1
|
Livingstone EJ, Cartwright JA, Campana L, Lewis PJS, Dwyer BJ, Aird R, Man TY, Vermeren M, Rossi AG, Boulter L, Forbes SJ. Semaphorin 7a is protective through immune modulation during acetaminophen-induced liver injury. J Inflamm (Lond) 2025; 22:13. [PMID: 40114253 PMCID: PMC11927371 DOI: 10.1186/s12950-025-00429-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/13/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND AND AIM Acetaminophen (APAP) induced acute liver injury (ALI), the leading cause acute liver failure in the western world, has limited treatment options. APAP toxicity results in massive hepatic necrosis and secondary infiltrating monocytes and neutrophils, which contribute to pathogenesis. Semaphorin 7a (Sema7a), a chemoattractant and modulator of monocytes and neutrophils, is a potential therapeutic target in other conditions, but its role in APAP-ALI is unexplored. METHODS Wild-type (WT) and Sema7a knockout (KO) mice were examined during APAP-ALI. Serum liver function tests, histological analysis and cellular localisation of Sema7a and its receptors, Plexin C1 and Integrin β1, were examined. Serum cytokines were quantified, tissue macrophages and neutrophils were localised, and in vivo phenotype, including phagocytosis, was assessed by immunohistochemistry and flow cytometry. RESULTS Sema7a was expressed by HNF4α + peri-necrotic hepatocytes circumferentially during APAP-ALI injury phases, and serum concentrations were increased, and correlated with hepatic injury. Sema7a KO mice had increased circulating inflammatory cytokines and significantly less hepatic F4/80 + macrophages, a cell type required for hepatic repair. Sema7a KO mice had higher necrotic area neutrophils, and increased neutrophil chemoattractant CXCL1. Without Sema7a expression, mice displayed increased necrosis and liver injury markers compared to Sema7a WT mice. Without peri-necrotic hepatocyte Sema7a expression, we also identified increased cell death and hepatic cellular stress outside of necrosis. CONCLUSION We have identified a novel protective role of Sema7a during injury phases of APAP-ALI. Without peri-necrotic hepatocyte Sema7a expression and secretion, there is increased inflammation, time specific worsened hepatic necrosis and increased hepatic cell stress and death outside of the necrotic zone.
Collapse
Affiliation(s)
- Eilidh J Livingstone
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Jennifer A Cartwright
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
- The Royal (Dick) School of Veterinary Studiesand Theaq , Roslin Institute, University of Edinburgh, Edinburgh, UK.
| | - Lara Campana
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Philip J Starkey Lewis
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Benjamin J Dwyer
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Rhona Aird
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Tak Yung Man
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Matthieu Vermeren
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Adriano Giorgio Rossi
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Stuart John Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
2
|
Chen YI, Tien SC, Ko YL, Chang CC, Hsu MF, Chien HJ, Peng HY, Jeng YM, Tien YW, Chang YT, Chang MC, Hu CM. SEMA7A-mediated juxtacrine stimulation of IGFBP-3 upregulates IL-17RB at pancreatic cancer invasive front. Cancer Gene Ther 2024; 31:1840-1855. [PMID: 39448803 PMCID: PMC11645274 DOI: 10.1038/s41417-024-00849-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
Tumor invasion is the hallmark of tumor malignancy. The invasive infiltration pattern of tumor cells located at the leading edge is highly correlated with metastasis and unfavorable patient outcomes. However, the regulatory mechanisms governing tumor malignancy at the invasive margin remain unclear. The IL-17B/IL-17RB pathway is known to promote pancreatic cancer invasion and metastasis, yet the specific mechanisms underlying IL-17RB upregulation during invasion are poorly understood. In this study, we unveiled a multistep process for IL-17RB upregulation at the invasive margin, which occurs through direct communication between tumor cells and fibroblasts. Tumor ATP1A1 facilitates plasma membrane expression of SEMA7A, which binds to and induces IGFBP-3 secretion from fibroblasts. The resulting gradient of IGFBP-3 influences the direction and enhances IL-17RB expression to regulate SNAI2 in invasion. These findings highlight the importance of local tumor-fibroblast interactions in promoting cancer cell invasiveness, potentially leading to the development of new therapeutic strategies targeting this communication.
Collapse
Affiliation(s)
- Yi-Ing Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Sui-Chih Tien
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-Ling Ko
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | - Min-Fen Hsu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hung Jen Chien
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hsuan-Yu Peng
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Ming Jeng
- Department of Pathology, National Taiwan University Hospital, Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yun-Wen Tien
- Department of Surgery, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Ting Chang
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
- National Taiwan University Hospital Hsin-Chu Branch, Hsinchu County, Taiwan
| | - Ming-Chu Chang
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Mei Hu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
3
|
Zhang S, Kong F, Zheng L, Li X, Jia L, Yang L. SEMA7A as a Novel Prognostic Biomarker and Its Correlation with Immune Infiltrates in Breast Cancer. Int J Gen Med 2024; 17:4081-4099. [PMID: 39295856 PMCID: PMC11410036 DOI: 10.2147/ijgm.s474827] [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: 04/21/2024] [Accepted: 09/12/2024] [Indexed: 09/21/2024] Open
Abstract
Background The role of Semaphorin 7a (SEMA7A) in the initiation and progression of different types of cancerous lesions has been extensively studied. However, the prognostic significance of SEMA7A, specifically in breast cancer (BC), lacks clarity. Methods We conducted an evaluation on the relationship between SEMA7A and the prognosis, immune invasion and tumor mutation burden in different types of cancer by analyzing data from The Cancer Genome Atlas database. The present study focused on investigating the expression level, mutation, immune correlation and coexpression of SEMA7A in BC, utilizing various databases such as the University of Alabama at Birmingham Cancer data analysis portal, cBioPortal and tumor immune estimation resource. Survival analysis was carried out using the Kaplan-Meier Plotter. Furthermore, we employed the R software package to generate receiver operating characteristic (ROC) curves and nomograms. Notably, P<0.05 was considered to indicate statistical significance. Results Using pancancer analysis, it has been observed that the expression of SEMA7A is elevated in various types of cancer and is strongly correlated with the prognosis of different cancer types. SEMA7A also exhibits a significant association with the tumor mutation burden of diverse types of cancer. Moreover, SEMA7A displays a notable increase in BC cases, and was indicated to have a substantial association with the abundance of immune infiltration. In-depth survival analysis demonstrated that elevated levels of SEMA7A expression are notably linked to shorter overall survival and distant metastasis-free survival among patients with BC. The efficiency of SEMA7A as a reliable prognostic biomarker for BC has been substantiated by the validation of ROC curves and nomograms. Conclusion SEMA7A has the potential to function as a prognostic indicator for BC, and its correlation with immune infiltration in BC is significant.
