51
|
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: 18] [Impact Index Per Article: 6.0] [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
|
52
|
Decreased Lymphangiogenic Activities and Genes Expression of Cord Blood Lymphatic Endothelial Progenitor Cells (VEGFR3 +/Pod +/CD11b + Cells) in Patient with Preeclampsia. Int J Mol Sci 2021; 22:ijms22084237. [PMID: 33921847 PMCID: PMC8073258 DOI: 10.3390/ijms22084237] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/12/2021] [Accepted: 04/16/2021] [Indexed: 12/26/2022] Open
Abstract
The abnormal development or disruption of the lymphatic vasculature has been implicated in metabolic and hypertensive diseases. Recent evidence suggests that the offspring exposed to preeclampsia (PE) in utero are at higher risk of long-term health problems, such as cardiovascular and metabolic diseases in adulthood, owing to in utero fetal programming. We aimed to investigate lymphangiogenic activities in the lymphatic endothelial progenitor cells (LEPCs) of the offspring of PE. Human umbilical cord blood LEPCs from pregnant women with severe PE (n = 10) and gestationally matched normal pregnancies (n = 10) were purified with anti-vascular endothelial growth factor receptor 3 (VEGFR3)/podoplanin/CD11b microbeads using a magnetic cell sorter device. LEPCs from PE displayed significantly delayed differentiation and reduced formation of lymphatic endothelial cell (LEC) colonies compared with the LEPCs from normal pregnancies. LECs differentiated from PE-derived LEPCs exhibited decreased tube formation, migration, proliferation, adhesion, wound healing, and 3D-sprouting activities as well as increased lymphatic permeability through the disorganization of VE-cadherin junctions, compared with the normal pregnancy-derived LECs. In vivo, LEPCs from PE showed significantly reduced lymphatic vessel formation compared to the LEPCs of the normal pregnancy. Gene expression analysis revealed that compared to the normal pregnancy-derived LECs, the PE-derived LECs showed a significant decrease in the expression of pro-lymphangiogenic genes (GREM1, EPHB3, VEGFA, AMOT, THSD7A, ANGPTL4, SEMA5A, FGF2, and GBX2). Collectively, our findings demonstrate, for the first time, that LEPCs from PE have reduced lymphangiogenic activities in vitro and in vivo and show the decreased expression of pro-lymphangiogenic genes. This study opens a new avenue for investigation of the molecular mechanism of LEPC differentiation and lymphangiogenesis in the offspring of PE and subsequently may impact the treatment of long-term health problems such as cardiovascular and metabolic disorders of offspring with abnormal development of lymphatic vasculature.
Collapse
|
53
|
Li W, Gauthier JM, Tong AY, Terada Y, Higashikubo R, Frye CC, Harrison MS, Hashimoto K, Bery AI, Ritter JH, Nava RG, Puri V, Wong BW, Lavine KJ, Bharat A, Krupnick AS, Gelman AE, Kreisel D. Lymphatic drainage from bronchus-associated lymphoid tissue in tolerant lung allografts promotes peripheral tolerance. J Clin Invest 2021; 130:6718-6727. [PMID: 33196461 DOI: 10.1172/jci136057] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 09/03/2020] [Indexed: 12/29/2022] Open
Abstract
Tertiary lymphoid organs are aggregates of immune and stromal cells including high endothelial venules and lymphatic vessels that resemble secondary lymphoid organs and can be induced at nonlymphoid sites during inflammation. The function of lymphatic vessels within tertiary lymphoid organs remains poorly understood. During lung transplant tolerance, Foxp3+ cells accumulate in tertiary lymphoid organs that are induced within the pulmonary grafts and are critical for the local downregulation of alloimmune responses. Here, we showed that tolerant lung allografts could induce and maintain tolerance of heterotopic donor-matched hearts through pathways that were dependent on the continued presence of the transplanted lung. Using lung retransplantation, we showed that Foxp3+ cells egressed from tolerant lung allografts via lymphatics and were recruited into donor-matched heart allografts. Indeed, survival of the heart allografts was dependent on lymphatic drainage from the tolerant lung allograft to the periphery. Thus, our work indicates that cellular trafficking from tertiary lymphoid organs regulates immune responses in the periphery. We propose that these findings have important implications for a variety of disease processes that are associated with the induction of tertiary lymphoid organs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jon H Ritter
- Pathology & Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | | | | | - Ankit Bharat
- Department of Surgery, Northwestern University, Chicago, Illinois, USA
| | | | - Andrew E Gelman
- Departments of Surgery.,Pathology & Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Daniel Kreisel
- Departments of Surgery.,Pathology & Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
54
|
Miyanaga T, Mizuguchi K, Hara S, Zoshima T, Inoue D, Nishioka R, Mizushima I, Ito K, Fuji H, Yamada K, Sato Y, Yanagita M, Kawano M. Tertiary lymphoid tissue in early-stage IgG4-related tubulointerstitial nephritis incidentally detected with a tumor lesion of the ureteropelvic junction: a case report. BMC Nephrol 2021; 22:34. [PMID: 33468063 PMCID: PMC7816437 DOI: 10.1186/s12882-021-02240-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 01/12/2021] [Indexed: 12/24/2022] Open
Abstract
Background IgG4-related kidney disease causes renal impairment of unknown pathogenesis that may progress to kidney failure. Although ectopic germinal centers contribute to the pathogenesis of the head and neck lesions of IgG4-related disease, the presence of tertiary lymphoid tissue (TLT) containing germinal centers in IgG4-RKD has rarely been reported. Case presentation We report a 72-year-old Japanese man who had IgG4-related tubulointerstitial nephritis (TIN) with TLT formation incidentally detected in a resected kidney with mass lesion of IgG4-related ureteritis in the ureteropelvic junction. During follow-up for past surgical resection of a bladder tumor, renal dysfunction developed and a ureter mass was found in the right ureteropelvic junction, which was treated by nephroureterectomy after chemotherapy. Pathology revealed no malignancy but abundant IgG4-positive cell infiltration, obliterative phlebitis and storiform fibrosis, confirming the diagnosis of IgG4-related ureteritis. In the resected right kidney, lymphoplasmacytes infiltrated the interstitium with focal distribution in the renal subcapsule and around medium vessels without storiform fibrosis, suggesting the very early stage of IgG4-TIN. Lymphocyte aggregates were also detected at these sites and consisted of B, T, and follicular dendritic cells, indicating TLT formation. IgG4-positive cells infiltrated around TLTs. Conclusions Our case suggests that TLT formation is related with the development of IgG4-TIN and our analysis of distribution of TLT have possibility to elucidate IgG4-TIN pathophysiology.
Collapse
Affiliation(s)
- Tatsuhito Miyanaga
- Division of Rheumatology, Department of Internal Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Keishi Mizuguchi
- Department of Diagnostic Pathology, Kanazawa University Hospital, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Satoshi Hara
- Division of Rheumatology, Department of Internal Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan.
| | - Takeshi Zoshima
- Division of Rheumatology, Department of Internal Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Dai Inoue
- Department of Radiology, Kanazawa University Graduate School of Medical Science, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Ryo Nishioka
- Division of Rheumatology, Department of Internal Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Ichiro Mizushima
- Division of Rheumatology, Department of Internal Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Kiyoaki Ito
- Division of Rheumatology, Department of Internal Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Hiroshi Fuji
- Division of Rheumatology, Department of Internal Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| | - Kazunori Yamada
- Department of Hematology and Immunology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, Japan
| | - Yuki Sato
- Department of Nephrology, Kyoto University Graduate School of Medicine, Yoshidakonoe-cho, Sakyo-ku, Kyoto, Japan.,Medical Innovation Center TMK Project, Graduate School of Medicine, Kyoto University, 53 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Motoko Yanagita
- Medical Innovation Center TMK Project, Graduate School of Medicine, Kyoto University, 53 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto, Japan.,Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Yoshida-honmachi, Sakyo-ku, Kyoto, Japan
| | - Mitsuhiro Kawano
- Division of Rheumatology, Department of Internal Medicine, 13-1 Takaramachi, Kanazawa, Ishikawa, Japan
| |
Collapse
|
55
|
Masum MA, Ichii O, Elewa YHA, Otani Y, Namba T, Kon Y. Vasculature-Associated Lymphoid Tissue: A Unique Tertiary Lymphoid Tissue Correlates With Renal Lesions in Lupus Nephritis Mouse Model. Front Immunol 2020; 11:595672. [PMID: 33384689 PMCID: PMC7770167 DOI: 10.3389/fimmu.2020.595672] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/09/2020] [Indexed: 12/30/2022] Open
Abstract
Lupus nephritis (LN) is a common complication in young patients and the most predominant cause of glomerulonephritis. Infiltrating immune cells and presence of immunocomplexes in the kidney are hallmarks of LN, which is closely associated with renal lesions (RLs). However, their regulatory mechanism in the kidney remains unclear, which is valuable for prevention of RL development. Here, we show the development of vasculature-associated lymphoid tissue (VALT) in LN, which is related to renal inflammatory cytokines, indicating that VALT is a unique tertiary lymphoid tissue. Transcriptomic analysis revealed different chemokines and costimulatory molecules for VALT induction and organization. Vascular and perivascular structures showed lymphoid tissue organization through lymphorganogenic chemokine production. Transcriptional profile and intracellular interaction also demonstrated antigen presentation, lymphocyte activity, clonal expansion, follicular, and germinal center activity in VALT. Importantly, VALT size was correlated with infiltrating immune cells in kidney and RLs, indicating its direct correlation with the development of RLs. In addition, dexamethasone administration reduced VALT size. Therefore, inhibition of VALT formation would be a novel therapeutic strategy against LN.
Collapse
Affiliation(s)
- Md Abdul Masum
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.,Department of Anatomy, Histology and Physiology, Faculty of Animal Science and Veterinary Medicine, Sher-e-Bangla Agricultural University, Dhaka, Bangladesh
| | - Osamu Ichii
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.,Laboratory of Agrobiomedical Science, Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Yaser Hosny Ali Elewa
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.,Department of Histology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Yuki Otani
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Takashi Namba
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Kon
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
56
|
Skin-Associated B Cells in the Pathogenesis of Cutaneous Autoimmune Diseases-Implications for Therapeutic Approaches. Cells 2020; 9:cells9122627. [PMID: 33297481 PMCID: PMC7762338 DOI: 10.3390/cells9122627] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022] Open
Abstract
B lymphocytes are crucial mediators of systemic immune responses and are known to be substantial in the pathogenesis of autoimmune diseases with cutaneous manifestations. Amongst them are lupus erythematosus, dermatomyositis, systemic sclerosis and psoriasis, and particularly those driven by autoantibodies such as pemphigus and pemphigoid. However, the concept of autoreactive skin-associated B cells, which may reside in the skin and locally contribute to chronic inflammation, is gradually evolving. These cells are believed to differ from B cells of primary and secondary lymphoid organs and may provide additional features besides autoantibody production, including cytokine expression and crosstalk to autoreactive T cells in an antigen-presenting manner. In chronically inflamed skin, B cells may appear in tertiary lymphoid structures. Those abnormal lymph node-like structures comprise a network of immune and stromal cells possibly enriched by vascular structures and thus constitute an ideal niche for local autoimmune responses. In this review, we describe current considerations of different B cell subsets and their assumed role in skin autoimmunity. Moreover, we discuss traditional and B cell-associated approaches for the treatment of autoimmune skin diseases, including drugs targeting B cells (e.g., CD19- and CD20-antibodies), plasma cells (e.g., proteasome inhibitors, CXCR4 antagonists), activated pathways (such as BTK- and PI3K-inhibitors) and associated activator molecules (BLyS, APRIL).
