell.2020.06.039] [Citation(s) in RCA: 420] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/17/2020] [Accepted: 06/25/2020] [Indexed: 12/19/2022]
Abstract
Mammals have two specialized vascular circulatory systems: the blood vasculature and the lymphatic vasculature. The lymphatic vasculature is a unidirectional conduit that returns filtered interstitial arterial fluid and tissue metabolites to the blood circulation. It also plays major roles in immune cell trafficking and lipid absorption. As we discuss in this review, the molecular characterization of lymphatic vascular development and our understanding of this vasculature's role in pathophysiological conditions has greatly improved in recent years, changing conventional views about the roles of the lymphatic vasculature in health and disease. Morphological or functional defects in the lymphatic vasculature have now been uncovered in several pathological conditions. We propose that subtle asymptomatic alterations in lymphatic vascular function could underlie the variability seen in the body's response to a wide range of human diseases.
Collapse
Affiliation(s)
- Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Natasha L Harvey
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| |
Collapse
|
8
|
Sulforaphane Elicits Protective Effects in Intestinal Ischemia Reperfusion Injury. Int J Mol Sci 2020; 21:ijms21155189. [PMID: 32707886 PMCID: PMC7432940 DOI: 10.3390/ijms21155189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
Intestinal ischemia reperfusion injury (IRI) is an inherent, unavoidable event of intestinal transplantation, contributing to allograft failure and rejection. The inflammatory state elicited by intestinal IRI is characterized by heightened leukocyte recruitment to the gut, which is amplified by a cross-talk with platelets at the endothelial border. Sulforaphane (SFN), a naturally occurring isothiocyanate, exhibits anti-inflammatory characteristics and has been shown to reduce platelet activation and block leukocyte adhesion. Thus, the aim of this study was to investigate protective effects and mechanism of action of SFN in a murine model of intestinal IRI. Intestinal IRI was induced by superior mesenteric artery occlusion for 30 min, followed by reperfusion for 2 h, 8 h or 24 h. To investigate cellular interactions, leukocytes were in vivo stained with rhodamine and platelets were harvested from donor animals and ex vivo stained. Mice (C57BL/6J) were divided into three groups: (1) control, (2) SFN treatment 24 h prior to reperfusion and (3) SFN treatment 24 h prior to platelet donation. Leukocyte and platelet recruitment was analyzed via intravital microscopy. Tissue was analyzed for morphological alterations in intestinal mucosa, barrier permeability, and leukocyte infiltration. Leukocyte rolling and adhesion was significantly reduced 2 h and 8 h after reperfusion. Mice receiving SFN treated platelets exhibited significantly decreased leukocyte and platelet recruitment. SFN showed protection for intestinal tissue with less damage observed in histopathological and ultrastructural evaluation. In summary, the data presented provide evidence for SFN as a potential therapeutic strategy against intestinal IRI.
Collapse
|
9
|
Becker F, Romero E, Goetzmann J, Hasselschwert DL, Dray B, Vanchiere J, Fontenot J, Yun JW, Norris PC, White L, Musso M, Serhan CN, Alexander JS, Gavins FNE. Endogenous Specialized Proresolving Mediator Profiles in a Novel Experimental Model of Lymphatic Obstruction and Intestinal Inflammation in African Green Monkeys. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 189:1953-1972. [PMID: 31547920 DOI: 10.1016/j.ajpath.2019.05.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 04/18/2019] [Accepted: 05/09/2019] [Indexed: 12/16/2022]
Abstract
Changes in the intestinal lymphatic vascular system, such as lymphatic obstruction, are characteristic features of inflammatory bowel diseases. The lymphatic vasculature forms a conduit to enable resolution of inflammation; this process is driven by specialized endogenous proresolving mediators (SPMs). To evaluate contributions of lymphatic obstruction to intestinal inflammation and to study profiles of SPMs, we generated a novel animal model of lymphatic obstruction using African green monkeys. Follow-up studies were performed at 7, 21, and 61 days. Inflammation was determined by histology. Luminex assays were performed to evaluate chemokine and cytokine levels. In addition, lipid mediator metabololipidomic profiling was performed to identify SPMs. After 7 days, lymphatic obstruction resulted in a localized inflammatory state, paralleled by an increase in inflammatory chemokines and cytokines, which were found to be up-regulated after 7 days but returned to baseline after 21 and 61 days. At the same time, a distinct pattern of SPMs was profiled, with an increase for D-series resolvins, protectins, maresins, and lipoxins at 61 days. These results indicate that intestinal lymphatic obstruction can lead to an acute inflammatory state, accompanied by an increase in proinflammatory mediators, followed by a phase of resolution, paralleled by an increase and decrease of respective SPMs.