Collapse
Affiliation(s)
- Shiyu Zhang
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| | - Fanting Kong
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| | - Lei Zheng
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| | - Xiaowei Li
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| | - Lining Jia
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| | - Lixian Yang
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, Hebei, 054000, People's Republic of China
| |
Collapse
|
4
|
Wen Y, Li C, Huang P, Liu Z, He Y, Liu B. Transcriptional landscape of intestinal environment in DSS-induced ulcerative colitis mouse model. BMC Gastroenterol 2024; 24:60. [PMID: 38308210 PMCID: PMC10836045 DOI: 10.1186/s12876-024-03128-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 01/05/2024] [Indexed: 02/04/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease that targets the colon and has seen an increasing prevalence worldwide. In our pursuit of new diagnostic and therapeutic approaches for UC, we undertook a sequencing of colons from UC mouse models. We focused on analyzing their differentially expressed genes (DEGs), enriching pathways, and constructing protein-protein interaction (PPI) and Competing Endogenous RNA (ceRNA) networks. Our analysis highlighted novel DEGs such as Tppp3, Saa3, Cemip, Pappa, and Nr1d1. These DEGs predominantly play roles in pathways like cytokine-mediated signaling, extracellular matrix organization, extracellular structure organization, and external encapsulating structure organization. This suggests that the UC pathogenesis is intricately linked to the interactions between immune and non-immune cells with the extracellular matrix (ECM). To corroborate our findings, we also verified certain DEGs through quantitative real-time PCR. Within the PPI network, nodes like Stat3, Il1b, Mmp3, and Lgals3 emerged as significant and were identified to be involved in the crucial cytokine-mediated signaling pathway, which is central to inflammation. Our ceRNA network analysis further brought to light the role of the Smad7 Long non-coding RNA (lncRNA). Key MicroRNA (miRNAs) in the ceRNA network were pinpointed as mmu-miR-17-5p, mmu-miR-93-5p, mmu-miR-20b-5p, mmu-miR-16-5p, and mmu-miR-106a-5p, while central mRNAs included Egln3, Plagl2, Sema7a, Arrdc3, and Stat3. These insights imply that ceRNA networks are influential in UC progression and could provide further clarity on its pathogenesis. In conclusion, this research deepens our understanding of UC pathogenesis and paves the way for potential new diagnostic and therapeutic methods. Nevertheless, to solidify our findings, additional experiments are essential to confirm the roles and molecular interplay of the identified DEGs in UC.
Collapse
Affiliation(s)
- Yuefei Wen
- Foshan Maternity and Child Health Care Hospital, Southern Medical University, 528000, Foshan, China
| | - Chenyang Li
- Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China
| | - Peng Huang
- Foshan Maternity and Child Health Care Hospital, Southern Medical University, 528000, Foshan, China
| | - Zhigang Liu
- Foshan Maternity and Child Health Care Hospital, Southern Medical University, 528000, Foshan, China
| | - Yanjun He
- Foshan Maternity and Child Health Care Hospital, Southern Medical University, 528000, Foshan, China.
- Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China.
| | - Bin Liu
- Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, China.
| |
Collapse
|
5
|
Körner A, Köhler D, Schneider M, Roth JM, Granja TF, Eggstein C, Mirakaj V, Rosenberger P. Semaphorin 7A is protective during inflammatory peritonitis through integrin receptor signaling. Front Immunol 2023; 14:1251026. [PMID: 38094294 PMCID: PMC10716335 DOI: 10.3389/fimmu.2023.1251026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction The study explores the role of endothelial Semaphorin 7A (SEMA7A) in inflammatory processes. SEMA7A is known for enhancing inflammation during tissue hypoxia and exhibiting anti-inflammatory properties in the intestinal system during colitis. This research extends the understanding of SEMA7A's function by examining its role in inflammatory peritonitis and intestinal inflammation. Methods The research involved inducing peritonitis in SEMA7A knockout (SEMA7A-/-) and wild-type (WT) animals through Zymosan A (ZyA) injection. The inflammatory response was assessed by measuring cell count and cytokine release. In parallel, the study investigated the expression of SEMA7A in intestinal epithelial cells under inflammatory stimuli and its impact on interleukin 10 (IL-10) production using an in vitro co-culture model of monocytes and epithelial cells. Additionally, the distribution of SEMA7A target receptors, particularly ITGAV/ITGB1 (CD51/CD29), was analyzed in WT animals. Results The results revealed that SEMA7A-/- animals exhibited increased inflammatory peritonitis compared to the WT animals. Inflammatory conditions in intestinal epithelial cells led to the induction of SEMA7A. The co-culture experiments demonstrated that SEMA7A induced IL-10 production, which depended on integrin receptors and was independent of PLXNC1 expression. Furthermore, ITGAV/ITGB1 emerged as the predominant SEMA7A receptor in the intestinal area of WT animals. Discussion These findings underscore the multifaceted role of SEMA7A in inflammatory processes. The differential responses in peritonitis and intestinal inflammation suggest that SEMA7A's function is significantly influenced by the expression and distribution of its target receptors within different organ systems. The study highlights the complex and context-dependent nature of SEMA7A in mediating inflammatory responses.
Collapse
Affiliation(s)
- Andreas Körner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - David Köhler
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Mariella Schneider
- Department of Obstetrics and Gynecology, Augsburg University Hospital, Augsburg, Germany
| | - Judith M. Roth
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Tiago F. Granja
- CBIOS-Universidade Research Center for Biosciences & Health Technologies, Universidade Lusófona, Lisbon, Portugal
| | - Claudia Eggstein
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Valbona Mirakaj
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| |
Collapse
|
6
|
Vargovic M, Papic N, Samadan L, Balen Topic M, Vince A. Association of Immune Semaphorins with COVID-19 Severity and Outcomes. Biomedicines 2023; 11:2786. [PMID: 37893159 PMCID: PMC10604420 DOI: 10.3390/biomedicines11102786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/08/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Semaphorins have recently been recognized as crucial modulators of immune responses. In the pathogenesis of COVID-19, the activation of immune responses is the key factor in the development of severe disease. This study aimed to determine the association of serum semaphorin concentrations with COVID-19 severity and outcomes. Serum semaphorin concentrations (SEMA3A, -3C, -3F, -4D, -7A) were measured in 80 hospitalized adult patients with COVID-19 (moderate (n = 24), severe (n = 32), critical, (n = 24)) and 40 healthy controls. While SEMA3C, SEMA3F and SEMA7A serum concentrations were significantly higher in patients with COVID-19, SEMA3A was significantly lower. Furthermore, SEMA3A and SEMA3C decreased with COVID-19 severity, while SEMA3F and SEMA7A increased. SEMA4D showed no correlation with disease severity. Serum semaphorin levels show better predictive values than CRP, IL-6 and LDH for differentiating critical from moderate/severe COVID-19. SEMA3F and SEMA7A serum concentrations were associated with the time to recovery, requirement of invasive mechanical ventilation, development of pulmonary thrombosis and nosocomial infections, as well as with in-hospital mortality. In conclusion, we provide the first evidence that SEMA3A, SEMA3C, SEMA3F and SEMA7A can be considered as new biomarkers of COVID-19 severity.
Collapse
Affiliation(s)
- Martina Vargovic
- Department for Infections in the Immunocompromised, University Hospital for Infectious Diseases, 10000 Zagreb, Croatia;
| | - Neven Papic
- Department for Viral Hepatitis, University Hospital for Infectious Diseases, 10000 Zagreb, Croatia;
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.S.); (M.B.T.)
| | - Lara Samadan
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.S.); (M.B.T.)
| | - Mirjana Balen Topic
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.S.); (M.B.T.)