Collapse
|
57
|
Ma Y, Xia Z, Ye C, Lu C, Zhou S, Pan J, Liu C, Zhang J, Liu T, Hu T, Xie L, Wu G, Zhao Y. AGTR1 promotes lymph node metastasis in breast cancer by upregulating CXCR4/SDF-1α and inducing cell migration and invasion. Aging (Albany NY) 2020; 11:3969-3992. [PMID: 31219799 PMCID: PMC6628987 DOI: 10.18632/aging.102032] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 06/13/2019] [Indexed: 12/21/2022]
Abstract
The angiotensin II type I receptor (AGTR1) has a strong influence on tumor growth, angiogenesis, inflammation and immunity. However, the role of AGTR1 on lymph node metastasis (LNM) in breast cancer, which correlates with tumor progression and patient survival, has not been examined. AGTR1 was highly expressed in lymph node-positive tumor tissues, which was confirmed by the Oncomine database. Next, inhibition of AGTR1 reduced tumor growth and LNM in orthotopic xenografts by bioluminescence imaging (BLI). Losartan, an AGTR1-specific inhibitor, decreased the chemokine pair CXCR4/SDF-1α levels in vivo and inhibited AGTR1-induced cell migration and invasion in vitro. Finally, the molecular mechanism of AGTR1-induced cell migration and LNM was assessed by knocking down AGTR1 in normal cells or CXCR4 in AGTR1high cells. AGTR1-silenced cells treated with losartan showed lower CXCR4 expression. AGTR1 overexpression caused the upregulation of FAK/RhoA signaling molecules, while knocking down CXCR4 in AGTR1high cells downregulated these molecules. Collectively, AGTR1 promotes LNM by increasing the chemokine pair CXCR4/SDF-1α and tumor cell migration and invasion. The potential mechanism of AGTR1-mediated cell movement relies on activating the FAK/RhoA pathway. Our study indicated that inhibiting AGTR1 may be a potential therapeutic target for LNM in early-stage breast cancer.
Collapse
Affiliation(s)
- Yuxi Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zihan Xia
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chunmei Ye
- Department of Breast Surgery, Wuhan Women and Children's Health Care Center, Wuhan 430022, China
| | - Chong Lu
- Department of Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sheng Zhou
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Juan Pan
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cuiwei Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jieying Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ting Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Linka Xie
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanxia Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
58
|
Tokunaga R, Nakagawa S, Sakamoto Y, Nakamura K, Naseem M, Izumi D, Kosumi K, Taki K, Higashi T, Miyata T, Miyamoto Y, Yoshida N, Baba H, Lenz HJ. 12-Chemokine signature, a predictor of tumor recurrence in colorectal cancer. Int J Cancer 2020; 147:532-541. [PMID: 32191346 DOI: 10.1002/ijc.32982] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 02/06/2020] [Accepted: 02/12/2020] [Indexed: 12/19/2022]
Abstract
Tertiary lymphoid structures (TLSs) provide an immunological antineoplastic effect. Recent evidences link a unique 12-chemokine (CCL2, -3, -4, -5, -8, -18, -19, -21, CXCL9, -10, -11, -13) signature status from tumor tissue and the TLS expression. However, the potential significance of 12-chemokine signature status for clinical use is unknown. We aimed to evaluate the association of 12-chemokine signature status with patient outcomes in colorectal cancer (CRC). We used integrated data of resected 975 CRC cases within three independent cohorts from France, Japan and the United States (GSE39582, KUMAMOTO from Kumamoto university hospital and TCGA). The association of 12-chemokine signature status with clinicopathological features, patient outcome, TLS expression status and key tumor molecular features was analyzed. Patients with low 12-chemokine signature status had a significant shorter relapse-free survival in discovery cohort (HR: 1.61, 95% CI: 1.11-2.39, p = 0.0123), which was confirmed in validation cohort (HR: 3.31, 95% CI: 1.33-10.08, p = 0.0087). High 12-chemokine signature status had significant associations with right-sided tumor, high tumor-localized TLS expression, BRAF mutant, CIMP-high status and MSI-high status. Furthermore, RNA-seq based analysis showed that high 12-chemokine signature status was strongly associated with inflammation-related, immune cells-related and apoptosis pathways (using gene set enrichment analysis), and more tumor-infiltrating immune cells, such as cytotoxic T lymphocytes and myeloid dendritic cells (using MCP-counter analysis). We investigated a promising effect of 12-chemokine signature status in CRC patients who underwent resection. Our data may be helpful in developing novel immunological treatment strategies for CRC.
Collapse
Affiliation(s)
- Ryuma Tokunaga
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuo Sakamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Nakamura
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Madiha Naseem
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daisuke Izumi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keisuke Kosumi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsunobu Taki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takaaki Higashi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tatsunori Miyata
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
59
|
Willard-Mack CL, Elmore SA, Hall WC, Harleman J, Kuper CF, Losco P, Rehg JE, Rühl-Fehlert C, Ward JM, Weinstock D, Bradley A, Hosokawa S, Pearse G, Mahler BW, Herbert RA, Keenan CM. Nonproliferative and Proliferative Lesions of the Rat and Mouse Hematolymphoid System. Toxicol Pathol 2020; 47:665-783. [PMID: 31526133 DOI: 10.1177/0192623319867053] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The INHAND Project (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions in Rats and Mice) is a joint initiative of the Societies of Toxicologic Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP), and North America (STP) to develop an internationally accepted nomenclature for proliferative and nonproliferative changes in rats and mice. The purpose of this publication is to provide a standardized nomenclature for classifying changes observed in the hematolymphoid organs, including the bone marrow, thymus, spleen, lymph nodes, mucosa-associated lymphoid tissues, and other lymphoid tissues (serosa-associated lymphoid clusters and tertiary lymphoid structures) with color photomicrographs illustrating examples of the lesions. Sources of material included histopathology databases from government, academia, and industrial laboratories throughout the world. Content includes spontaneous lesions as well as lesions induced by exposure to test materials. The nomenclature for these organs is divided into 3 terminologies: descriptive, conventional, and enhanced. Three terms are listed for each diagnosis. The rationale for this approach and guidance for its application to toxicologic pathology are described in detail below.
Collapse
Affiliation(s)
| | - Susan A Elmore
- Thymus subgroup lead.,National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | | - Johannes Harleman
- Lymph node subgroup lead.,Neoplasm subgroup leads.,Independent Consultant, Darmstadt, Germany
| | - C Frieke Kuper
- Associated lymphoid organs subgroup lead.,Independent Consultant, Utrecht, the Netherlands
| | - Patricia Losco
- General hematolymphoid subgroup lead.,Independent Consultant, West Chester, PA, USA
| | - Jerold E Rehg
- Spleen subgroup leads.,Neoplasm subgroup leads.,Saint Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Jerrold M Ward
- Spleen subgroup leads.,Neoplasm subgroup leads.,Global VetPathology, Montgomery Village, MD, USA
| | | | - Alys Bradley
- Charles River Laboratories, Tranent, Scotland, United Kingdom
| | - Satoru Hosokawa
- Eisai Co, Ltd, Drug Safety Research Laboratories, Ibaraki, Japan
| | | | - Beth W Mahler
- Experimental Pathology Laboratories, Research Triangle Park, NC, USA
| | - Ronald A Herbert
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | |
Collapse
|
60
|
Helmink BA, Reddy SM, Gao J, Zhang S, Basar R, Thakur R, Yizhak K, Sade-Feldman M, Blando J, Han G, Gopalakrishnan V, Xi Y, Zhao H, Amaria RN, Tawbi HA, Cogdill AP, Liu W, LeBleu VS, Kugeratski FG, Patel S, Davies MA, Hwu P, Lee JE, Gershenwald JE, Lucci A, Arora R, Woodman S, Keung EZ, Gaudreau PO, Reuben A, Spencer CN, Burton EM, Haydu LE, Lazar AJ, Zapassodi R, Hudgens CW, Ledesma DA, Ong S, Bailey M, Warren S, Rao D, Krijgsman O, Rozeman EA, Peeper D, Blank CU, Schumacher TN, Butterfield LH, Zelazowska MA, McBride KM, Kalluri R, Allison J, Petitprez F, Fridman WH, Sautès-Fridman C, Hacohen N, Rezvani K, Sharma P, Tetzlaff MT, Wang L, Wargo JA. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020; 577:549-555. [DOI: 10.1038/s41586-019-1922-8] [Citation(s) in RCA: 863] [Impact Index Per Article: 215.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 12/04/2019] [Indexed: 12/28/2022]
|
61
|
Aging Induces an Nlrp3 Inflammasome-Dependent Expansion of Adipose B Cells That Impairs Metabolic Homeostasis. Cell Metab 2019; 30:1024-1039.e6. [PMID: 31735593 PMCID: PMC6944439 DOI: 10.1016/j.cmet.2019.10.006] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 08/10/2019] [Accepted: 10/15/2019] [Indexed: 12/23/2022]
Abstract
During aging, visceral adiposity is often associated with alterations in adipose tissue (AT) leukocytes, inflammation, and metabolic dysfunction. However, the contribution of AT B cells in immunometabolism during aging is unexplored. Here, we show that aging is associated with an expansion of a unique population of resident non-senescent aged adipose B cells (AABs) found in fat-associated lymphoid clusters (FALCs). AABs are transcriptionally distinct from splenic age-associated B cells (ABCs) and show greater expansion in female mice. Functionally, whole-body B cell depletion restores proper lipolysis and core body temperature maintenance during cold stress. Mechanistically, the age-induced FALC formation, AAB, and splenic ABC expansion is dependent on the Nlrp3 inflammasome. Furthermore, AABs express IL-1R, and inhibition of IL-1 signaling reduces their proliferation and increases lipolysis in aging. These data reveal that inhibiting Nlrp3-dependent B cell accumulation can be targeted to reverse metabolic impairment in aging AT.
Collapse
|
62
|
Jeucken KCM, Koning JJ, Mebius RE, Tas SW. The Role of Endothelial Cells and TNF-Receptor Superfamily Members in Lymphoid Organogenesis and Function During Health and Inflammation. Front Immunol 2019; 10:2700. [PMID: 31824495 PMCID: PMC6879661 DOI: 10.3389/fimmu.2019.02700] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/04/2019] [Indexed: 01/02/2023] Open
Abstract
Lymph nodes (LNs) are crucial for the orchestration of immune responses. LN reactions depend on interactions between incoming and local immune cells, and stromal cells. To mediate these cellular interactions an organized vascular network within the LN exists. In general, the LN vasculature can be divided into two components: blood vessels, which include the specialized high endothelial venules that recruit lymphocytes from the bloodstream, and lymphatic vessels. Signaling via TNF receptor (R) superfamily (SF) members has been implicated as crucial for the development and function of LNs and the LN vasculature. In recent years the role of cell-specific signaling of TNFRSF members in different endothelial cell (EC) subsets and their roles in development and maintenance of lymphoid organs has been elucidated. Here, we discuss recent insights into EC-specific TNFRSF member signaling and highlight its importance in different EC subsets in LN organogenesis and function during health, and in lymphocyte activation and tertiary lymphoid structure formation during inflammation.