Collapse
Affiliation(s)
- Felix Becker
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of General, Visceral and Transplant Surgery, University of Münster, Münster, Germany
| | - Emily Romero
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana
| | - Jason Goetzmann
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana
| | - Dana L Hasselschwert
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana
| | - Beth Dray
- Department of Veterinary Science and Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer Center, Bastrop, Texas
| | - John Vanchiere
- Section of Pediatric Infectious Diseases, Department of Pediatrics, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Jane Fontenot
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana
| | - J Winny Yun
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Paul C Norris
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Luke White
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Melany Musso
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - J Steven Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Felicity N E Gavins
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of Life Sciences, Brunel University London, London, United Kingdom.
| |
Collapse
|
10
|
Nunes NS, Kim S, Sundby M, Chandran P, Burks SR, Paz AH, Frank JA. Temporal clinical, proteomic, histological and cellular immune responses of dextran sulfate sodium-induced acute colitis. World J Gastroenterol 2018; 24:4341-4355. [PMID: 30344419 PMCID: PMC6189848 DOI: 10.3748/wjg.v24.i38.4341] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/06/2018] [Accepted: 08/24/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the temporal clinical, proteomic, histological and cellular immune profiles of dextran sulfate sodium (DSS)-induced acute colitis.
METHODS Acute colitis was induced in C57Bl/6 female mice by administration of 1%, 2% or 3% DSS in drinking water for 7 d. Animals were monitored daily for weight loss, stool consistency and blood in the stool, while spleens and colons were harvested on day 8. A time course analysis was performed in mice ingesting 3% DSS, which included colon proteomics through multiplex assay, colon histological scoring by a blinded investigator, and immune response through flow cytometry or immunohistochemistry of the spleen, mesenteric lymph node and colon.
RESULTS Progressive worsening of clinical colitis was observed with increasing DSS from 1% to 3%. In mice ingesting 3% DSS, colon shortening and increase in pro-inflammatory factors starting at day 3 was observed, with increased spleen weights at day 6 and day 8. This coincided with cellular infiltration in the colon from day 2 to day 8, with progressive accumulation of macrophages F4/80+, T helper CD4+ (Th), T cytotoxic CD8+ (Tcyt) and T regulatory CD25+ (Treg) cells, and progressive changes in colonic pathology including destruction of crypts, loss of goblet cells and depletion of the epithelial barrier. Starting on day 4, mesenteric lymph node and/or spleen presented with lower levels of Treg, Th and Tcyt cells, suggesting an immune cell tropism to the gut.
CONCLUSION These results demonstrate that the severity of experimental colitis is dependent on DSS concentration, correlated with clinical, proteomic, histological and cellular immune response on 3% DSS.