- Department for Gastrointestinal Infections, University Hospital for Infectious Diseases, 10000 Zagreb, Croatia
| | - Adriana Vince
- Department for Viral Hepatitis, University Hospital for Infectious Diseases, 10000 Zagreb, Croatia;
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.S.); (M.B.T.)
| |
Collapse
|
7
|
Schimpel C, Passegger C, Egger S, Tam-Amersdorfer C, Strobl H. A novel 3D cell culture model to study the human small intestinal immune landscape. Eur J Immunol 2023; 53:e2250131. [PMID: 36527196 DOI: 10.1002/eji.202250131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/21/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Several subsets of mononuclear phagocytes and DCs (MDC) populate the small intestine (SI), and these cells reportedly exert specialized functions in anti-microbial immunity and tolerance. Given the specialized phenotype of these cells, differing from other MDC family members, including their putative circulating blood precursors, local intestinal factors play key instructive roles in their differentiation. We designed an SI cell culture model composed of three intestinal epithelial cell (IEC) types, including absorptive enterocytes (E cells), antigen delivering microfold (M) cells, and mucus-producing goblet (G) cells plus T lymphocytes and soluble B cell-derived factors. This model was used to study the differentiation fate of CD34+ hematopoietic progenitor cell-derived monocyte/DC precursors. Progeny cells can be analyzed after a 3-week co-culture period, mimicking the physiologic turn-over time of intestinal MDC. A dominant monocyte differentiation pathway was suppressed, in favor of partial differentiation along DC and macrophage pathways, with low percentages of cells acquired DC or macrophage markers. Moreover, E and G cells play opposing roles in CX3CR1+ vs CD103dim cell differentiation, indicating that both together might counter-balance M/DC differentiation. Thus, SI epithelial cells suppress M/DC differentiation, supporting a key role for exogenous factors in M/DC differentiation.
Collapse
Affiliation(s)
- Christa Schimpel
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Christina Passegger
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Simone Egger
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Carmen Tam-Amersdorfer
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Herbert Strobl
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| |
Collapse
|
8
|
Šamadan L, Papić N, Mijić M, Knežević Štromar I, Gašparov S, Vince A. Do Semaphorins Play a Role in Development of Fibrosis in Patients with Nonalcoholic Fatty Liver Disease? Biomedicines 2022; 10:3014. [PMID: 36551769 PMCID: PMC9775767 DOI: 10.3390/biomedicines10123014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is associated with systemic changes in immune response linked with chronic low-grade inflammation and disease progression. Semaphorins, a large family of biological response modifiers, were recently recognized as one of the key regulators of immune responses, possibly also associated with chronic liver diseases. The aim of this study was to identify semaphorins associated with NAFLD and their relationship with steatosis and fibrosis stages. In this prospective, case-control study, serum semaphorin concentrations (SEMA3A, -3C, -4A, -4D, -5A and -7A) were measured in 95 NAFLD patients and 35 healthy controls. Significantly higher concentrations of SEMA3A, -3C and -4D and lower concentrations of SEAMA5A and -7A were found in NAFLD. While there was no difference according to steatosis grades, SEMA3C and SEMA4D significantly increased and SEMA3A significantly decreased with fibrosis stages and had better accuracy in predicting fibrosis compared to the FIB-4 score. Immunohistochemistry confirmed higher expression of SEMA4D in hepatocytes, endothelial cells and lymphocytes in NAFLD livers. The SEMA5A rs1319222 TT genotype was more frequent in the NAFLD group and was associated with higher liver stiffness measurements. In conclusion, we provide the first evidence of the association of semaphorins with fibrosis in patients with NAFLD.
Collapse
Affiliation(s)
- Lara Šamadan
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Neven Papić
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department for Viral Hepatitis, University Hospital for Infectious Diseases Zagreb, 10000 Zagreb, Croatia
| | - Maja Mijić
- Department of Internal Medicine, Division of Gastroenterology, University Hospital Merkur, 10000 Zagreb, Croatia
| | - Ivana Knežević Štromar
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Slavko Gašparov
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Pathology, University Hospital Merkur, 10000 Zagreb, Croatia
| | - Adriana Vince
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department for Viral Hepatitis, University Hospital for Infectious Diseases Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
9
|
Crump LS, Kines KT, Richer JK, Lyons TR. Breast cancers co-opt normal mechanisms of tolerance to promote immune evasion and metastasis. Am J Physiol Cell Physiol 2022; 323:C1475-C1495. [PMID: 36189970 PMCID: PMC9662806 DOI: 10.1152/ajpcell.00189.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022]
Abstract
Normal developmental processes, such as those seen during embryonic development and postpartum mammary gland involution, can be reactivated by cancer cells to promote immune suppression, tumor growth, and metastatic spread. In mammalian embryos, paternal-derived antigens are at risk of being recognized as foreign by the maternal immune system. Suppression of the maternal immune response toward the fetus, which is mediated in part by the trophoblast, is critical to ensure embryonic survival and development. The postpartum mammary microenvironment also exhibits immunosuppressive mechanisms accompanying the massive cell death and tissue remodeling that occurs during mammary gland involution. These normal immunosuppressive mechanisms are paralleled during malignant transformation, where tumors can develop neoantigens that may be recognized as foreign by the immune system. To circumvent this, tumors can dedifferentiate and co-opt immune-suppressive mechanisms normally utilized during fetal tolerance and postpartum mammary involution. In this review, we discuss those similarities and how they can inform our understanding of cancer progression and metastasis.
Collapse
Affiliation(s)
- Lyndsey S Crump
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kelsey T Kines
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, Aurora, Colorado
| | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, Aurora, Colorado
| |
Collapse
|
10
|
Zhao N, Zhang X, Ding J, Pan Q, Zheng MH, Liu WY, Luo G, Qu J, Li M, Li L, Cheng Y, Peng Y, Xie Q, Wei Q, Li Q, Zou L, Ouyang X, Cai SY, Boyer JL, Chai J. SEMA7AR148W mutation promotes lipid accumulation and NAFLD progression via increased localization on the hepatocyte surface. JCI Insight 2022; 7:e154113. [PMID: 35938531 PMCID: PMC9462498 DOI: 10.1172/jci.insight.154113] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 06/27/2022] [Indexed: 12/30/2022] Open
Abstract
Genetic polymorphisms are associated with the development of nonalcoholic fatty liver disease (NAFLD). Semaphorin7a (Sema7a) deficiency in mouse peritoneal macrophages reduces fatty acid (FA) oxidation. Here, we identified 17 individuals with SEMA7A heterozygous mutations in 470 patients with biopsy-proven NAFLD. SEMA7A heterozygous mutations increased susceptibility to NAFLD, steatosis severity, and NAFLD activity scores in humans and mice. The Sema7aR145W mutation (equivalent to human SEMA7AR148W) significantly induced small lipid droplet accumulation in mouse livers compared with WT mouse livers. Mechanistically, the Sema7aR145W mutation increased N-glycosylated Sema7a and its receptor integrin β1 proteins in the cell membranes of hepatocytes. Furthermore, Sema7aR145W mutation enhanced its protein interaction with integrin β1 and PKC-α and increased PKC-α phosphorylation, which were both abrogated by integrin β1 silencing. Induction of PKCα_WT, but not PKCα_dominant negative, overexpression induced transcriptional factors Srebp1, Chrebp, and Lxr expression and their downstream Acc1, Fasn, and Cd36 expression in primary mouse hepatocytes. Collectively, our findings demonstrate that the SEMA7AR148W mutation is a potentially new strong genetic determinant of NAFLD and promotes intrahepatic lipid accumulation and NAFLD in mice by enhancing PKC-α-stimulated FA and triglyceride synthesis and FA uptake. The inhibition of hepatic PKC-α signaling may lead to novel NAFLD therapies.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaoxun Zhang