Collapse
Affiliation(s)
- Kim C M Jeucken
- Amsterdam Rheumatology and Immunology Center (ARC), Department of Rheumatology and Clinical Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jasper J Koning
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center (ARC), Department of Rheumatology and Clinical Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
63
|
Abstract
Immunosenescence involves a series of ageing-induced alterations in the immune system and is characterized by two opposing hallmarks: defective immune responses and increased systemic inflammation. The immune system is modulated by intrinsic and extrinsic factors and undergoes profound changes in response to the ageing process. Immune responses are therefore highly age-dependent. Emerging data show that immunosenescence underlies common mechanisms responsible for several age-related diseases and is a plastic state that can be modified and accelerated by non-heritable environmental factors and pharmacological intervention. In the kidney, resident macrophages and fibroblasts are continuously exposed to components of the external environment, and the effects of cellular reprogramming induced by local immune responses, which accumulate with age, might have a role in the increased susceptibility to kidney disease among elderly individuals. Additionally, because chronic kidney disease, especially end-stage renal disease, is often accompanied by immunosenescence, which affects these patients independently of age, and many kidney diseases are strongly age-associated, treatment approaches that target immunosenescence might be particularly clinically relevant.
Collapse
|
64
|
A potential role of toll-like receptors, IFN-γ and the phosphatidylinositol 3-kinase pathway in the pathogenesis of acquired mediastinal lymphatic malformation. Med Hypotheses 2019; 131:109287. [PMID: 31443764 DOI: 10.1016/j.mehy.2019.109287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/23/2019] [Indexed: 11/20/2022]
Abstract
Sarcoidosis is a multisystem disorder with non-caseating granulomas in various organs. The etiology of sarcoid granuloma formation is not clear and likely an antigen-induced process. We came across a previously treated sarcoidosis patient who presented with worsening dyspnea on exertion for several months and several days of difficulty swallowing. On Chest CT imaging, large posterior mediastinal mass was found that subsequently diagnosed as macrocystic lymphatic malformation after surgical resection. Pathophysiology of development of acquired lymphatic malformations in a sarcoidosis patient is currently not clear. We hypothesize there might be a complex interplay of Toll-like receptors, IFN-γ and the phosphatidylinositol 3-kinase pathway in the pathogenesis.
Collapse
|
65
|
Frasca D, Blomberg BB. Adipose Tissue: A Tertiary Lymphoid Organ: Does It Change with Age? Gerontology 2019; 66:114-121. [PMID: 31412335 DOI: 10.1159/000502036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/10/2019] [Indexed: 12/16/2022] Open
Abstract
In this manuscript, we summarize published results showing that obesity and aging are inflammatory conditions associated with serious health problems, increased risk for disease and death. We show that fat mass increases with age and represents a major contributor to insulin resistance and the metabolic syndrome. We summarize the effects of age on the adipose tissue (AT), related to the abundance, distribution, cellular composition, endocrine signaling and function of the tissue. The AT is an immunological tissue, with several hallmarks of innate and adaptive immune responses. We show that in both mice and humans, the AT is heavily infiltrated by immune cells that have receptors for pro-inflammatory cytokines and chemokines secreted by the adipocytes and also by the immune cells that have infiltrated the AT. We also show that the AT provides an environment for the secretion of IgG antibodies with anti-self (autoimmune) reactivity. As we have previously shown, this is due to the release of self antigens following cell death due to hypoxia, as well as to the expression of activation-induced cytidine deaminase, the enzyme of class switch recombination, and the transcription factor T-bet by the resident B cells, which also express the membrane marker CD11c, both involved in the production of autoimmune IgG antibodies. We show data in support of the AT as a tertiary lymphoid organ (TLO), showing the examples of TLOs that develop within the AT, such as fat-associated lymphoid clusters and milky spots, as well as artery TLOs that develop in the adventitia areas of the aorta.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA,
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
66
|
Pfuderer PL, Ballhausen A, Seidler F, Stark HJ, Grabe N, Frayling IM, Ager A, von Knebel Doeberitz M, Kloor M, Ahadova A. High endothelial venules are associated with microsatellite instability, hereditary background and immune evasion in colorectal cancer. Br J Cancer 2019; 121:395-404. [PMID: 31358939 PMCID: PMC6738093 DOI: 10.1038/s41416-019-0514-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Microsatellite-unstable (MSI) tumours show a high load of mutational neoantigens, as a consequence of DNA mismatch repair deficiency. Consequently, MSI tumours commonly present with dense immune infiltration and develop immune evasion mechanisms. Whether improved lymphocyte recruitment contributes to the pronounced immune infiltration in MSI tumours is unknown. We analysed the density of high endothelial venules (HEV) and postcapillary blood vessels specialised for lymphocyte trafficking, in MSI colorectal cancers (CRC). METHODS HEV density was determined by immunohistochemical staining of FFPE tissue sections from MSI (n = 48) and microsatellite-stable (MSS, n = 35) CRCs. Associations with clinical and pathological variables were analysed. RESULTS We found elevated HEV densities in MSI compared with MSS CRCs (median 0.049 vs 0.000 counts/mm2, respectively, p = 0.0002), with the highest densities in Lynch syndrome MSI CRCs. Dramatically elevated HEV densities were observed in B2M-mutant Lynch syndrome CRCs, pointing towards a link between lymphocyte recruitment and immune evasion (median 0.485 vs 0.0885 counts/mm2 in B2M-wild-type tumours, p = 0.0237). CONCLUSIONS Our findings for the first time indicate a significant contribution of lymphocyte trafficking in immune responses against MSI CRC, particularly in the context of Lynch syndrome. High HEV densities in B2M-mutant tumours underline the significance of immunoediting during tumour evolution.
Collapse
Affiliation(s)
- Pauline L Pfuderer
- Department of Applied Tumour Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumour Biology, DKFZ, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University Hospital Heidelberg, Heidelberg, Germany
| | - Alexej Ballhausen
- Department of Applied Tumour Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumour Biology, DKFZ, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University Hospital Heidelberg, Heidelberg, Germany
| | - Florian Seidler
- Department of Applied Tumour Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumour Biology, DKFZ, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University Hospital Heidelberg, Heidelberg, Germany
| | - Hans-Jürgen Stark
- Department of Applied Tumour Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumour Biology, DKFZ, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University Hospital Heidelberg, Heidelberg, Germany
| | - Niels Grabe
- Hamamatsu Tissue Imaging and Analysis (TIGA) Center, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumour Diseases (NCT), Heidelberg, Germany
| | - Ian M Frayling
- Inherited Tumour Syndromes Research Group, Institute of Cancer & Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - Ann Ager
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Magnus von Knebel Doeberitz
- Department of Applied Tumour Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumour Biology, DKFZ, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumour Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumour Biology, DKFZ, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University Hospital Heidelberg, Heidelberg, Germany
| | - Aysel Ahadova
- Department of Applied Tumour Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.
- Clinical Cooperation Unit Applied Tumour Biology, DKFZ, Heidelberg, Germany.
- Molecular Medicine Partnership Unit (MMPU), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
67
|
Sarfarazi A, Lee G, Mirjalili SA, Phillips ARJ, Windsor JA, Trevaskis NL. Therapeutic delivery to the peritoneal lymphatics: Current understanding, potential treatment benefits and future prospects. Int J Pharm 2019; 567:118456. [PMID: 31238102 DOI: 10.1016/j.ijpharm.2019.118456] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/21/2019] [Accepted: 06/21/2019] [Indexed: 12/20/2022]
Abstract
The interest in approaches to deliver therapeutics to the lymphatic system has increased in recent years as the lymphatics have been discovered to play an important role in a range of disease states such as cancer metastases, inflammatory and metabolic disease, and acute and critical illness. Therapeutic delivery to lymph has the potential to enhance treatment of these conditions. Currently much of the existing data explores therapeutic delivery to the lymphatic vessels and nodes that drain peripheral tissues and the intestine. Relatively little focus has been given to understanding the anatomy, function and therapeutic delivery to the peritoneal lymphatics. Gaining a better understanding of peritoneal lymphatic structure and function would contribute to the understanding of disease processes involving these lymphatics and facilitate the development of delivery systems to target therapeutics to the peritoneal lymphatics. This review explores the basic anatomy and ultrastructure of the peritoneal lymphatics system, the lymphatic drainage pathways from the peritoneum, and therapeutic and delivery system characteristics (size, lipophilicity and surface properties) that favour lymph uptake and retention after intraperitoneal delivery. Finally, techniques that can be used to quantify uptake into peritoneal lymph are outlined, providing a platform for future studies.
Collapse
Affiliation(s)
- Ali Sarfarazi
- Surgical and Translational Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Applied Surgery and Metabolism Laboratory, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Given Lee
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - S Ali Mirjalili
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Anthony R J Phillips
- Surgical and Translational Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Applied Surgery and Metabolism Laboratory, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - John A Windsor
- Surgical and Translational Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Applied Surgery and Metabolism Laboratory, School of Biological Sciences, University of Auckland, Auckland, New Zealand; HBP/Upper GI Unit, Department of General Surgery, Auckland City Hospital, Auckland, New Zealand
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia.
| |
Collapse
|
68
|
Lisovska N, Shanazarov N. Tumor progression mechanisms: Insights from the central immune regulation of tissue homeostasis. Oncol Lett 2019; 17:5311-5318. [PMID: 31186747 PMCID: PMC6507387 DOI: 10.3892/ol.2019.10218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/07/2019] [Indexed: 11/29/2022] Open
Abstract
Knowledge of the mechanisms underlying the spread of cancer at the cellular and molecular levels is expanding rapidly. However, the central regulators governing the initiation and the rate of tumor growth remain poorly established. The fundamental principles of innate and adaptive immunity may explain how immune cells generate a specific response to tumor tissue. In the current review, the functional features of the immune system that contribute to the maintenance of normal tissue homeostasis, as well as their disruption in malignant transformations, were analyzed. Experimental and clinical studies previously demonstrated the involvement of regulatory T-cells in the process of tumor metastasis in a tissue-specific manner. An understanding of the cross talk between lymphoid and tumor cells may provide an insight into cancer evolution in terms of the mechanisms of T-cell competency formation. Elucidating the mechanisms of tumor progression via central immune regulation has implications for the development of novel therapeutic agents that target immune checkpoints.
Collapse
Affiliation(s)
- Natalya Lisovska
- Department of Chemotherapy, Center of Oncology, Cyber Clinic of Spizhenko, Kyiv 08112, Ukraine, Republic of Kazakhstan
| | - Nasrulla Shanazarov
- Department of General Surgery, Medical Center Hospital of The President's Affairs Administration of The Republic of Kazakhstan, Astana 010000, Republic of Kazakhstan
| |
Collapse
|
69
|
Chakraborty A, Barajas S, Lammoglia GM, Reyna AJ, Morley TS, Johnson JA, Scherer PE, Rutkowski JM. Vascular Endothelial Growth Factor-D (VEGF-D) Overexpression and Lymphatic Expansion in Murine Adipose Tissue Improves Metabolism in Obesity. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:924-939. [PMID: 30878136 DOI: 10.1016/j.ajpath.2018.12.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/13/2018] [Accepted: 12/11/2018] [Indexed: 12/18/2022]
Abstract
Obese adipose tissue expansion is an inflammatory process that results in dysregulated lipolysis, increased circulating lipids, ectopic lipid deposition, and systemic insulin resistance. Lymphatic vessels provide a route of fluid, macromolecule, and immune cell clearance, and lymphangiogenesis increases this capability. Indeed, inflammation-associated lymphangiogenesis is critical in resolving acute and chronic inflammation, but it is largely absent in obese adipose tissue. Enhancing adipose tissue lymphangiogenesis could, therefore, improve metabolism in obesity. To test this hypothesis, transgenic mice with doxycycline-inducible expression of murine vascular endothelial growth factor (VEGF)-D under a tightly controlled Tet-On promoter were crossed with adipocyte-specific adiponectin-reverse tetracycline-dependent transactivator mice (Adipo-VD) to stimulate adipose tissue-specific lymphangiogenesis during 16-week high-fat diet-induced obesity. Adipose VEGF-D overexpression induced de novo lymphangiogenesis in murine adipose tissue, and obese Adipo-VD mice exhibited enhanced glucose clearance, lower insulin levels, and reduced liver triglycerides. On β-3 adrenergic stimulation, Adipo-VD mice exhibited more rapid and increased glycerol flux from adipose tissue, suggesting that the lymphatics are a potential route of glycerol clearance. Resident macrophage crown-like structures were scarce and total F4/80+ macrophages were reduced in obese Adipo-VD s.c. adipose tissue with evidence of increased immune trafficking from the tissue. Augmenting VEGF-D signaling and lymphangiogenesis specifically in adipose tissue, therefore, reduces obesity-associated immune accumulation and improves metabolic responsiveness.