Collapse
Affiliation(s)
- Natalia Schneider Nunes
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States
- Gastroenterology and Hepatology Sciences Graduate Program, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-093, Brazil
| | - Saejeong Kim
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States
| | - Maggie Sundby
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States
| | - Parwathy Chandran
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States
| | - Scott Robert Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States
| | - Ana Helena Paz
- Gastroenterology and Hepatology Sciences Graduate Program, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-093, Brazil
| | - Joseph Alan Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, United States
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, United States
| |
Collapse
|
11
|
Li X, Jusko WJ, Cao Y. Role of Interstitial Fluid Turnover on Target Suppression by Therapeutic Biologics Using a Minimal Physiologically Based Pharmacokinetic Model. J Pharmacol Exp Ther 2018; 367:1-8. [PMID: 30002096 PMCID: PMC6123664 DOI: 10.1124/jpet.118.250134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/09/2018] [Indexed: 02/06/2023] Open
Abstract
For therapeutic biologics against soluble ligands, the magnitude and duration of target suppression affect their therapeutic efficacy. Many factors have been evaluated in relation to target suppression but the interstitial fluid turnover rate in target tissues has not been considered. Inspired by the fact that etanercept exerts limited efficacy in Crohn's disease despite its high efficacy in rheumatoid arthritis, we developed a minimal physiologically based pharmacokinetic model to investigate the role of the tissue fluid turnover rate on soluble target suppression and assessed the interrelationships between binding constants and tissue fluid turnover. Interstitial fluid turnover rates in target tissues were found to strongly influence target binding kinetics. For tissues with low fluid turnover, stable binders (low koff) exhibit greater target suppression, but efficacy is often restricted by accumulation of the drug-target complex. For tissues with high fluid turnover, fast binders (high kon) are generally favored, but a plateau effect is present for antibodies with low dissociation rates (koff). Etanercept is often regarded as a fast tumor necrosis factor-α (TNF-α) binder (high kon) despite comparable binding affinity (KD, koff/kon) with adalimumab and infliximab. Crohn's disease largely involves the colon, a tissue with relatively slower fluid turnover than arthritis-associated joint synovium; this may explain why etanercept exerts poor TNF-α suppressive effect in Crohn's disease. This study highlights the importance of tissue interstitial fluid turnover in evaluation of therapeutic antibodies bound to soluble antigens.
Collapse
Affiliation(s)
- Xiaobing Li
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China (X.L.); Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (X.L., Y.C.); and Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical, Sciences, State University of New York at Buffalo, Buffalo, New York (W.J.J.)
| | - William J Jusko
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China (X.L.); Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (X.L., Y.C.); and Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical, Sciences, State University of New York at Buffalo, Buffalo, New York (W.J.J.)
| | - Yanguang Cao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China (X.L.); Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (X.L., Y.C.); and Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical, Sciences, State University of New York at Buffalo, Buffalo, New York (W.J.J.)
| |
Collapse
|
12
|
Seamons A, Treuting PM, Meeker S, Hsu C, Paik J, Brabb T, Escobar SS, Alexander JS, Ericsson AC, Smith JG, Maggio-Price L. Obstructive Lymphangitis Precedes Colitis in Murine Norovirus-Infected Stat1-Deficient Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1536-1554. [PMID: 29753791 PMCID: PMC6109697 DOI: 10.1016/j.ajpath.2018.03.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 03/07/2018] [Accepted: 03/26/2018] [Indexed: 12/16/2022]
Abstract
Murine norovirus (MNV) is an RNA virus that can prove lethal in mice with impaired innate immunity. We found that MNV-4 infection of Stat1-/- mice was not lethal, but produced a 100% penetrant, previously undescribed lymphatic phenotype characterized by chronic-active lymphangitis with hepatitis, splenitis, and chronic cecal and colonic inflammation. Lesion pathogenesis progressed from early ileal enteritis and regional dilated lymphatics to lymphangitis, granulomatous changes in the liver and spleen, and, ultimately, typhlocolitis. Lesion development was neither affected by antibiotics nor reproduced by infection with another enteric RNA virus, rotavirus. MNV-4 infection in Stat1-/- mice decreased expression of vascular endothelial growth factor (Vegf) receptor 3, Vegf-c, and Vegf-d and increased interferon (Ifn)-γ, tumor necrosis factor-α, and inducible nitric oxide synthase. However, anti-IFN-γ and anti-tumor necrosis factor-α antibody treatment did not attenuate the histologic lesions. Studies in Ifnαβγr-/- mice suggested that canonical signaling via interferon receptors did not cause MNV-4-induced disease. Infected Stat1-/- mice had increased STAT3 phosphorylation and expressed many STAT3-regulated genes, consistent with our findings of increased myeloid cell subsets and serum granulocyte colony-stimulating factor, which are also associated with increased STAT3 activity. In conclusion, in Stat1-/- mice, MNV-4 induces lymphatic lesions similar to those seen in Crohn disease as well as hepatitis, splenitis, and typhlocolitis. MNV-4-infected Stat1-/- mice may be a useful model to study mechanistic associations between viral infections, lymphatic dysfunction, and intestinal inflammation in a genetically susceptible host.