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Jingjing Ding
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiong Pan
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | | | - Wen-Yue Liu
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gang Luo
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiaquan Qu
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Mingqiao Li
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Ling Li
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Ying Cheng
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Ying Peng
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiaoling Xie
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Qinglin Wei
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiao Li
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| | - Lingyun Zou
- Bioinformatics Center, Department of Microbiology of Third Military Medical University, Chongqing, China
- Bao’an Maternal and Child Health Hospital of Jinan University, Shenzhen, China
| | - Xinshou Ouyang
- Department of Internal Medicine, Section of Digestive Diseases, and
| | - Shi-Ying Cai
- Department of Internal Medicine and Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - James L. Boyer
- Department of Internal Medicine and Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jin Chai
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
11
|
Cao Q, Mertens RT, Sivanathan KN, Cai X, Xiao P. Macrophage orchestration of epithelial and stromal cell homeostasis in the intestine. J Leukoc Biol 2022; 112:313-331. [PMID: 35593111 PMCID: PMC9543232 DOI: 10.1002/jlb.3ru0322-176r] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 11/06/2022] Open
Abstract
The intestinal tract is a complex ecosystem where numerous cell types of epithelial, immune, neuronal, and endothelial origin coexist in an intertwined, highly organized manner. The functional equilibrium of the intestine relies heavily on the proper crosstalk and cooperation among each cell population. Furthermore, macrophages are versatile, innate immune cells that participate widely in the modulation of inflammation and tissue remodeling. Emerging evidence suggest that macrophages are central in orchestrating tissue homeostasis. Herein, we describe how macrophages interact with epithelial cells, neurons, and other types of mesenchymal cells under the context of intestinal inflammation, followed by the therapeutic implications of cellular crosstalk pertaining to the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Qian Cao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Randall Tyler Mertens
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Kisha Nandini Sivanathan
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Xuechun Cai
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China.,Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
12
|
Nakanishi Y, Kang S, Kumanogoh A. Crosstalk between axon guidance signaling and bone remodeling. Bone 2022; 157:116305. [PMID: 34973495 DOI: 10.1016/j.bone.2021.116305] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 01/04/2023]
Abstract
The maintenance of skeletal integrity is tightly regulated by two cell types, bone forming osteoblasts and bone resorbing osteoclasts. Although the role of the nervous system in regulating osteoblasts and osteoclasts was identified over a decade ago, the molecular mechanism of skeletal-neural interactions in bone homeostasis has only been studied recently. In particular, the complex roles of axon guidance molecules, such as semaphorins and ephrins, in the bone have been studied extensively. In this review, we highlight the latest advances in determining the functions of semaphorins and ephrins in the establishment and maintenance of the skeletal system, with a focus on the functional interaction between the skeletal and nervous systems.
Collapse
Affiliation(s)
- Yoshimitsu Nakanishi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita City, Osaka 565-0871, Japan
| | - Sujin Kang
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita City, Osaka 565-0871, Japan; Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
13
|
Chen W, Liu D, Ren C, Su X, Wong CK, Yang R. A Special Network Comprised of Macrophages, Epithelial Cells, and Gut Microbiota for Gut Homeostasis. Cells 2022; 11:cells11020307. [PMID: 35053422 PMCID: PMC8774616 DOI: 10.3390/cells11020307] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 12/15/2022] Open
Abstract
A number of gut epithelial cells derived immunological factors such as cytokines and chemokines, which are stimulated by the gut microbiota, can regulate host immune responses to maintain a well-balance between gut microbes and host immune system. Multiple specialized immune cell populations, such as macrophages, dendritic cells (DCs), innate lymphoid cells, and T regulatory (Treg) cells, can communicate with intestinal epithelial cells (IEC) and/or the gut microbiota bi-directionally. The gut microbiota contributes to the differentiation and function of resident macrophages. Situated at the interface between the gut commensals and macrophages, the gut epithelium is crucial for gut homeostasis in microbial recognition, signaling transformation, and immune interactions, apart from being a physical barrier. Thus, three distinct but interactive components—macrophages, microbiota, and IEC—can form a network for the delicate and dynamic regulation of intestinal homeostasis. In this review, we will discuss the crucial features of gut microbiota, macrophages, and IEC. We will also summarize recent advances in understanding the cooperative and dynamic interactions among the gut microbiota, gut macrophages, and IEC, which constitute a special network for gut homeostasis.
Collapse
Affiliation(s)
- Wei Chen
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China; (W.C.); (D.L.); (C.R.); (X.S.)
| | - Dan Liu
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China; (W.C.); (D.L.); (C.R.); (X.S.)
| | - Changhao Ren
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China; (W.C.); (D.L.); (C.R.); (X.S.)
| | - Xiaomin Su
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China; (W.C.); (D.L.); (C.R.); (X.S.)
| | - Chun-Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong 999077, China;
| | - Rongcun Yang
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China; (W.C.); (D.L.); (C.R.); (X.S.)
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- Correspondence:
| |
Collapse
|
14
|
Li Q, Lian Y, Deng Y, Chen J, Wu T, Lai X, Zheng B, Qiu C, Peng Y, Li W, Xiang AP, Zhang X, Ren J. mRNA-engineered mesenchymal stromal cells expressing CXCR2 enhances cell migration and improves recovery in IBD. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:222-236. [PMID: 34513306 PMCID: PMC8413681 DOI: 10.1016/j.omtn.2021.07.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/13/2021] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) have shown significant heterogeneity in terms of therapeutic efficacy for inflammatory bowel disease (IBD) treatment, which may be due to an insufficient number of MSCs homing to the damaged tissue of the colon. Engineering MSCs with specific chemokine receptors can enhance the homing ability by lentiviral transduction. However, the unclear specific chemokine profile related to IBD and the safety concerns of viral-based gene delivery limit its application. Thus, a new strategy to modify MSCs to express specific chemokine receptors using mRNA engineering is developed to evaluate the homing ability of MSCs and its therapeutic effects for IBD. We found that CXCL2 and CXCL5 were highly expressed in the inflammatory colon, while MSCs minimally expressed the corresponding receptor CXCR2. Transient expression of CXCR2 in MSC was constructed and exhibited significantly enhanced migration to the inflamed colons, leading to a robust anti-inflammatory effect and high efficacy. Furthermore, the high expression of semaphorins7A on MSCs were found to induce the macrophages to produce IL-10, which may play a critical therapeutic role. This study demonstrated that the specific chemokine receptor CXCR2 mRNA-engineered MSCs not only improves the therapeutic efficacy of IBD but also provides an efficient and safe MSC modification strategy.