Collapse
Affiliation(s)
- Adri Chakraborty
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M College of Medicine, College Station
| | - Sheridan Barajas
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M College of Medicine, College Station
| | - Gabriela M Lammoglia
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M College of Medicine, College Station
| | - Andrea J Reyna
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M College of Medicine, College Station
| | - Thomas S Morley
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Joshua A Johnson
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Joseph M Rutkowski
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M College of Medicine, College Station.
| |
Collapse
|
70
|
Asam S, Neag G, Berardicurti O, Gardner D, Barone F. The role of stroma and epithelial cells in primary Sjögren's syndrome. Rheumatology (Oxford) 2019; 60:3503-3512. [PMID: 30945742 DOI: 10.1093/rheumatology/kez050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/19/2018] [Indexed: 12/27/2022] Open
Abstract
Primary SS (pSS) is a chronic autoimmune condition characterized by infiltration of the exocrine glands and systemic B cell hyperactivation. This glandular infiltration is associated with loss of glandular function, with pSS patients primarily presenting with severe dryness of the eyes and mouth. Within the affected glands, the infiltrating lymphocytes are organized in tertiary lymphoid structures. Tertiary lymphoid structures subvert normal tissue architecture and impact on organ function, by promoting the activation and maintenance of autoreactive lymphocytes. This review summarizes the current knowledge about the role of stromal cells (including endothelium, epithelium, nerves and fibroblasts) in the pathogenesis of pSS, in particular the interactions taking place between stromal cells and infiltrating lymphocytes. We will provide evidences pointing towards the driving role of stromal cells in the orchestration of the local inflammatory milieu, thus highlighting the need for therapies aimed at targeting this compartment alongside classical immunosuppression in pSS.
Collapse
Affiliation(s)
- Saba Asam
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Georgiana Neag
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | | | - David Gardner
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Francesca Barone
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
71
|
Witzel II, Nasser R, Garcia-Sabaté A, Sapudom J, Ma C, Chen W, Teo JCM. Deconstructing Immune Microenvironments of Lymphoid Tissues for Reverse Engineering. Adv Healthc Mater 2019; 8:e1801126. [PMID: 30516005 DOI: 10.1002/adhm.201801126] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/25/2018] [Indexed: 01/01/2023]
Abstract
The immune microenvironment presents a diverse panel of cues that impacts immune cell migration, organization, differentiation, and the immune response. Uniquely, both the liquid and solid phases of every specific immune niche within the body play an important role in defining cellular functions in immunity at that particular location. The in vivo immune microenvironment consists of biomechanical and biochemical signals including their gradients, surface topography, dimensionality, modes of ligand presentation, and cell-cell interactions, and the ability to recreate these immune biointerfaces in vitro can provide valuable insights into the immune system. This manuscript reviews the critical roles played by different immune cells and surveys the current progress of model systems for reverse engineering of immune microenvironments with a focus on lymphoid tissues.
Collapse
Affiliation(s)
- Ini-Isabée Witzel
- Core Technology Platforms; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Rasha Nasser
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Anna Garcia-Sabaté
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
- Department of Biomedical Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| | - Jeremy C. M. Teo
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| |
Collapse
|
72
|
Breslin JW, Yang Y, Scallan JP, Sweat RS, Adderley SP, Murfee WL. Lymphatic Vessel Network Structure and Physiology. Compr Physiol 2018; 9:207-299. [PMID: 30549020 PMCID: PMC6459625 DOI: 10.1002/cphy.c180015] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The lymphatic system is comprised of a network of vessels interrelated with lymphoid tissue, which has the holistic function to maintain the local physiologic environment for every cell in all tissues of the body. The lymphatic system maintains extracellular fluid homeostasis favorable for optimal tissue function, removing substances that arise due to metabolism or cell death, and optimizing immunity against bacteria, viruses, parasites, and other antigens. This article provides a comprehensive review of important findings over the past century along with recent advances in the understanding of the anatomy and physiology of lymphatic vessels, including tissue/organ specificity, development, mechanisms of lymph formation and transport, lymphangiogenesis, and the roles of lymphatics in disease. © 2019 American Physiological Society. Compr Physiol 9:207-299, 2019.
Collapse
Affiliation(s)
- Jerome W. Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Joshua P. Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Richard S. Sweat
- Department of Biomedical Engineering, Tulane University, New Orleans, LA
| | - Shaquria P. Adderley
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - W. Lee Murfee
- Department of Biomedical Engineering, University of Florida, Gainesville, FL
| |
Collapse
|
73
|
Chatterjee G, Pai T, Hardiman T, Avery-Kiejda K, Scott RJ, Spencer J, Pinder SE, Grigoriadis A. Molecular patterns of cancer colonisation in lymph nodes of breast cancer patients. Breast Cancer Res 2018; 20:143. [PMID: 30458865 PMCID: PMC6247766 DOI: 10.1186/s13058-018-1070-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Lymph node (LN) metastasis is an important prognostic parameter in breast carcinoma, a crucial site for tumour–immune cell interaction and a gateway for further dissemination of tumour cells to other metastatic sites. To gain insight into the underlying molecular changes from the pre-metastatic, via initial colonisation to the fully involved LN, we reviewed transcriptional research along the evolving microenvironment of LNs in human breast cancers patients. Gene expression studies were compiled and subjected to pathway-based analyses, with an emphasis on immune cell-related genes. Of 366 studies, 14 performed genome-wide gene expression comparisons and were divided into six clinical-biological scenarios capturing different stages of the metastatic pathway in the LN, as follows: metastatically involved LNs are compared to their patient-matched primary breast carcinomas (scenario 1) or the normal breast tissue (scenario 2). In scenario 3, uninvolved LNs were compared between LN-positive patients and LN-negative patients. Scenario 4 homed in on the residual uninvolved portion of involved LNs and compared it to the patient-matched uninvolved LNs. Scenario 5 contrasted uninvolved and involved LNs, whilst in scenario 6 involved (sentinel) LNs were assessed between patients with other either positive or negative LNs (non-sentinel). Gene lists from these chronological steps of LN metastasis indicated that gene patterns reflecting deficiencies in dendritic cells and hyper-proliferation of B cells parallel to tumour promoting pathways, including cell adhesion, extracellular matrix remodelling, cell motility and DNA repair, play key roles in the changing microenvironment of a pro-metastatic to a metastatically involved LN. Similarities between uninvolved LNs and the residual uninvolved portion of involved LNs hinted that LN alterations expose systemic tumour-related immune responses in breast cancer patients. Despite the diverse settings, gene expression patterns at different stages of metastatic colonisation in LNs were recognised and may provide potential avenues for clinical interventions to counteract disease progression for breast cancer patients.
Collapse
Affiliation(s)
- Gaurav Chatterjee
- Cancer Bioinformatics, King's College London, Innovation Hub, Cancer Centre at Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.,School of Cancer & Pharmaceutical Sciences, CRUK King's Health Partners Centre, King's College London, Innovation Hub, Comprehensive Cancer Centre at Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.,Department of Pathology, Tata Memorial Centre, 8th Floor, Annexe Building, Mumbai, India
| | - Trupti Pai
- Cancer Bioinformatics, King's College London, Innovation Hub, Cancer Centre at Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.,School of Cancer & Pharmaceutical Sciences, CRUK King's Health Partners Centre, King's College London, Innovation Hub, Comprehensive Cancer Centre at Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.,Department of Pathology, Tata Memorial Centre, 8th Floor, Annexe Building, Mumbai, India
| | - Thomas Hardiman
- Cancer Bioinformatics, King's College London, Innovation Hub, Cancer Centre at Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.,School of Cancer & Pharmaceutical Sciences, CRUK King's Health Partners Centre, King's College London, Innovation Hub, Comprehensive Cancer Centre at Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Kelly Avery-Kiejda
- Priority Research Centre for Cancer, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Rodney J Scott
- Priority Research Centre for Cancer, School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Jo Spencer
- Peter Gorer Department of Immunobiology, King's College London, Guy's Hospital, 2nd Floor, Borough Wing, London, SE1 9RT, UK
| | - Sarah E Pinder
- School of Cancer & Pharmaceutical Sciences, CRUK King's Health Partners Centre, King's College London, Innovation Hub, Comprehensive Cancer Centre at Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Anita Grigoriadis
- Cancer Bioinformatics, King's College London, Innovation Hub, Cancer Centre at Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK. .,School of Cancer & Pharmaceutical Sciences, CRUK King's Health Partners Centre, King's College London, Innovation Hub, Comprehensive Cancer Centre at Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK. .,Breast Cancer Now Research Unit, Innovation Hub, Cancer Centre at Guy's Hospital, King's College London, Faculty of Life Sciences and Medicine, London, SE1 9RT, UK.
| |
Collapse
|
74
|
Chen M, Lin X, Olsen N, He X, Zheng SG. Advances in T follicular helper and T follicular regulatory cells in transplantation immunity. Transplant Rev (Orlando) 2018; 32:187-193. [PMID: 30139705 DOI: 10.1016/j.trre.2018.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/08/2018] [Accepted: 07/20/2018] [Indexed: 12/14/2022]
Abstract
B cells play a crucial role in alloreactivity of organ transplant rejection and graft versus host diseases (GVHD). Over the past decade, it has been well recognized that B-cell infiltration in allografts and de novo donor-specific antibodies (DSA) were strongly associated with severe graft rejection and loss, as well as glucocorticoid resistance. Emerging evidence has demonstrated that Follicular T helper (Tfh) cells are key effectors to promote the proliferation and differentiation of B cells into antibody-producing plasmablasts and memory B cells. T-follicular regulatory (Tfr) cells are a recently recognized cell population that has a negative regulatory role on Tfh cells in the follicle, preventing incessant antibody production. It is still less clear how those humoral immunoreactive cells affect transplant rejection and allograft loss. This review focuses on the production and function of Tfr/Tfh cells in the transplant environment. Better understanding of the functions and mechanisms of Tfr/Tfh cells will help to design new strategies to prevent allograft rejection and prolong graft survival.