Collapse
Affiliation(s)
- Audrey Seamons
- Department of Comparative Medicine, University of Washington, Seattle, Washington.
| | - Piper M Treuting
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Stacey Meeker
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Charlie Hsu
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Jisun Paik
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Thea Brabb
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Sabine S Escobar
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Jonathan S Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University, Shreveport, Louisiana
| | - Aaron C Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Jason G Smith
- Department of Microbiology, University of Washington, Seattle, Washington
| | - Lillian Maggio-Price
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
13
|
Merigo F, Brandolese A, Facchin S, Missaggia S, Bernardi P, Boschi F, D’Incà R, Savarino EV, Sbarbati A, Sturniolo GC. Glucose transporter expression in the human colon. World J Gastroenterol 2018; 24:775-793. [PMID: 29467549 PMCID: PMC5807937 DOI: 10.3748/wjg.v24.i7.775] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 12/13/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate by immunostaining glucose transporter expression in human colorectal mucosa in controls and patients with inflammatory bowel disease (IBD).
METHODS Colorectal samples were obtained from patients undergoing lower endoscopic colonoscopy or recto-sigmoidoscopy. Patients diagnosed with ulcerative colitis (n = 18) or Crohn’s disease (n = 10) and scheduled for diagnostic colonoscopy were enrolled. Patients who underwent colonoscopy for prevention screening of colorectal cancer or were followed-up after polypectomy or had a history of lower gastrointestinal symptoms were designated as the control group (CTRL, n = 16). Inflammatory status of the mucosa at the sampling site was evaluated histologically and/or endoscopically. A total of 147 biopsies of colorectal mucosa were collected and processed for immunohistochemistry analysis. The expression of GLUT2, SGLT1, and GLUT5 glucose transporters was investigated using immunoperoxidase labeling. To compare immunoreactivity of GLUT5 and LYVE-1, which is a marker for lymphatic vessel endothelium, double-labeled confocal microscopy was used.
RESULTS Immunohistochemical analysis revealed that GLUT2, SGLT1, and GLUT5 were expressed only in short epithelial portions of the large intestinal mucosa. No important differences were observed in glucose transporter expression between the samples obtained from the different portions of the colorectal tract and between the different patient groups. Unexpectedly, GLUT5 expression was also identified in vessels, mainly concentrated in specific areas where the vessels were clustered. Immunostaining with LYVE-1 and GLUT5 antibodies revealed that GLUT5-immunoreactive (-IR) clusters of vessels were concentrated in areas internal to those that were LYVE-1 positive. GLUT5 and LYVE-1 did not appear to be colocalized but rather showed a close topographical relationship on the endothelium. Based on their LYVE-1 expression, GLUT5-IR vessels were identified as lymphatic. Both inflamed and non-inflamed mucosal colorectal tissue biopsies from the IBD and CTRL patients showed GLUT5-IR clusters of lymphatic vessels.
CONCLUSION Glucose transporter immunoreactivity is present in colorectal mucosa in controls and IBD patients. GLUT5 expression is also associated with lymphatic vessels. This novel finding aids in the characterization of lymphatic vasculature in IBD patients.