Collapse
Affiliation(s)
- Qiaojia Li
- Department of Medical Ultrasonic, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou 510630, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yufan Lian
- Department of Medical Ultrasonic, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou 510630, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yiwen Deng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou 510630, China
| | - Jieying Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou 510630, China
| | - Tao Wu
- Department of Medical Ultrasonic, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou 510630, China
| | - Xinqiang Lai
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
- Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Bowen Zheng
- Department of Medical Ultrasonic, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou 510630, China
| | - Chen Qiu
- Department of Medical Ultrasonic, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou 510630, China
| | - Yanwen Peng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou 510630, China
| | - Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou 510630, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou 510630, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou 510630, China
| | - Jie Ren
- Department of Medical Ultrasonic, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou 510630, China
| |
Collapse
|
15
|
Abstract
Nonresolving inflammation, a hallmark of sepsis and/or multi-organ failure, still poses a challenge in medicine. The mortality rate is enormous, and so far no adequate curative therapy is available. Here we identify a previously unrecognized role of the neuronal guidance protein semaphorin 7A in the transition to resolution processes in severe systematic inflammation such as sepsis. Endogenous mediators regulating acute inflammatory responses in both the induction and resolution phases of inflammatory processes are pivotal in host defense and tissue homeostasis. Recent studies have identified neuronal guidance proteins characterized in axonal development that display immunomodulatory functions. Here, we identify the neuroimmune guidance cue Semaphorin 7A (Sema7A), which appears to link macrophage (MΦ) metabolic remodeling to inflammation resolution. Sema7A orchestrated MΦ chemotaxis and chemokinesis, activated MΦ differentiation and polarization toward the proresolving M2 phenotype, and promoted leukocyte clearance. Peritoneal MΦSema7A−/− displayed metabolic reprogramming, characterized by reductions in fatty acid oxidation and oxidative phosphorylation, increases in glycolysis and the pentose phosphate pathway, and truncation of the tricarboxylic acid cycle, which resulted in increased levels of the intermediates succinate and fumarate. The low accumulation of citrate in MΦSema7A−/− correlated with the decreased synthesis of prostaglandins, leading to a reduced impact on lipid-mediator class switching and the generation of specialized pro resolving lipid mediators. Signaling network analysis indicated that Sema7A induced the metabolic reprogramming of MΦ by activating the mTOR- and AKT2-signaling pathways. Administration of Sema7ASL4cd orchestrated the resolution response to tissue homeostasis by shortening the resolution interval, promoting tissue protection in murine peritonitis, and enhancing survival in polymicrobial sepsis.
Collapse
|
16
|
Nakanishi Y, Kang S, Kumanogoh A. Neural guidance factors as hubs of immunometabolic crosstalk. Int Immunol 2021; 33:749-754. [PMID: 34174067 PMCID: PMC8633672 DOI: 10.1093/intimm/dxab035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/24/2021] [Indexed: 11/14/2022] Open
Abstract
Semaphorins were originally identified as axon-guidance molecules essential for neural development. In addition to their functions in the neural system, members of the semaphorin family have critical functions in many pathophysiological processes, including immune responses, bone homeostasis, cancer and metabolic disorders. In particular, several lines of evidence indicate that mammalian/mechanistic target of rapamycin (mTOR), a central regulator of cell metabolism, regulates the functions of semaphorins in various types of cells, revealing a novel link between semaphorins and cell metabolism. In this review, we discuss recent advances in the immunometabolic functions of semaphorins, with a particular focus on mTOR signaling.
Collapse
Affiliation(s)
- Yoshimitsu Nakanishi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita City, Osaka 565-0871, Japan.,Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita City, Osaka 565-0871, Japan
| | - Sujin Kang
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita City, Osaka 565-0871, Japan.,Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita City, Osaka 565-0871, Japan
| |
Collapse
|
17
|
Song Y, Wang L, Li J, Yang F, Gao Y, Song D, Sun J, Ye L, Zhang L, Huang D. The Expression of Semaphorin 7A in Human Periapical Lesions. J Endod 2021; 47:1631-1639. [PMID: 34126161 DOI: 10.1016/j.joen.2021.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Semaphorin 7A (SEMA7A) is a membrane-bound or secretory protein exerting multiple functions in the regulation of inflammation, neural degradation, and cancer progression. Human periapical lesions are chronic and infectious diseases mainly caused by bacteria. However, the involvement of SEMA7A in human periapical lesions is still unclear. This study aimed to explore the expression of SEMA7A in human periapical lesions accompanied by the potential association of SEMA7A with matrix metalloproteinase (MMP)-1 and MMP-3 during the progression of apical periodontitis. METHODS Samples of periapical lesions and healthy controls were collected. Total RNA and protein were extracted respectively for quantitative real-time polymerase chain reaction and Western blot analysis. Additionally, 6 healthy samples and 27 periapical lesion samples were fixed, dehydrated, and embedded for further histologic and immunochemical analysis. The expression of SEMA7A was quantified by average integrated optical density. Immunofluorescence analysis was conducted to explore the colocalization of SEMA7A/MMP-1 and SEMA7A/MMP-3. RESULTS Compared with healthy controls, the messenger RNA and protein expression of SEMA7A was markedly up-regulated in periapical lesions. A stronger expression of MMP-1, MMP-3, and inflammatory cytokines was exhibited in periapical lesions than in healthy groups. An increasing expression of SEMA7A can be observed in both the periapical granuloma group and the radicular cyst group compared with the normal group (P < .01). Immunofluorescence results showed the colocalization of SEMA7A with both MMP-1 and MMP-3 in vascular vessels and extracellular matrix. CONCLUSIONS SEMA7A was up-regulated in periapical periodontitis and might be involved in the tissue destruction and infiltration of immune cells in periapical lesions.
Collapse
Affiliation(s)
- Yao Song
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liu Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiatong Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fan Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuxuan Gao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dongzhe Song
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianxun Sun
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lan Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Dingming Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
18
|
Song Y, Wang L, Zhang L, Huang D. The involvement of semaphorin 7A in tumorigenic and immunoinflammatory regulation. J Cell Physiol 2021; 236:6235-6248. [PMID: 33611799 DOI: 10.1002/jcp.30340] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/12/2021] [Accepted: 02/05/2021] [Indexed: 02/04/2023]
Abstract
Semaphorins, a large group of highly conserved proteins, consist of eight subfamilies that are widely expressed in vertebrates, invertebrates, and viruses and exist in membrane-bound or secreted forms. First described as axon guidance cues during neurogenesis, semaphorins also perform physiological functions in other organ systems, such as bone homeostasis, immune response, and tumor progression. Semaphorin 7A (SEMA7A), also known as CDw108, is an immune semaphorin that modulates diverse immunoinflammatory processes, including immune cell interactions, inflammatory infiltration, and cytokine production. In addition, SEMA7A regulates the proliferation, migration, invasion, lymph formation, and angiogenesis of multiple types of tumor cells, and these effects are mediated by the interaction of SEMA7A with two specific receptors, PLXNC1 and integrins. Thus, SEMA7A is intimately related to the pathogenesis of multiple autoimmune and inflammation-related diseases and tumors. This review focuses on the role of SEMA7A in the pathogenesis of autoimmune disorders, inflammatory diseases, and tumors, as well as the underlying mechanisms. Furthermore, strategies targeting SEMA7A as a potential predictive, diagnostic, and therapeutic agent for these diseases are also addressed.