Collapse
Affiliation(s)
- Maogen Chen
- Organ transplant center, First affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou 510080, PR China; Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou 510080, PR China
| | - Xiaohong Lin
- Division of general surgery, The Eastern Hospital of the First affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China
| | - Nancy Olsen
- Division of Rheumatology, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Xiaoshun He
- Organ transplant center, First affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China; Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou 510080, PR China; Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou 510080, PR China
| | - Song Guo Zheng
- Division of Rheumatology, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033, USA.
| |
Collapse
|
75
|
Pipi E, Nayar S, Gardner DH, Colafrancesco S, Smith C, Barone F. Tertiary Lymphoid Structures: Autoimmunity Goes Local. Front Immunol 2018; 9:1952. [PMID: 30258435 PMCID: PMC6143705 DOI: 10.3389/fimmu.2018.01952] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 08/07/2018] [Indexed: 12/18/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are frequently observed in target organs of autoimmune diseases. TLS present features of secondary lymphoid organs such as segregated T and B cell zones, presence of follicular dendritic cell networks, high endothelial venules and specialized lymphoid fibroblasts and display the mechanisms to support local adaptive immune responses toward locally displayed antigens. TLS detection in the tissue is often associated with poor prognosis of disease, auto-antibody production and malignancy development. This review focuses on the contribution of TLS toward the persistence of the inflammatory drive, the survival of autoreactive lymphocyte clones and post-translational modifications, responsible for the pathogenicity of locally formed autoantibodies, during autoimmune disease development.
Collapse
Affiliation(s)
- Elena Pipi
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,Experimental Medicine Unit, Immuno-Inflammation Therapeutic Area, GSK Medicines Research Centre, Stevenage, United Kingdom
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - David H Gardner
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | | | - Charlotte Smith
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
76
|
Rivellese F, Mauro D, Nerviani A, Pagani S, Fossati-Jimack L, Messemaker T, Kurreeman FAS, Toes REM, Ramming A, Rauber S, Schett G, Jones GW, Jones SA, Rossi FW, de Paulis A, Marone G, El Shikh MEM, Humby F, Pitzalis C. Mast cells in early rheumatoid arthritis associate with disease severity and support B cell autoantibody production. Ann Rheum Dis 2018; 77:1773-1781. [PMID: 30127058 DOI: 10.1136/annrheumdis-2018-213418] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Mast cells (MCs) are involved in the pathogenesis of rheumatoid arthritis (RA). However, their contribution remains controversial. To establish their role in RA, we analysed their presence in the synovium of treatment-naïve patients with early RA and their association and functional relationship with histological features of synovitis. METHODS Synovial tissue was obtained by ultrasound-guided biopsy from treatment-naïve patients with early RA (n=99). Immune cells (CD3/CD20/CD138/CD68) and their relationship with CD117+MCs in synovial tissue were analysed by immunohistochemistry (IHC) and immunofluorescence (IF). The functional involvement of MCs in ectopic lymphoid structures (ELS) was investigated in vitro, by coculturing MCs with naïve B cells and anticitrullinated protein antibodies (ACPA)-producing B cell clones, and in vivo in interleukin-27 receptor alpha (IL27ra)-deficient and control mice during antigen-induced arthritis (AIA). RESULTS High synovial MC counts are associated with local and systemic inflammation, autoantibody positivity and high disease activity. IHC/IF showed that MCs reside at the outer border of lymphoid aggregates. Furthermore, human MCs promote the activation and differentiation of naïve B cells and induce the production of ACPA, mainly via contact-dependent interactions. In AIA, synovial MC numbers increase in IL27ra deficient mice, in association with ELS and worse disease activity. CONCLUSIONS Synovial MCs identify early RA patients with a severe clinical form of synovitis characterised by the presence of ELS.
Collapse
Affiliation(s)
- Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Daniele Mauro
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alessandra Nerviani
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sara Pagani
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Liliane Fossati-Jimack
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tobias Messemaker
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Fina A S Kurreeman
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - René E M Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andreas Ramming
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Simon Rauber
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gareth W Jones
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Simon A Jones
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Francesca Wanda Rossi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), World Allergy Organization (WAO) Center of Excellence, University of Naples Federico II, Naples, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), World Allergy Organization (WAO) Center of Excellence, University of Naples Federico II, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), World Allergy Organization (WAO) Center of Excellence, University of Naples Federico II, Naples, Italy.,Institute of Experimental Endocrinology and Oncology 'Gateano Salvatore' (IEOS), National Research Council (CNR), Naples, Italy
| | - Mohey Eldin M El Shikh
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Frances Humby
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
77
|
Rice LM, Mantero JC, Stratton EA, Warburton R, Roberts K, Hill N, Simms RW, Domsic R, Farber HW, Layfatis R. Serum biomarker for diagnostic evaluation of pulmonary arterial hypertension in systemic sclerosis. Arthritis Res Ther 2018; 20:185. [PMID: 30115106 PMCID: PMC6097341 DOI: 10.1186/s13075-018-1679-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023] Open
Abstract
Background Systemic sclerosis-associated pulmonary arterial hypertension (SSc-PAH) is one of the leading causes of death in SSc. Identification of a serum-based proteomic diagnostic biomarker for SSc-PAH would allow for rapid non-invasive screening and could positively impact patient survival. Identification and validation of novel proteins could potentially facilitate the identification of SSc-PAH, and might also point to important protein mediators in pathogenesis. Methods Thirteen treatment-naïve SSc-PAH patients had serum collected at time of diagnosis and were used as the discovery cohort for the protein-expression biomarker. Two proteins, Midkine and Follistatin-like 3 (FSTL3) were then validated by enzyme-linked immunosorbent assays. Midkine and FSTL3 were tested in combination to identify SSc-PAH and were validated in two independent cohorts of SSc-PAH (n = 23, n = 11). Results Eighty-two proteins were found to be differentially regulated in SSc-PAH sera. Two proteins (Midkine and FSTL3) were also shown to be elevated in publicly available data and their expression was evaluated in independent cohorts. In the validation cohorts, the combination of Midkine and FSTL3 had an area under the receiver operating characteristic curve (AUC) of 0.85 and 0.92 with respective corresponding measures of sensitivity of 76% and 91%, and specificity measures of 76% and 80%. Conclusions These findings indicate that there is a clear delineation between overall protein expression in sera from SSc patients and those with SSc-PAH. The combination of Midkine and FSTL3 can serve as an SSc-PAH biomarker and are potential drug targets for this rare disease population. Electronic supplementary material The online version of this article (10.1186/s13075-018-1679-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lisa M Rice
- Boston University School of Medicine, E5 Arthritis Center, 72 E Concord Street, Boston, MA, 0211, USA.
| | - Julio C Mantero
- Boston University School of Medicine, E5 Arthritis Center, 72 E Concord Street, Boston, MA, 0211, USA
| | - Eric A Stratton
- Boston University School of Medicine, E5 Arthritis Center, 72 E Concord Street, Boston, MA, 0211, USA
| | | | | | | | - Robert W Simms
- Boston University School of Medicine, E5 Arthritis Center, 72 E Concord Street, Boston, MA, 0211, USA
| | - Robyn Domsic
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Harrison W Farber
- Boston University School of Medicine, E5 Arthritis Center, 72 E Concord Street, Boston, MA, 0211, USA
| | - Robert Layfatis
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
78
|
Kilarski WW. Physiological Perspective on Therapies of Lymphatic Vessels. Adv Wound Care (New Rochelle) 2018; 7:189-208. [PMID: 29984111 PMCID: PMC6032671 DOI: 10.1089/wound.2017.0768] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/26/2018] [Indexed: 12/16/2022] Open
Abstract
Significance: Growth of distinctive blood vessels of granulation tissue is a central step in the post-developmental tissue remodeling. Even though lymphangiogenesis is a part of the regeneration process, the significance of the controlled restoration of lymphatic vessels has only recently been recognized. Recent Advances: Identification of lymphatic markers and growth factors paved the way for the exploration of the roles of lymphatic vessels in health and disease. Emerging pro-lymphangiogenic therapies use vascular endothelial growth factor (VEGF)-C to combat fluid retention disorders such as lymphedema and to enhance the local healing process. Critical Issues: The relevance of recently identified lymphatic functions awaits verification by their association with pathologic conditions. Further, despite a century of research, the complete etiology of secondary lymphedema, a fluid retention disorder directly linked to the lymphatic function, is not understood. Finally, the specificity of pro-lymphangiogenic therapy depends on VEGF-C transfection efficiency, dose exposure, and the age of the subject, factors that are difficult to standardize in a heterogeneous human population. Future Directions: Further research should reveal the role of lymphatic circulation in internal organs and connect its impairment with human diseases. Pro-lymphangiogenic therapies that aim at the acceleration of tissue healing should focus on the controlled administration of VEGF-C to increase their capillary specificity, whereas regeneration of collecting vessels might benefit from balanced maturation and differentiation of pre-existing lymphatics. Unique features of pre-nodal lymphatics, fault tolerance and functional hyperplasia of capillaries, may find applications outreaching traditional pro-lymphangiogenic therapies, such as immunomodulation or enhancement of subcutaneous grafting.
Collapse
Affiliation(s)
- Witold W. Kilarski
- Institute for Molecular Engineering, The University of Chicago, Chicago, Illinois
| |
Collapse
|
79
|
Hadamitzky C, Perić H, Theobald SJ, Gratz KF, Spohr H, Pabst R, Vogt PM. Effect of cryopreservation on lymph node fragment regeneration after autologous transplantation in the minipig model. Innov Surg Sci 2018; 3:139-146. [PMID: 31579777 PMCID: PMC6604575 DOI: 10.1515/iss-2018-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/10/2018] [Indexed: 02/02/2023] Open
Abstract
Introduction Lymphoedema is a worldwide pandemic causing swelling of tissues due to dysfunctional transport of lymph fluid. Present management concepts are based in conservative palliation of symptoms through manual lymphatic drainage, use of compression garments, manual lymph drainage, exercise, and skin care. Nevertheless, some curative options as autologous lymph node transplantation were shown to reduce lymphoedema in selected cases. Lately, some concern has arisen due to reports of donor site morbidity. A possible solution could be the development of artificial lymph node scaffolds as niches of lymphatic regeneration. Engineering these scaffolds has included cryopreservation of lymph node stroma. However, the effects of cryopreservation on the regeneration capacities of these organs were unknown. Materials and methods Here, we used the minipig animal model to assess lymphatic regeneration processes after cryopreservation of autologous lymph nodes. Superficial inguinal lymph nodes were excised and conserved at -80°C for 1 month. Thereafter, lymph node fragments were transplanted in the subcutaneous tissue. Results Regeneration of the lymph nodes was assessed five months after transplantation. We show that lymph node fragment regeneration takes place in spite of former cryopreservation. Transplanted fragments presented typical histological appearance. Their draining capacity was documented by macroscopic transport of Berlin Blue dye as well as through SPECT-CT hybrid imaging. Discussion In conclusion, our results suggest that processes of cryopreservation can be used in the creation of artificial lymph node scaffolds without major impairment of lymph node fragments regeneration.