Collapse
Affiliation(s)
- Flavia Merigo
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, Verona I-37134, Italy
| | - Alessandro Brandolese
- Department of Medicine, Gastroenterology Section, University of Verona, Verona I-37134, Italy
| | - Sonia Facchin
- Department of Surgery, Oncology and Gastroenterology, Gastroenterology Section, University Hospital of Padua, Padua I-35128, Italy
| | - Silvia Missaggia
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, Verona I-37134, Italy
| | - Paolo Bernardi
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, Verona I-37134, Italy
| | - Federico Boschi
- Department of Computer Science, University of Verona, Verona I-37134, Italy
| | - Renata D’Incà
- Department of Surgery, Oncology and Gastroenterology, Gastroenterology Section, University Hospital of Padua, Padua I-35128, Italy
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology and Gastroenterology, Gastroenterology Section, University Hospital of Padua, Padua I-35128, Italy
| | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, Verona I-37134, Italy
| | - Giacomo Carlo Sturniolo
- Department of Surgery, Oncology and Gastroenterology, Gastroenterology Section, University Hospital of Padua, Padua I-35128, Italy
| |
Collapse
|
14
|
Bernier-Latmani J, Petrova TV. Intestinal lymphatic vasculature: structure, mechanisms and functions. Nat Rev Gastroenterol Hepatol 2017; 14:510-526. [PMID: 28655884 DOI: 10.1038/nrgastro.2017.79] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The mammalian intestine is richly supplied with lymphatic vasculature, which has functions ranging from maintenance of interstitial fluid balance to transport of antigens, antigen-presenting cells, dietary lipids and fat-soluble vitamins. In this Review, we provide in-depth information concerning the organization and structure of intestinal lymphatics, the current view of their developmental origins, as well as molecular mechanisms of intestinal lymphatic patterning and maintenance. We will also discuss physiological aspects of intestinal lymph flow regulation and the known and emerging roles of intestinal lymphatic vessels in human diseases, such as IBD, infection and cancer.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland
| | - Tatiana V Petrova
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland.,Swiss Institute for Experimental Cancer Research, School of Life Sciences, Swiss Federal Institute of Technology Lausanne, Route Cantonale 1015, Lausanne, Switzerland
| |
Collapse
|
15
|
Downregulation of CX 3CR1 ameliorates experimental colitis: evidence for CX 3CL1-CX 3CR1-mediated immune cell recruitment. Int J Colorectal Dis 2017; 32:315-324. [PMID: 27942903 DOI: 10.1007/s00384-016-2735-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/05/2016] [Indexed: 02/04/2023]
Abstract
PURPOSE Inflammatory conditions like inflammatory bowel diseases (IBD) are characterized by increased immune cell infiltration. The chemokine ligand CX3CL1 and its receptor CX3CR1 have been shown to be involved in leukocyte adhesion, transendothelial recruitment, and chemotaxis. Therefore, the objective of this study was to describe CX3CL1-CX3CR1-mediated signaling in the induction of immune cell recruitment during experimental murine colitis. METHODS Acute colitis was induced by dextran sodium sulfate (DSS), and sepsis was induced by injection of lipopolysaccharide (LPS). Serum concentrations of CX3CR1 and CX3CL1 were measured by ELISA. Wild-type and CX3CR1-/- mice were challenged with DSS, and on day 6, intravital microscopy was performed to monitor colonic leukocyte and platelet recruitment. Intestinal inflammation was assessed by disease activity, histopathology, and neutrophil infiltration. RESULTS CX3CR1 was upregulated in DSS colitis and LPS-induced sepsis. CX3CR1-/- mice were protected from disease severity and intestinal injury in DSS colitis, and CX3CR1 deficiency resulted in reduced rolling of leukocytes and platelets. CONCLUSIONS In the present study, we provide evidence for a crucial role of CX3CL1-CX3CR1 in experimental colitis, in particular for intestinal leukocyte recruitment during murine colitis. Our findings suggest that CX3CR1 blockade represents a potential therapeutic strategy for treatment of IBD.