Collapse
Affiliation(s)
- Yao Song
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liu Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lan Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
19
|
Li J, Zhou H, Fu X, Zhang M, Sun F, Fan H. Dynamic role of macrophage CX3CR1 expression in inflammatory bowel disease. Immunol Lett 2021; 232:39-44. [PMID: 33582183 DOI: 10.1016/j.imlet.2021.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/23/2021] [Accepted: 02/02/2021] [Indexed: 12/18/2022]
Abstract
Inflammatory bowel disease (IBD), consisting of ulcerative colitis (UC) and Crohn's disease (CD), is featured by overactive immune response and enduring course of unrestrained colitis. Genetic predisposition and environmental factors are fundamental in disease progression. Notably, microbiota dysregulation and its interaction with host mucosal barrier perplex disease phenotype. Under experimental setting, distinct mouse models are established to mimic human colitis process, including infection induced dysbiosis, dextran sulfate sodium (DSS) etc. induced barrier destruction, anti-CD40 L induced innate immunity dominant colitis and T cell transfer colitis model. Thus, from a more detailed aspect, IBD is heterogeneous and can be further classified into different subtypes based on the specific etiological pathways. As a typical inflammatory disorder, various immune cell types are involved in IBD pathogenesis. Among them, macrophages are believed to play a pivotal role. CX3CR1+ macrophages, deriving from peripheral patrolling CD14+ Ly6Chi monocytes, are specified cell population dwelling in the gut. Accumulating evidence suggests that CX3CR1+ macrophages are critical for mucosal homeostasis and IBD pathogenesis, while some conflicts exist in current studies with both protective and harmful effects being revealed. Herein, we reviewed published literatures and found that the observed discrepancies stem from many aspects: the expression level of CX3CR1, the confounding dendritic cell subsets and most importantly, the different colitis stages and subtypes. Overall, CX3CR1 targeting strategy could be powerful weapon in fighting against colitis, but at the same time, the precise etiological and pathological mechanisms should be cautiously examined concerning the appropriate usage of CX3CR1 targeted therapy.
Collapse
Affiliation(s)
- Junyi Li
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haifeng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Fu
- Department of Traditional Chinese Medicine, Guangzhou Eighth People's Hospital, Guangzhou, China
| | - Meng Zhang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Sun
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
20
|
Wang P, Mao YM, Liu LN, Zhao CN, Li XM, Pan HF. Decreased Expression of Semaphorin 3A and Semaphorin 7A Levels and Its Association with Systemic Lupus Erythematosus. Immunol Invest 2019; 49:69-80. [PMID: 31412748 DOI: 10.1080/08820139.2019.1649280] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
A growing body of data suggests that semaphorins are involved in both normal and pathological immune responses, as well as autoimmune pathologies. To investigate the plasma semaphorin 3A (Sema3A) and semaphorin 7A (Sema7A) levels in systemic lupus erythematosus (SLE) patients and their correlation with clinical manifestations and laboratory indexes, a two-step method was applied. First, 80 SLE patients and 80 healthy controls were recruited for comparing serum Sema3A and Sema7A concentrations. Second, 40 rheumatoid arthritis (RA) patients and 40 sjögren's syndrome (SS) patients were then included as disease controls. Plasma Sema3A and Sema7A concentrations were detected by ELISA. There were significant differences in Sema3A and Sema7A among four groups. When compared to healthy controls, both Sema3A and Sema7A levels were decreased in SLE and increased in RA; increased Sema3A level and decreased Sema7A level were found in SS. There were significant differences in Sema3A concentration between SLE and RA, SLE and SS. Moreover, there were significant differences in Sema7A level between SLE and RA, SS and RA. However, no significant differences in Sema3A between SS and RA and no significant differences in Sema7A between SS and SLE were observed. Both plasma Sema3A and Sema7A levels were correlated with anti-SSA and IgM. Area under curve (AUC) of the receiver operating characteristic (ROC) curve for Sema3A and Sema7A were 0.535 (0.455-0.613) and 0.671 (0.594-0.742), respectively. Aberrant Sema3A and Sema7A expression and their clinical associations in SLE suggest their important role in this disease.
Collapse
Affiliation(s)
- Peng Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yan-Mei Mao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Department of Epidemiology, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, China
| | - Li-Na Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Department of Epidemiology, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, China
| | - Chan-Na Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Department of Epidemiology, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, China
| | - Xiao-Mei Li
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, Anhui, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.,Department of Epidemiology, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, China
| |
Collapse
|
21
|
Han YF, Liu Z, Wang B, Zhu W, Li JZ, Qi YQ, Li XJ, Xu YY, Dou XX, Mu GY. Semaphorin 7a participants in pterygium by regulating vascular endothelial growth factor. Int J Ophthalmol 2019; 12:892-897. [PMID: 31236342 PMCID: PMC6580203 DOI: 10.18240/ijo.2019.06.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/03/2019] [Indexed: 11/23/2022] Open
Abstract
AIM To investigate the relationship between semaphorin 7a expression and cell proliferation and migration in pterygium fibroblasts. METHODS Twenty-six patients with surgically diagnosed pterygium were enrolled, including 15 cases of primary pterygium and 11 cases of recurrent pterygium. In addition, 12 cases of normal conjunctival tissue were collected. The expression of semaphorin 7a in normal conjunctival tissue, primary pterygium and recurrent pterygium was detected by real-time polymerase chain reaction. Recurrent pterygium fibroblasts were isolated and cultured, and the expression of semaphorin 7a was silenced by small interfering RNA (siRNA) interference technique. Furthermore, the effects of si-semaphorin 7a interference on the mRNA and protein levels of β1-integrin, vascular endothelial growth factor A (VEGFA) and vascular endothelial growth factor receptor (VEGFR), and on fibroblast proliferation were analyzed. Transwell assay was used to detect the effect of semaphorin 7a interference on fibroblast migration. RESULTS Semaphorin 7a was highly expressed in the primary pterygium and recurrent pterygium samples than that of the normal conjunctival tissue. Compared with the primary pterygium, the expression of semaphoring 7a in the recurrent pterygium samples was significantly increased (P<0.05). The mRNA and protein expression levels of β1-integrin, VEGFA and VEGFR were decreased after si-semaphorin 7a transfection, and as well as the cell proliferation and migration. CONCLUSION Semaphorin 7a might play important roles in the pathogenesis of pterygium by affecting the expression of β1-integrin, VEGFA and VEGFR.
Collapse
Affiliation(s)
- Yun-Fei Han
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China
- Aier Eye Hospital Group, Hubin Aier Eye Hospital, Binzhou 256600, Shandong Province, China
| | - Zhen Liu
- Department of Ophthalmology, the Second People's Hospital of Liaocheng, Linqing 252600, Shandong Province, China
| | - Bang Wang
- Neonatal Intensive Care Unit, Binzhou Medical University Hospital, Binzhou 256600, Shandong Province, China
| | - Wei Zhu
- Department of Ophthalmology, Jinan Central Hospital Affiliated to Shandong University, Jinan 250013, Shandong Province, China
| | - Jing-Zhen Li
- Aier Eye Hospital Group, Hubin Aier Eye Hospital, Binzhou 256600, Shandong Province, China
| | - Yue-Qin Qi
- Aier Eye Hospital Group, Hubin Aier Eye Hospital, Binzhou 256600, Shandong Province, China
| | - Xiao-Jing Li
- Aier Eye Hospital Group, Hubin Aier Eye Hospital, Binzhou 256600, Shandong Province, China
| | - Yan-Yun Xu
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China
| | - Xiao-Xiao Dou
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Guo-Ying Mu
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China
| |
Collapse
|
22
|
Kinehara Y, Nagatomo I, Koyama S, Ito D, Nojima S, Kurebayashi R, Nakanishi Y, Suga Y, Nishijima-Futami Y, Osa A, Nakatani T, Kato Y, Nishide M, Hayama Y, Higashiguchi M, Morimura O, Miyake K, Kang S, Minami T, Hirata H, Iwahori K, Takimoto T, Takamatsu H, Takeda Y, Hosen N, Hoshino S, Shintani Y, Okumura M, Kumagai T, Nishino K, Imamura F, Nakatsuka SI, Kijima T, Kida H, Kumanogoh A. Semaphorin 7A promotes EGFR-TKI resistance in EGFR mutant lung adenocarcinoma cells. JCI Insight 2018; 3:123093. [PMID: 30568033 DOI: 10.1172/jci.insight.123093] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 11/06/2018] [Indexed: 12/17/2022] Open
Abstract
Although responses to EGFR tyrosine kinase inhibitors (EGFR-TKIs) are initially positive, 30%-40% of patients with EGFR-mutant tumors do not respond well to EGFR-TKIs, and most lung cancer patients harboring EGFR mutations experience relapse with resistance. Therefore, it is necessary to identify not only the mechanisms underlying EGFR-TKI resistance, but also potentially novel therapeutic targets and/or predictive biomarkers for EGFR-mutant lung adenocarcinoma. We found that the GPI-anchored protein semaphorin 7A (SEMA7A) is highly induced by the EGFR pathway, via mTOR signaling, and that expression levels of SEMA7A in human lung adenocarcinoma specimens were correlated with mTOR activation. Investigations using cell culture and animal models demonstrated that loss or overexpression of SEMA7A made cells less or more resistant to EGFR-TKIs, respectively. The resistance was due to the inhibition of apoptosis by aberrant activation of ERK. The ERK signal was suppressed by knockdown of integrin β1 (ITGB1). Furthermore, in patients with EGFR mutant tumors, higher SEMA7A expression in clinical samples predicted poorer response to EGFR-TKI treatment. Collectively, these data show that the SEMA7A-ITGB1 axis plays pivotal roles in EGFR-TKI resistance mediated by ERK activation and apoptosis inhibition. Moreover, our results reveal the potential utility of SEMA7A not only as a predictive biomarker, but also as a potentially novel therapeutic target in EGFR-mutant lung adenocarcinoma.