Collapse
Affiliation(s)
- Catarina Hadamitzky
- Practice for Lympho-Vascular Diseases Bahnhofstrasse 12, Hannover, Germany.,Catarina Hadamitzky and Hanes Perić contributed equally to the manuscript
| | - Hanes Perić
- myDent GmbH Tiergartenstrasse 114, 30559 Hannover, Germany.,Catarina Hadamitzky and Hanes Perić contributed equally to the manuscript
| | - Sebastian J Theobald
- Clinic of Haematology, Haemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | | | - Hendrik Spohr
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Reinhard Pabst
- Institute of Immunomorphology, Hannover Medical School, Hannover, Germany
| | - Peter M Vogt
- Clinic of Plastic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
80
|
Maehara T, Mattoo H, Mahajan VS, Murphy SJ, Yuen GJ, Ishiguro N, Ohta M, Moriyama M, Saeki T, Yamamoto H, Yamauchi M, Daccache J, Kiyoshima T, Nakamura S, Stone JH, Pillai S. The expansion in lymphoid organs of IL-4 + BATF + T follicular helper cells is linked to IgG4 class switching in vivo. Life Sci Alliance 2018; 1. [PMID: 29984361 PMCID: PMC6034714 DOI: 10.26508/lsa.201800050] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Distinct T follicular helper (TFH) subsets that influence specific class-switching events are assumed to exist, but the accumulation of isotype-specific TFH subsets in secondary lymphoid organs (SLOs) and tertiary lymphoid organs has not been hitherto demonstrated. IL-4-expressing TFH cells are surprisingly sparse in human SLOs. In contrast, in IgG4-related disease (IgG4-RD), a disorder characterized by polarized Ig class switching, most TFH cells in tertiary and SLOs make IL-4. Human IL-4+ TFH cells do not express GATA-3 but express nuclear BATF, and the transcriptomes of IL-4-secreting TFH cells differ from both PD1hi TFH cells that do not secrete IL-4 and IL-4-secreting non-TFH cells. Unlike IgG4-RD, IL-4+ TFH cells are rarely found in tertiary lymphoid organs in Sjögren's syndrome, a disorder in which IgG4 is not elevated. The proportion of CD4+IL-4+BATF+ T cells and CD4+IL-4+CXCR5+ T cells in IgG4-RD tissues correlates tightly with tissue IgG4 plasma cell numbers and plasma IgG4 levels in patients but not with the total plasma levels of other isotypes. These data describe a disease-related TFH subpopulation in human tertiary lymphoid organs and SLOs that is linked to IgG4 class switching.
Collapse
Affiliation(s)
- Takashi Maehara
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hamid Mattoo
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vinay S Mahajan
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Samuel Jh Murphy
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Grace J Yuen
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Noriko Ishiguro
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Miho Ohta
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Masafumi Moriyama
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takako Saeki
- Department of Internal Medicine, Nagaoka Red Cross Hospital, Nagaoka, Japan
| | - Hidetaka Yamamoto
- Division of Diagnostic Pathology, Kyushu University Hospital, Fukuoka, Japan.,Department of Anatomic Pathology, Kyushu University, Fukuoka, Japan
| | - Masaki Yamauchi
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Joe Daccache
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tamotsu Kiyoshima
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Seiji Nakamura
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - John H Stone
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shiv Pillai
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
81
|
Xiao X, Putatunda R, Zhang Y, Soni PV, Li F, Zhang T, Xin M, Luo JJ, Bethea JR, Cheng Y, Hu W. Lymphotoxin β receptor-mediated NFκB signaling promotes glial lineage differentiation and inhibits neuronal lineage differentiation in mouse brain neural stem/progenitor cells. J Neuroinflammation 2018; 15:49. [PMID: 29463313 PMCID: PMC5819232 DOI: 10.1186/s12974-018-1074-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 01/22/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Lymphotoxin (LT) is a lymphokine mainly expressed in lymphocytes. LTα binds one or two membrane-associated LTβ to form LTα2β1 or LTα1β2 heterotrimers. The predominant LTα1β2 binds to LTβ receptor (LTβR) primarily expressed in epithelial and stromal cells. Most studies on LTβR signaling have focused on the organization, development, and maintenance of lymphoid tissues. However, the roles of LTβR signaling in the nervous system, particularly in neurogenesis, remain unknown. Here, we investigated the role of LTβR-mediated NFκB signaling in regulating neural lineage differentiation. METHODS The C57BL/6J wild-type and GFAP-dnIκBα transgenic mice were used. Serum-free embryoid bodies were cultured from mouse embryonic stem cells and further induced into neural stem/progenitor cells (NSCs/NPCs). Primary neurospheres were cultured from embryonic and adult mouse brains followed by monolayer culture for amplification/passage. NFκB activation was determined by adenovirus-mediated NFκB-firefly-luciferase reporter assay and p65/RelB/p52 nuclear translocation assay. LTβR mRNA expression was evaluated by quantitative RT-PCR and LTβR protein expression was determined by immunohistochemistry and Western blot analysis. Multilabeled immunocytochemistry or immunohistochemistry followed by fluorescent confocal microscopy and quantitative analysis of neural lineage differentiation were performed. Graphing and statistical analysis were performed with GraphPad Prism software. RESULTS In cultured NSCs/NPCs, LTα1β2 stimulation induced an activation of classical and non-classical NFκB signaling. The expression of LTβR-like immunoreactivity in GFAP+/Sox2+ NSCs was identified in well-established neurogenic zones of adult mouse brain. Quantitative RT-PCR and Western blot analysis validated the expression of LTβR in cultured NSCs/NPCs and brain neurogenic regions. LTβR expression was significantly increased during neural induction. LTα1β2 stimulation in cultured NSCs/NPCs promoted astroglial and oligodendrocytic lineage differentiation, but inhibited neuronal lineage differentiation. Astroglial NFκB inactivation in GFAP-dnIκBα transgenic mice rescued LTβR-mediated abnormal phenotypes of cultured NSCs/NPCs. CONCLUSION This study provides the first evidence for the expression and function of LTβR signaling in NSCs/NPCs. Activation of LTβR signaling promotes glial lineage differentiation. Our results suggest that neurogenesis is regulated by the adaptive immunity and inflammatory responses.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Raj Putatunda
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Yonggang Zhang
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Priya V Soni
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Fang Li
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Ting Zhang
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Mingyang Xin
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Jin Jun Luo
- Department of Neurology, Temple University Lewis Katz School of Medicine, 3401 N Broad Street, Philadelphia, PA, USA
| | - John R Bethea
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Wenhui Hu
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
82
|
Kraft JC, Treuting PM, Ho RJY. Indocyanine green nanoparticles undergo selective lymphatic uptake, distribution and retention and enable detailed mapping of lymph vessels, nodes and abnormalities. J Drug Target 2018; 26:494-504. [PMID: 29388438 DOI: 10.1080/1061186x.2018.1433681] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The distributed network of lymph vessels and nodes in the body, with its complex architecture and physiology, presents a major challenge for whole-body lymphatic-targeted drug delivery. To gather physiological and pathological information of the lymphatics, near-infrared (NIR) fluorescence imaging of NIR fluorophores is used in clinical practice due to its tissue-penetrating optical radiation (700-900 nm) that safely provides real-time high-resolution in vivo images. However, indocyanine green (ICG), a common clinical NIR fluorophore, is unstable in aqueous environments and under light exposure, and its poor lymphatic distribution and retention limits its use as a NIR lymphatic tracer. To address this, we investigated in mice the distribution pathways of a novel nanoparticle formulation that stabilises ICG and is optimised for lymphatic drug delivery. From the subcutaneous space, ICG particles provided selective lymphatic uptake, lymph vessel and node retention, and extensive first-pass lymphatic distribution of ICG, enabling 0.2 mm and 5-10 cell resolution of lymph vessels, and high signal-to-background ratios for lymphatic vessel and node networks. Soluble (free) ICG readily dissipated from lymph vessels local to the injection site and absorbed into the blood. These unique characteristics of ICG particles could enable mechanistic studies of the lymphatics and diagnosis of lymphatic abnormalities.
Collapse
Affiliation(s)
- John C Kraft
- a Department of Pharmaceutics , University of Washington , Seattle , WA , USA
| | - Piper M Treuting
- b Department of Comparative Medicine , University of Washington , Seattle , WA , USA
| | - Rodney J Y Ho
- a Department of Pharmaceutics , University of Washington , Seattle , WA , USA.,c Department of Bioengineering , University of Washington , Seattle , WA , USA
| |
Collapse
|
83
|
Ren X, Tian Y, Liu L, Liu X. A reaction–diffusion within-host HIV model with cell-to-cell transmission. J Math Biol 2018; 76:1831-1872. [DOI: 10.1007/s00285-017-1202-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/26/2017] [Indexed: 02/07/2023]
|
84
|
Abstract
For decades, the brain has been considered an immune-privileged organ, meaning that the brain was mainly ignored by the immune system and that the presence of immune cells, notably of the adaptive arm, was a hallmark of pathological conditions. Over the past few decades, the definition of the immune privilege continues to be refined. There has been evidence accumulating that shows that the immune system plays a role in proper brain function. This evidence may represent an effective source of therapeutic targets for neurological disorders. In this chapter, we discuss the recent advances in understanding the immunity of the brain and describe how tertiary lymphoid structures can be generated in the central nervous system, which might represent a new avenue to treat neurological disorders.
Collapse
|
85
|
Mueller CG, Nayar S, Gardner D, Barone F. Cellular and Vascular Components of Tertiary Lymphoid Structures. Methods Mol Biol 2018; 1845:17-30. [PMID: 30141005 DOI: 10.1007/978-1-4939-8709-2_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Inflammatory immune cells recruited at the site of chronic inflammation form structures that resemble secondary lymphoid organs (SLO). These are characterized by segregated areas of prevalent T- or B-cell aggregation, differentiation of high endothelial venules, and local activation of resident stromal cells, including lymphatic endothelial cells. B-cell proliferation and affinity maturation toward locally displayed autoantigens have been demonstrated at these sites, known as tertiary lymphoid structures (TLS). TLS formation during chronic inflammation has been associated with local disease persistence and progression, as well as increased systemic manifestations. While bearing a similar histological structure to SLO, the signals that regulate TLS and SLO formation can diverge and a series of pro-inflammatory cytokines have been ascribed as responsible for TLS formation at different anatomical sites. Moreover, for a long time the structural compartment that regulates TLS homeostasis, including survival and recirculation of leucocytes has been neglected. In this chapter, we summarize the novel data available on TLS formation, structural organization, and the functional and anatomical links connecting TLS and SLOs.
Collapse
Affiliation(s)
- Christopher George Mueller
- Laboratoire d'Immunologie, Immunopathologie et Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire (IBMC), CNRS UPR 3572, University of Strasbourg, Strasbourg, France
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - David Gardner
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK.
| |
Collapse
|
86
|
Yang JG, Sun YF, He KF, Ren JG, Liu ZJ, Liu B, Zhang W, Zhao YF. Lymphotoxins Promote the Progression of Human Lymphatic Malformation by Enhancing Lymphatic Endothelial Cell Proliferation. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2602-2615. [DOI: 10.1016/j.ajpath.2017.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/24/2017] [Accepted: 07/26/2017] [Indexed: 12/31/2022]
|
87
|
Kogame T, Nomura T, Kataoka T, Hirata M, Ueshima C, Matsui M, Kabashima K. Possible inducible skin-associated lymphoid tissue (iSALT)-like structures with CXCL13+
fibroblast-like cells in secondary syphilis. Br J Dermatol 2017; 177:1737-1739. [DOI: 10.1111/bjd.15349] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- T. Kogame
- Department of Dermatology; Kyoto University Graduate School of Medicine; 54 Shogoin Kawara-cho Sakyo Kyoto 606-8507 Japan
| | - T. Nomura
- Department of Dermatology; Kyoto University Graduate School of Medicine; 54 Shogoin Kawara-cho Sakyo Kyoto 606-8507 Japan
| | - T. Kataoka
- Department of Diagnostic Pathology; Kyoto University Hospital; Kyoto Japan
| | - M. Hirata
- Department of Diagnostic Pathology; Kyoto University Hospital; Kyoto Japan
| | - C. Ueshima
- Department of Diagnostic Pathology; Kyoto University Hospital; Kyoto Japan
| | - M. Matsui
- Ijinkai Takeda General Hospital; Kyoto Japan
| | - K. Kabashima
- Department of Dermatology; Kyoto University Graduate School of Medicine; 54 Shogoin Kawara-cho Sakyo Kyoto 606-8507 Japan
- Singapore Immunology Network (SIgN) and Institute of Medical Biology; Agency for Science, Technology and Research (A*STAR); Biopolis Singapore
- PRESTO; Japan Science and Technology Agency; Saitama Japan
| |
Collapse
|
88
|
S1P Provokes Tumor Lymphangiogenesis via Macrophage-Derived Mediators Such as IL-1 β or Lipocalin-2. Mediators Inflamm 2017; 2017:7510496. [PMID: 28804221 PMCID: PMC5539930 DOI: 10.1155/2017/7510496] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/15/2017] [Indexed: 12/17/2022] Open
Abstract
A pleiotropic signaling lipid, sphingosine-1-phosphate (S1P), has been implicated in various pathophysiological processes supporting tumor growth and metastasis. However, there are only a few descriptive studies suggesting a role of S1P in tumor lymphangiogenesis, which is critical for tumor growth and dissemination. Corroborating own data, the literature suggests that apoptotic tumor cell-derived S1P alters the phenotype of tumor-associated macrophages (TAMs) to gain protumor functions. However, mechanistically, the role of TAM-induced lymphangiogenesis has only been poorly described, mostly linked to the production of lymphangiogenic factors such as vascular endothelial growth factor C (VEGF-C) and VEGF-D, or transdifferentiation into lymphatic endothelial cells. Recent findings highlight a rather underappreciated role of S1P in tumor lymphangiogenesis, referring to the production of interleukin-1β (IL-1β) and lipocalin-2 (LCN2) by a tumor-promoting macrophage phenotype. In this review, we aim to provide to the readers with the current understanding of the molecular mechanism how apoptotic cell-derived S1P triggers TAMs to promote lymphangiogenesis.