Collapse
|
16
|
Becker F, Kurmaeva E, Gavins FNE, Stevenson EV, Navratil AR, Jin L, Tsunoda I, Orr AW, Alexander JS, Ostanin DV. A Critical Role for Monocytes/Macrophages During Intestinal Inflammation-associated Lymphangiogenesis. Inflamm Bowel Dis 2016; 22:1326-1345. [PMID: 26950310 PMCID: PMC4912566 DOI: 10.1097/mib.0000000000000731] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Inflammation-associated lymphangiogenesis (IAL) is frequently observed in inflammatory bowel diseases. IAL is believed to limit inflammation by enhancing fluid and immune cell clearance. Although monocytes/macrophages (MΦ) are known to contribute to intestinal pathology in inflammatory bowel disease, their role in intestinal IAL has never been studied mechanistically. We investigated contributions of monocytes/MΦ to the development of intestinal inflammation and IAL. METHODS Because inflammatory monocytes express CC chemokine receptor 2 (CCR2), we used CCR2 diphtheria toxin receptor transgenic (CCR2.DTR) mice, in which monocytes can be depleted by diphtheria toxin injection, and CCR2 mice, which have reduced circulating monocytes. Acute or chronic colitis was induced by dextran sodium sulfate or adoptive transfer of CD4CD45RB T cells, respectively. Intestinal inflammation was assessed by flow cytometry, immunofluorescence, disease activity, and histopathology, whereas IAL was assessed by lymphatic vessel morphology and density. RESULTS We demonstrated that intestinal MΦ expressed vascular endothelial growth factor-C/D. In acute colitis, monocyte-depleted mice were protected from intestinal injury and showed reduced IAL, which was reversed after transfer of wild-type monocytes into CCR2 mice. In chronic colitis, CCR2 deficiency did not attenuate inflammation but reduced IAL. CONCLUSIONS We propose a dual role of MΦ in (1) promoting acute inflammation and (2) contributing to IAL. Our data suggest that intestinal inflammation and IAL could occur independently, because IAL was reduced in the absence of monocytes/MΦ, even when inflammation was present. Future inflammatory bowel disease therapies might exploit promotion of IAL and suppression of MΦ independently, to restore lymphatic clearance and reduce inflammation.
Collapse
Affiliation(s)
- Felix Becker
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
- Department of General and Visceral Surgery, University Hospital Muenster, Germany
| | - Elvira Kurmaeva
- Department of Medicine, Center of Excellence for Arthritis and Rheumatology, Louisiana State University Health Sciences Center-Shreveport, Louisiana
| | - Felicity N. E. Gavins
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Emily V. Stevenson
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Aaron R. Navratil
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Long Jin
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Ikuo Tsunoda
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - A. Wayne Orr
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
- Department of Pathology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Jonathan S. Alexander
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Dmitry V. Ostanin
- Department of Medicine, Center of Excellence for Arthritis and Rheumatology, Louisiana State University Health Sciences Center-Shreveport, Louisiana
| |
Collapse
|
17
|
Okuda KS, Misa JP, Oehlers SH, Hall CJ, Ellett F, Alasmari S, Lieschke GJ, Crosier KE, Crosier PS, Astin JW. A zebrafish model of inflammatory lymphangiogenesis. Biol Open 2015; 4:1270-80. [PMID: 26369931 PMCID: PMC4610225 DOI: 10.1242/bio.013540] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a disabling chronic inflammatory disease of the gastrointestinal tract. IBD patients have increased intestinal lymphatic vessel density and recent studies have shown that this may contribute to the resolution of IBD. However, the molecular mechanisms involved in IBD-associated lymphangiogenesis are still unclear. In this study, we established a novel inflammatory lymphangiogenesis model in zebrafish larvae involving colitogenic challenge stimulated by exposure to 2,4,6-trinitrobenzenesulfonic acid (TNBS) or dextran sodium sulphate (DSS). Treatment with either TNBS or DSS resulted in vascular endothelial growth factor receptor (Vegfr)-dependent lymphangiogenesis in the zebrafish intestine. Reduction of intestinal inflammation by the administration of the IBD therapeutic, 5-aminosalicylic acid, reduced intestinal lymphatic expansion. Zebrafish macrophages express vascular growth factors vegfaa, vegfc and vegfd and chemical ablation of these cells inhibits intestinal lymphatic expansion, suggesting that the recruitment of macrophages to the intestine upon colitogenic challenge is required for intestinal inflammatory lymphangiogenesis. Importantly, this study highlights the potential of zebrafish as an inflammatory lymphangiogenesis model that can be used to investigate the role and mechanism of lymphangiogenesis in inflammatory diseases such as IBD.