Collapse
Affiliation(s)
- Yuhei Kinehara
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Izumi Nagatomo
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shohei Koyama
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Daisuke Ito
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Satoshi Nojima
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan.,Department of Pathology, Osaka University Graduate school of Medicine, Suita, Osaka, Japan
| | - Ryota Kurebayashi
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshimitsu Nakanishi
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Yasuhiko Suga
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Yu Nishijima-Futami
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Akio Osa
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takeshi Nakatani
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Yasuhiro Kato
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Masayuki Nishide
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Yoshitomo Hayama
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Masayoshi Higashiguchi
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Osamu Morimura
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kotaro Miyake
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Sujin Kang
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan.,Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Toshiyuki Minami
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Division of Respiratory Medicine, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kota Iwahori
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takayuki Takimoto
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hyota Takamatsu
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Naoki Hosen
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | - Yasushi Shintani
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Meinoshin Okumura
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | | | | | - Shin-Ichi Nakatsuka
- Department of Pathology, Osaka International Cancer Institute, Osaka, Osaka, Japan
| | - Takashi Kijima
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Division of Respiratory Medicine, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hiroshi Kida
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,The Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| |
Collapse
|
23
|
Jiang R, Prell C, Lönnerdal B. Milk osteopontin promotes brain development by up-regulating osteopontin in the brain in early life. FASEB J 2018; 33:1681-1694. [PMID: 30199283 DOI: 10.1096/fj.201701290rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Osteopontin (OPN) is a pleiotropic protein and is abundantly present in milk. Its functions include immune modulation and cellular proliferation and differentiation. OPN is highly expressed in the brain. We investigated the effects of milk-derived OPN on brain development of mouse pups. Wild-type (WT) dams producing OPN+ milk and OPN knockout (KO) dams producing OPN- milk nursed WT pups (OPN+/+), yielding 2 pup treatment groups, OPN+ OPN+/+ and OPN- OPN+/+, for comparison. Preliminary studies supported use of this model by showing high concentrations of OPN in milk of WT dams and no OPN in milk of OPN KO dams, and production of similar amounts of milk by WT and KO dams. The ability of ingested milk OPN to enter the brain was revealed by appearance of orally gavaged [125I]-labeled and antibody-probed milk OPN in brains of pups. Brain OPN mRNA levels were similar in both nursed groups, but the brain OPN protein level was significantly lower in the OPN- OPN+/+ group at postnatal days 6 and 8. Behavior tests showed impaired memory and learning ability in OPN- OPN+/+ pups. In addition, our study revealed increased expression of myelination-related proteins and elevated proliferation and differentiation of NG-2 glia into oligodendrocytes in the brain of OPN+ OPN+/+ pups, accompanied by increased activation of ERK-1/2 and PI3K/Akt signaling. We concluded that milk OPN can play an important role in brain development and behavior in infancy by promoting myelination.-Jiang, R., Prell, C., Lönnerdal, B. Milk osteopontin promotes brain development by up-regulating osteopontin in the brain in early life.
Collapse
Affiliation(s)
- Rulan Jiang
- Department of Nutrition, University of California Davis, Davis, California, USA
| | - Christine Prell
- Dr. von Hauner Children's Hospital, Ludwig Maximilians University, Munich, Germany
| | - Bo Lönnerdal
- Department of Nutrition, University of California Davis, Davis, California, USA
| |
Collapse
|
24
|
Kang S, Nakanishi Y, Kioi Y, Okuzaki D, Kimura T, Takamatsu H, Koyama S, Nojima S, Nishide M, Hayama Y, Kinehara Y, Kato Y, Nakatani T, Shimogori T, Takagi J, Toyofuku T, Kumanogoh A. Semaphorin 6D reverse signaling controls macrophage lipid metabolism and anti-inflammatory polarization. Nat Immunol 2018; 19:561-570. [PMID: 29777213 DOI: 10.1038/s41590-018-0108-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 04/04/2018] [Indexed: 12/21/2022]
Abstract
Polarization of macrophages into pro-inflammatory or anti-inflammatory states has distinct metabolic requirements, with mechanistic target of rapamycin (mTOR) kinase signaling playing a critical role. However, it remains unclear how mTOR regulates metabolic status to promote polarization of these cells. Here we show that an mTOR-Semaphorin 6D (Sema6D)-Peroxisome proliferator receptor γ (PPARγ) axis plays critical roles in macrophage polarization. Inhibition of mTOR or loss of Sema6D blocked anti-inflammatory macrophage polarization, concomitant with severe impairments in PPARγ expression, uptake of fatty acids, and lipid metabolic reprogramming. Macrophage expression of the receptor Plexin-A4 is responsible for Sema6D-mediated anti-inflammatory polarization. We found that a tyrosine kinase, c-Abl, which associates with the cytoplasmic region of Sema6D, is required for PPARγ expression. Furthermore, Sema6D is important for generation of intestinal resident CX3CR1hi macrophages and prevents development of colitis. Collectively, these findings highlight crucial roles for Sema6D reverse signaling in macrophage polarization, coupling immunity, and metabolism via PPARγ.