Collapse
|
89
|
Lymphatic Endothelial Cells Control Initiation of Lymph Node Organogenesis. Immunity 2017; 47:80-92.e4. [DOI: 10.1016/j.immuni.2017.05.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/14/2017] [Accepted: 05/26/2017] [Indexed: 10/19/2022]
|
90
|
Okada S, Kobayashi-Fujiwara Y, Oga A, Furuta T, Ikemoto K, Fujii H, Sakata Y, Suzuki Y, Hasegawa S, Kusuda T, Itoh H, Yamashita H, Ohga S. Distinct Distribution of Immunocytes in a Retropharyngeal Lymphadenopathy Associated with Kawasaki Disease: A Case Study Compared with Tonsillitis. Cardiology 2017; 137:237-243. [PMID: 28467980 DOI: 10.1159/000467388] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 02/27/2017] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Retropharyngeal lesions are often associated with Kawasaki disease (KD). A 4-year-old male first presented a peritonsillar and retropharyngeal abscess-like lesion. Surgical tonsillectomy was performed to avoid a risk of mediastinal abscess, but he fulfilled the diagnostic criteria of KD after the operation. This prompted us to perform a histological study on the KD tonsils. METHODS The histopathology of the KD tonsil specimens were compared with hypertrophic tonsils obtained from 4 patients with chronic tonsillitis unrelated to KD assessed by the immunostainings. RESULTS KD tonsils showed small lymphatic follicles and neutrophil infiltration in the peritonsillar muscle layer, with no evidence of vasculitis or abscess formation. The KD tonsils exclusively showed (1) predominant activated CD4+ T cells in the perifollicular interstitium, (2) sparse scattering of CD68+ monocytes/macrophages in the lymphatic follicles, and (3) polyclonal carcinoembryonic antigen-positive cells in the lymphatic follicles and venules with the high endothelial cells. CONCLUSIONS The uniquely distributed immunocytes suggest the inflammatory process of KD involving the pathogen-associated molecules.
Collapse
Affiliation(s)
- Seigo Okada
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Weinstein AM, Chen L, Brzana EA, Patil PR, Taylor JL, Fabian KL, Wallace CT, Jones SD, Watkins SC, Lu B, Stroncek DF, Denning TL, Fu YX, Cohen PA, Storkus WJ. Tbet and IL-36γ cooperate in therapeutic DC-mediated promotion of ectopic lymphoid organogenesis in the tumor microenvironment. Oncoimmunology 2017; 6:e1322238. [PMID: 28680760 DOI: 10.1080/2162402x.2017.1322238] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/31/2022] Open
Abstract
We have previously reported that direct injection of dendritic cells (DC) engineered to express the Type-1 transactivator Tbet (i.e., DC.Tbet) into murine tumors results in antitumor efficacy in association with the development of structures resembling tertiary lymphoid organs (TLO) in the tumor microenvironment (TME). These TLO contained robust infiltrates of B cells, DC, NK cells, and T cells in proximity to PNAd+ blood vessels; however, they were considered incomplete, since the recruited B cells failed to organize into classic germinal center-like structures. We now report that antitumor efficacy and TLO-inducing capacity of DC.Tbet-based i.t. therapy is operational in peripheral lymph node-deficient LTA-/- mice, and that it is highly dependent upon a direct Tbet target gene product, IL-36γ/IL-1F9. Intratumoral DC.Tbet fails to provide protection to tumor-bearing IL-36R-/- hosts, or to tumor-bearing wild-type recipient mice co-administered rmIL-1F5/IL-36RN, a natural IL-36R antagonist. Remarkably, the injection of tumors with DC engineered to secrete a bioactive form of mIL-36γ (DC.IL36γ) also initiated therapeutic TLO and slowed tumor progression in vivo. Furthermore, DC.IL36γ cells strongly upregulated their expression of Tbet, suggesting that Tbet and IL-36γ cooperate to reinforce each other's expression in DC, rendering them competent to promote TLO formation in an "immunologically normalized," therapeutic TME.
Collapse
Affiliation(s)
- Aliyah M Weinstein
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | - Lu Chen
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | | | | | | | - Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | - Callen T Wallace
- Department of Cell Biology and Physiology, UPSOM, Pittsburgh, PA, USA
| | | | - Simon C Watkins
- Department of Cell Biology and Physiology, UPSOM, Pittsburgh, PA, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | - David F Stroncek
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, PA, USA
| | - Timothy L Denning
- Center for Inflammation, Immunity & Infection at Georgia State University, Atlanta, GA, USA
| | - Yang-Xin Fu
- Departments of Pathology and Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Peter A Cohen
- Department of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA.,Department of Dermatology, UPSOM, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Bioengineering, UPSOM, Pittsburgh, PA, USA.,University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
92
|
Popovic ZV, Embgenbroich M, Chessa F, Nordström V, Bonrouhi M, Hielscher T, Gretz N, Wang S, Mathow D, Quast T, Schloetel JG, Kolanus W, Burgdorf S, Gröne HJ. Hyperosmolarity impedes the cross-priming competence of dendritic cells in a TRIF-dependent manner. Sci Rep 2017; 7:311. [PMID: 28331179 PMCID: PMC5428499 DOI: 10.1038/s41598-017-00434-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 02/21/2017] [Indexed: 12/19/2022] Open
Abstract
Tissue osmolarity varies among different organs and can be considerably increased under pathologic conditions. Hyperosmolarity has been associated with altered stimulatory properties of immune cells, especially macrophages and dendritic cells. We have recently reported that dendritic cells upon exposure to hypertonic stimuli shift their profile towards a macrophage-M2-like phenotype, resulting in attenuated local alloreactivity during acute kidney graft rejection. Here, we examined how hyperosmotic microenvironment affects the cross-priming capacity of dendritic cells. Using ovalbumin as model antigen, we showed that exposure of dendritic cells to hyperosmolarity strongly inhibits activation of antigen-specific T cells despite enhancement of antigen uptake, processing and presentation. We identified TRIF as key mediator of this phenomenon. Moreover, we detected a hyperosmolarity-triggered, TRIF-dependent clustering of MHCI loaded with the ovalbumin-derived epitope, but not of overall MHCI molecules, providing a possible explanation for a reduced T cell activation. Our findings identify dendritic cells as important players in hyperosmolarity-mediated immune imbalance and provide evidence for a novel pathway of inhibition of antigen specific CD8+ T cell response in a hypertonic micromilieu.
Collapse
Affiliation(s)
- Zoran V Popovic
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany. .,Institute of Pathology, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany.
| | - Maria Embgenbroich
- Department of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Federica Chessa
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Viola Nordström
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Mahnaz Bonrouhi
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Thomas Hielscher
- Department of Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Norbert Gretz
- Medical Research Center, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Shijun Wang
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Daniel Mathow
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Thomas Quast
- Department of Molecular Immunology and Cell Biology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Jan-Gero Schloetel
- Department of Membrane Biochemistry, LIMES Institute, University of Bonn, Bonn, Germany
| | - Waldemar Kolanus
- Department of Molecular Immunology and Cell Biology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Sven Burgdorf
- Department of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Hermann-Josef Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany. h.-
| |
Collapse
|
93
|
Riquet M, Pricopi C, Legras A, Arame A, Badia A, Le Pimpec Barthes F. Can mathematics replace anatomy to establish recommendations in lung cancer surgery? J Thorac Dis 2017; 9:E327-E332. [PMID: 28449533 DOI: 10.21037/jtd.2017.03.46] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The greater the number of lymph node (LN) sampled (NLNsS) during lung cancer surgery, the lower the risk of underestimating the pN-status and the better the outcome of the pN0-patients due to stage-migration. Thus, regarding LN sampling "to be or not to be", number is the question. Recent studies advocate removing 10 LNs. The most suitable NLNsS is unfortunately impossible to establish by mathematics. A too high NLNsS variability exists, based on anatomy, surgery and pathology. The methodology may vary according to Inter-institutional differences in the surgical approach regarding LN inspection and number sampling. The NLNsS increases with the type of resection: sublobar, lobectomy or pneumonectomy. Concerning pathology, one LN may be divided into several pieces, leading to number overestimation. The pathological examination is limited by the number of slices analyzed by LN. The examined LNs can arbitrarily depend on the probability of detecting nodal metastasis. In fact, the only way to ensure the best NLNsS and the best pN-staging is to remove all LNs from the ipsilateral mediastinal and hilar LN-stations as they are discovered by thoroughly dissecting their anatomical locations. In doing so, a deliberate lack of harvest of LNs is unlikely, number turns out not to be the question anymore and a low NLNsS no longer means incomplete surgery. This prevents from judging as incomplete a complete LN dissection in a patient with a small NLNsS and from considering as complete a true incomplete one in a patient with a great NLNsS. Precise information describing the course of the operation and furnished in the surgeon's reports is also advisable to further improve the quality of LN-dissection, which ultimately might be beneficial in the long-term to patients. However, that procedure is of limited interest in pN-staging if LNs are not thoroughly examined and also described by the pathologist.