Collapse
Affiliation(s)
- Kazuhide S Okuda
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - June Pauline Misa
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Stefan H Oehlers
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham 27710, USA
| | - Christopher J Hall
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Felix Ellett
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Sultan Alasmari
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Graham J Lieschke
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Kathryn E Crosier
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Philip S Crosier
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Jonathan W Astin
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
18
|
Al-Kofahi M, Becker F, Gavins FNE, Woolard MD, Tsunoda I, Wang Y, Ostanin D, Zawieja DC, Muthuchamy M, von der Weid PY, Alexander JS. IL-1β reduces tonic contraction of mesenteric lymphatic muscle cells, with the involvement of cycloxygenase-2 and prostaglandin E2. Br J Pharmacol 2015; 172:4038-4051. [PMID: 25989136 PMCID: PMC4543611 DOI: 10.1111/bph.13194] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 04/02/2015] [Accepted: 04/28/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE The lymphatic system maintains tissue homeostasis by unidirectional lymph flow, maintained by tonic and phasic contractions within subunits, 'lymphangions'. Here we have studied the effects of the inflammatory cytokine IL-1β on tonic contraction of rat mesenteric lymphatic muscle cells (RMLMC). EXPERIMENTAL APPROACH We measured IL-1β in colon-conditioned media (CM) from acute (AC-CM, dextran sodium sulfate) and chronic (CC-CM, T-cell transfer) colitis-induced mice and corresponding controls (Con-AC/CC-CM). We examined tonic contractility of RMLMC in response to CM, the cytokines h-IL-1β or h-TNF-α (5, 10, 20 ng·mL(-1) ), with or without COX inhibitors [TFAP (10(-5) M), diclofenac (0.2 × 10(-5) M)], PGE2 (10(-5) M)], IL-1-receptor antagonist, Anakinra (5 μg·mL(-1) ), or a selective prostanoid EP4 receptor antagonist, GW627368X (10(-6) and 10(-7) M). KEY RESULTS Tonic contractility of RMLMC was reduced by AC- and CC-CM compared with corresponding control culture media, Con-AC/CC-CM. IL-1β or TNF-α was not found in Con-AC/CC-CM, but detected in AC- and CC-CM. h-IL-1β concentration-dependently decreased RMLMC contractility, whereas h-TNF-α showed no effect. Anakinra blocked h-IL-1β-induced RMLMC relaxation, and with AC-CM, restored contractility to RMLMC. IL-1β increased COX-2 protein and PGE2 production in RMLMC.. PGE2 induced relaxations in RMLMC, comparable to h-IL-1β. Conversely, COX-2 and EP4 receptor inhibition reversed relaxation induced by IL-1β. CONCLUSIONS AND IMPLICATIONS The IL-1β-induced decrease in RMLMC tonic contraction was COX-2 dependent, and mediated by PGE2 . In experimental colitis, IL-1β and tonic lymphatic contractility were causally related, as this cytokine was critical for the relaxation induced by AC-CM and pharmacological blockade of IL-1β restored tonic contraction.
Collapse
Affiliation(s)
- M Al-Kofahi
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center – ShreveportShreveport, LA, USA
| | - F Becker
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center – ShreveportShreveport, LA, USA
- Department for General and Visceral Surgery, University Hospital MuensterMuenster, Germany
| | - F N E Gavins
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center – ShreveportShreveport, LA, USA
| | - M D Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center – ShreveportShreveport, LA, USA
| | - I Tsunoda
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center – ShreveportShreveport, LA, USA
| | - Y Wang
- Department of Obstetrics and Gynecology, Louisiana State University Health Sciences Center – ShreveportShreveport, LA, USA
| | - D Ostanin
- Department of Medicine, Division of Rheumatology, Louisiana State University Health Sciences Center – ShreveportShreveport, LA, USA
| | - D C Zawieja
- Department of Medicine, Cardiovascular Research Institute, Texas A&M Health Science CenterCollege Station, TX, USA
| | - M Muthuchamy
- Department of Medicine, Cardiovascular Research Institute, Texas A&M Health Science CenterCollege Station, TX, USA
| | - P Y von der Weid
- Department of Physiology and Pharmacology, University of CalgaryCalgary, Alberta, Canada
| | - J S Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center – ShreveportShreveport, LA, USA
| |
Collapse
|