Collapse
Affiliation(s)
- Marc Riquet
- General Thoracic Surgery Department, Georges Pompidou European Hospital, Paris, France
| | - Ciprian Pricopi
- General Thoracic Surgery Department, Georges Pompidou European Hospital, Paris, France
| | - Antoine Legras
- General Thoracic Surgery Department, Georges Pompidou European Hospital, Paris, France
| | - Alex Arame
- General Thoracic Surgery Department, Georges Pompidou European Hospital, Paris, France
| | - Alain Badia
- General Thoracic Surgery Department, Georges Pompidou European Hospital, Paris, France
| | | |
Collapse
|
94
|
Crescenzi R, Donahue PMC, Hartley KG, Desai AA, Scott AO, Braxton V, Mahany H, Lants SK, Donahue MJ. Lymphedema evaluation using noninvasive 3T MR lymphangiography. J Magn Reson Imaging 2017; 46:1349-1360. [PMID: 28245075 DOI: 10.1002/jmri.25670] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/30/2017] [Indexed: 01/05/2023] Open
Abstract
PURPOSE To exploit the long 3.0T relaxation times and low flow velocity of lymphatic fluid to develop a noninvasive 3.0T lymphangiography sequence and evaluate its relevance in patients with lymphedema. MATERIALS AND METHODS A 3.0T turbo-spin-echo (TSE) pulse train with long echo time (TEeffective = 600 msec; shot-duration = 13.2 msec) and TSE-factor (TSE-factor = 90) was developed and signal evolution simulated. The method was evaluated in healthy adults (n = 11) and patients with unilateral breast cancer treatment-related lymphedema (BCRL; n = 25), with a subgroup (n = 5) of BCRL participants scanned before and after manual lymphatic drainage (MLD) therapy. Maximal lymphatic vessel cross-sectional area, signal-to-noise-ratio (SNR), and results from a five-point categorical scoring system were recorded. Nonparametric tests were applied to evaluate study parameter differences between controls and patients, as well as between affected and contralateral sides in patients (significance criteria: two-sided P < 0.05). RESULTS Patient volunteers demonstrated larger lymphatic cross-sectional areas in the affected (arm = 12.9 ± 6.3 mm2 ; torso = 17.2 ± 15.6 mm2 ) vs. contralateral (arm = 9.4 ± 3.9 mm2 ; torso = 9.1 ± 4.6 mm2 ) side; this difference was significant both for the arm (P = 0.014) and torso (P = 0.025). Affected (arm: P = 0.010; torso: P = 0.016) but not contralateral (arm: P = 0.42; torso: P = 0.71) vessel areas were significantly elevated compared with control values. Lymphatic cross-sectional areas reduced following MLD on the affected side (pre-MLD: arm = 8.8 ± 1.8 mm2 ; torso = 31.4 ± 26.0 mm2 ; post-MLD: arm = 6.6 ± 1.8 mm2 ; torso = 23.1 ± 24.3 mm2 ). This change was significant in the torso (P = 0.036). The categorical scoring was found to be less specific for detecting lateralizing disease compared to lymphatic-vessel areas. CONCLUSION A 3.0T lymphangiography sequence is proposed, which allows for upper extremity lymph stasis to be detected in ∼10 minutes without exogenous contrast agents. LEVEL OF EVIDENCE 1 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2017;46:1349-1360.
Collapse
Affiliation(s)
- Rachelle Crescenzi
- Radiology and Radiological Science, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Paula M C Donahue
- Physical Medicine and Rehabilitation, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Dayani Center for Health and Wellness, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Katherine G Hartley
- Radiology and Radiological Science, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Aditi A Desai
- Radiology and Radiological Science, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Allison O Scott
- Radiology and Radiological Science, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Vaughn Braxton
- Radiology and Radiological Science, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Helen Mahany
- Radiology and Radiological Science, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Sarah K Lants
- Radiology and Radiological Science, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Manus J Donahue
- Radiology and Radiological Science, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Neurology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
95
|
Review on Toll-Like Receptor Activation in Myasthenia Gravis: Application to the Development of New Experimental Models. Clin Rev Allergy Immunol 2017; 52:133-147. [PMID: 27207173 DOI: 10.1007/s12016-016-8549-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Abnormal toll-like receptor (TLR) activation and uncontrolled resolution of inflammation are suspected to play a key role in the development of autoimmune diseases. Acquired myasthenia gravis (MG) is an invalidating neuromuscular disease leading to muscle weaknesses. MG is mainly mediated by anti-acetylcholine receptor (AChR) autoantibodies, and thymic hyperplasia characterized by ectopic germinal centers is a common feature in MG. An abnormal expression of certain TLRs is observed in the thymus of MG patients associated with the overexpression of interferon (IFN)-β, the orchestrator of thymic changes in MG. Experimental models have been developed for numerous autoimmune diseases. These models are induced by animal immunization with a purified antigen solubilized in complete Freund's adjuvant (CFA) containing heat-inactivated mycobacterium tuberculosis (MTB). Sensitization against the antigen is mainly due to the activation of TLR signaling pathways by the pathogen motifs displayed by MTB, and attempts have been made to substitute the use of CFA by TLR agonists. AChR emulsified in CFA is used to induce the classical experimental autoimmune MG model (EAMG). However, the TLR4 activator lipopolysaccharide (LPS) has proved to be efficient to replace MTB and induce a sensitization against purified AChR. Poly(I:C), the well-known TLR3 agonist, is also able by itself to induce MG symptoms in mice associated with early thymic changes as observed in human MG. In this review, we discuss the abnormal expression of TLRs in MG patients and we describe the use of TLR agonists to induce EAMG in comparison with other autoimmune experimental models.
Collapse
|
96
|
Innate lymphoid cells in autoimmunity: emerging regulators in rheumatic diseases. Nat Rev Rheumatol 2017; 13:164-173. [PMID: 28148916 DOI: 10.1038/nrrheum.2016.218] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Innate lymphoid cells (ILCs) are important in the regulation of barrier homeostasis. These cells do not express T cell receptors but share many functional similarities with T helper cells and cytotoxic CD8+ T lymphocytes. ILCs are divided into three groups, namely group 1 ILCs, group 2 ILCs and group 3 ILCs, based on the transcription factors they depend on for their development and function, and the cytokines they produce. Emerging data indicate that ILCs not only have protective functions but can also have detrimental effects when dysregulated, leading to chronic inflammation and autoimmune diseases, including asthma, inflammatory bowel disease, graft-versus-host disease, psoriasis, rheumatoid arthritis and atopic dermatitis. Elucidation of the cytokine pathways involved in various autoimmune diseases - and the identification of ILCs as potent producers of these cytokines - points towards a potential role for these cellular players in the pathophysiology of these diseases. In this Review we discuss the current knowledge of the role of ILCs in the pathogenesis of rheumatic and other autoimmune diseases.
Collapse
|
97
|
Ji RC. Lymph Nodes and Cancer Metastasis: New Perspectives on the Role of Intranodal Lymphatic Sinuses. Int J Mol Sci 2016; 18:ijms18010051. [PMID: 28036019 PMCID: PMC5297686 DOI: 10.3390/ijms18010051] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/21/2016] [Accepted: 12/23/2016] [Indexed: 02/07/2023] Open
Abstract
The lymphatic system is essential for transporting interstitial fluid, soluble antigen, and immune cells from peripheral tissues to lymph nodes (LNs). Functional integrity of LNs is dependent on intact lymphatics and effective lymph drainage. Molecular mechanisms that facilitate interactions between tumor cells and lymphatic endothelial cells (LECs) during tumor progression still remain to be identified. The cellular and molecular structures of LNs are optimized to trigger a rapid and efficient immune response, and to participate in the process of tumor metastasis by stimulating lymphangiogenesis and establishing a premetastatic niche in LNs. Several molecules, e.g., S1P, CCR7-CCL19/CCL21, CXCL12/CXCR4, IL-7, IFN-γ, TGF-β, and integrin α4β1 play an important role in controlling the activity of LN stromal cells including LECs, fibroblastic reticular cells (FRCs) and follicular dendritic cells (DCs). The functional stromal cells are critical for reconstruction and remodeling of the LN that creates a unique microenvironment of tumor cells and LECs for cancer metastasis. LN metastasis is a major determinant for the prognosis of most human cancers and clinical management. Ongoing work to elucidate the function and molecular regulation of LN lymphatic sinuses will provide insight into cancer development mechanisms and improve therapeutic approaches for human malignancy.
Collapse
Affiliation(s)
- Rui-Cheng Ji
- Faculty of Welfare and Health Science, Oita University, Oita 870-1192, Japan.
| |
Collapse
|
98
|
Endomysial germinal centres in Hashimoto's thyroiditis with myopathic symptoms. Pathology 2016; 49:97-100. [PMID: 27913040 DOI: 10.1016/j.pathol.2016.08.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/27/2016] [Accepted: 08/31/2016] [Indexed: 11/22/2022]
|
99
|
Ruddle NH. High Endothelial Venules and Lymphatic Vessels in Tertiary Lymphoid Organs: Characteristics, Functions, and Regulation. Front Immunol 2016; 7:491. [PMID: 27881983 PMCID: PMC5101196 DOI: 10.3389/fimmu.2016.00491] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/25/2016] [Indexed: 12/27/2022] Open
Abstract
High endothelial venules (HEVs) and lymphatic vessels (LVs) are essential for the function of the immune system, by providing communication between the body and lymph nodes (LNs), specialized sites of antigen presentation and recognition. HEVs bring in naïve and central memory cells and LVs transport antigen, antigen-presenting cells, and lymphocytes in and out of LNs. Tertiary lymphoid organs (TLOs) are accumulations of lymphoid and stromal cells that arise and organize at ectopic sites in response to chronic inflammation in autoimmunity, microbial infection, graft rejection, and cancer. TLOs are distinguished from primary lymphoid organs – the thymus and bone marrow, and secondary lymphoid organs (SLOs) – the LNs, spleen, and Peyer’s patches, in that they arise in response to inflammatory signals, rather than in ontogeny. TLOs usually do not have a capsule but are rather contained within the confines of another organ. Their structure, cellular composition, chemokine expression, and vascular and stromal support resemble SLOs and are the defining aspects of TLOs. T and B cells, antigen-presenting cells, fibroblast reticular cells, and other stromal cells and vascular elements including HEVs and LVs are all typical components of TLOs. A key question is whether the HEVs and LVs play comparable roles and are regulated similarly to those in LNs. Data are presented that support this concept, especially with regard to TLO HEVs. Emerging data suggest that the functions and regulation of TLO LVs are also similar to those in LNs. These observations support the concept that TLOs are not merely cellular accumulations but are functional entities that provide sites to generate effector cells, and that their HEVs and LVs are crucial elements in those activities.
Collapse
Affiliation(s)
- Nancy H Ruddle
- Department of Epidemiology of Microbial Diseases, School of Public Health, Yale University School of Medicine , New Haven, CT , USA
| |
Collapse
|
100
|
Barone F, Gardner DH, Nayar S, Steinthal N, Buckley CD, Luther SA. Stromal Fibroblasts in Tertiary Lymphoid Structures: A Novel Target in Chronic Inflammation. Front Immunol 2016; 7:477. [PMID: 27877173 PMCID: PMC5100680 DOI: 10.3389/fimmu.2016.00477] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are organized aggregates of lymphocytes, myeloid, and stromal cells that provide ectopic hubs for acquired immune responses. TLS share phenotypical and functional features with secondary lymphoid organs (SLO); however, they require persistent inflammatory signals to arise and are often observed at target sites of autoimmune disease, chronic infection, cancer, and organ transplantation. Over the past 10 years, important progress has been made in our understanding of the role of stromal fibroblasts in SLO development, organization, and function. A complex and stereotyped series of events regulate fibroblast differentiation from embryonic life in SLOs to lymphoid organ architecture observed in adults. In contrast, TLS-associated fibroblasts differentiate from postnatal, locally activated mesenchyme, predominantly in settings of inflammation and persistent antigen presentation. Therefore, there are critical differences in the cellular and molecular requirements that regulate SLO versus TLS development that ultimately impact on stromal and hematopoietic cell function. These differences may contribute to the pathogenic nature of TLS in the context of chronic inflammation and malignant transformation and offer a window of opportunity for therapeutic interventions in TLS associated pathologies.
Collapse
Affiliation(s)
- Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - David H Gardner
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Nathalie Steinthal
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Christopher D Buckley
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Sanjiv A Luther
- Department of Biochemistry, Center for Immunity and Infection, University of Lausanne , Lausanne , Switzerland
| |
Collapse
|