Collapse
Affiliation(s)
- Sujin Kang
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan. .,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan. .,Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.
| | - Yoshimitsu Nakanishi
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Yoshiyuki Kioi
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita City, Osaka, Japan
| | - Tetsuya Kimura
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan
| | - Hyota Takamatsu
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Shohei Koyama
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Satoshi Nojima
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Pathology, Graduate School of Medicine, Osaka University, Suita City, Osaka, Japan
| | - Masayuki Nishide
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Yoshitomo Hayama
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Yuhei Kinehara
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Yasuhiro Kato
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Takeshi Nakatani
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan
| | - Tomomi Shimogori
- RIKEN Brain Science Institute, Laboratory for Molecular Mechanisms of Thalamus Development, Saitama, Japan
| | - Junichi Takagi
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita City, Osaka, Japan
| | - Toshihiko Toyofuku
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan.,Department of Immunology and Regenerative Medicine, Graduate School of Medicine, Osaka University, Suita City, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka, Japan. .,Department of Respiratory Medicine and Clinical Immunology, Graduate School of medicine, Osaka University, Suita City, Osaka, Japan.
| |
Collapse
|
25
|
Abstract
Several neuronal guidance proteins, known as semaphorin molecules, function in the immune system. This dual tissue performance has led to them being defined as "neuroimmune semaphorins". They have been shown to regulate T cell activation by serving as costimulatory molecules. Similar to classical costimulatory molecules, neuroimmune semaphorins are either constitutively or inducibly expressed on immune cells. In contrast to the classical costimulatory molecule function, the action of neuroimmune semaphorins requires the presence of two signals, the first one provided by TCR/MHC engagement, and the second one provided by B7/CD28 interaction. Thus, neuroimmune semaphorins serve as a "signal three" for immune cell activation and regulate the overall intensity of immune response. The current knowledge on their structures, multiple receptors, specific cell/tissue/organ expression, and distinct functions in different diseases are summarized and discussed in this review.
Collapse
Affiliation(s)
- Svetlana P Chapoval
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA.
- Program in Oncology at the Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.
- SemaPlex LLC, Ellicott City, MD, USA.
| |
Collapse
|
26
|
Abstract
Semaphorins are extracellular signaling proteins that are essential for the development and maintenance of many organs and tissues. The more than 20-member semaphorin protein family includes secreted, transmembrane and cell surface-attached proteins with diverse structures, each characterized by a single cysteine-rich extracellular sema domain, the defining feature of the family. Early studies revealed that semaphorins function as axon guidance molecules, but it is now understood that semaphorins are key regulators of morphology and motility in many different cell types including those that make up the nervous, cardiovascular, immune, endocrine, hepatic, renal, reproductive, respiratory and musculoskeletal systems, as well as in cancer cells. Semaphorin signaling occurs predominantly through Plexin receptors and results in changes to the cytoskeletal and adhesive machinery that regulate cellular morphology. While much remains to be learned about the mechanisms underlying the effects of semaphorins, exciting work has begun to reveal how semaphorin signaling is fine-tuned through different receptor complexes and other mechanisms to achieve specific outcomes in various cellular contexts and physiological systems. These and future studies will lead to a more complete understanding of semaphorin-mediated development and to a greater understanding of how these proteins function in human disease.
Collapse
Affiliation(s)
- Laura Taylor Alto
- Departments of Neuroscience and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jonathan R Terman
- Departments of Neuroscience and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
27
|
Wu X, Meng Y, Wang C, Yue Y, Dong C, Xiong S. Semaphorin7A aggravates coxsackievirusB3-induced viral myocarditis by increasing α1β1-integrin macrophages and subsequent enhanced inflammatory response. J Mol Cell Cardiol 2018; 114:48-57. [DOI: 10.1016/j.yjmcc.2017.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 11/01/2017] [Accepted: 11/02/2017] [Indexed: 12/22/2022]
|
28
|
Westendorp BF, Büller NV, Karpus ON, van Dop WA, Koster J, Versteeg R, Koelink PJ, Snel CY, Meisner S, Roelofs JJ, Uhmann A, Ver Loren van Themaat E, Heijmans J, Hahn H, Muncan V, Wildenberg ME, van den Brink GR. Indian Hedgehog Suppresses a Stromal Cell-Driven Intestinal Immune Response. Cell Mol Gastroenterol Hepatol 2017; 5:67-82.e1. [PMID: 29276753 PMCID: PMC5738458 DOI: 10.1016/j.jcmgh.2017.08.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 08/29/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND & AIMS Upon intestinal epithelial damage a complex wound healing response is initiated to restore epithelial integrity and defend against pathogenic invasion. Epithelium-derived Indian Hedgehog (Ihh) functions as a critical sensor in this process. Signaling occurs in a paracrine manner because the receptor for Ihh is expressed only in the mesenchyme, but the exact Hedgehog target cell has remained elusive. The aim of this study was to elucidate further the nature of this target cell in the context of intestinal inflammation. METHODS Hedgehog activity was modulated genetically in both cell type-specific and body-wide models and the resulting animals were analyzed for gene expression profiles and sensitivity for dextran sodium sulfate (DSS) colitis. To characterize the Hedgehog target cell, Gli1-CreERT2-Rosa26-ZsGreen animals were generated, which express ZsGreen in all Hedgehog-responsive cells. These cells were characterized using flow cytometry and immunofluorescence. RESULTS Loss of Indian Hedgehog from the intestinal epithelium resulted in a rapid increase in expression of inflammation-related genes, accompanied by increased influx of immune cells. Animals with epithelium-specific deletion of Ihh or lacking the Hedgehog receptor Smoothened from Hedgehog target cells were more sensitive to DSS colitis. In contrast, specific deletion of Smoothened in the myeloid compartment did not alter the response to DSS. This suggests that Hedgehog signaling does not repress intestinal immunity through an effect on myeloid cells. Indeed, we found that Hedgehog-responsive cells expressed gp38, smooth muscle actin, and desmin, indicating a fibroblastic nature. Ihh signaling inhibited expression of C-X-C motif chemokine ligand 12 (CXCL12) in fibroblasts in vitro and in vivo, thereby impairing the recruitment of immune cells. CONCLUSIONS We show that epithelium-derived Indian Hedgehog signals exclusively to fibroblasts in the intestine. Loss of Ihh leads to a rapid immune response with up-regulation of fibroblast-derived CXCL12, and migration of immune cells into the lamina propria.
Collapse
Key Words
- CXCL, C-X-C motif chemokine ligand
- CXCL12
- CXCR, C-X-C motif chemokine receptor
- DMEM, Dulbecco's modified Eagle medium
- DSS, dextran sodium sulfate
- FCS, fetal calf serum
- Gli, glioma-associated oncogene proteins
- Hedgehog
- Hhip, Hedgehog interacting protein
- IBD, inflammatory bowel disease
- IL, interleukin
- Ihh+/+, Villin-CreERT2-ZsGreen-Ihh+/+
- Ihh, Indian Hedgehog
- IhhΔ, Villin-CreERT2-ZsGreen-Ihhfl/fl
- Inflammation
- Intestine
- MPO, myeloperoxidase
- PBT, PBS/BSA/Triton
- Ptch1, Patched1
- RT-PCR, reverse-transcription polymerase chain reaction
- Smo, Smoothened
- Stroma
- α-SMA, α smooth muscle actin
Collapse
Affiliation(s)
- B. Florien Westendorp
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Nikè V.J.A. Büller
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Olga N. Karpus
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Willemijn A. van Dop
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Jan Koster
- Department of Oncogenomics and Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands
| | - Rogier Versteeg
- Department of Oncogenomics and Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands
| | - Pim J. Koelink
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Clinton Y. Snel
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Sander Meisner
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Anja Uhmann
- Institute of Human Genetics, Georg August University of Göttingen, Göttingen, Germany
| | | | - Jarom Heijmans
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Heidi Hahn
- Institute of Human Genetics, Georg August University of Göttingen, Göttingen, Germany
| | - Vanesa Muncan
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Manon E. Wildenberg
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Gijs R. van den Brink
- Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
- GlaxoSmithKline, Medicines Research Center, Stevenage, United Kingdom
- Correspondence Address correspondence to: Gijs R. van den Brink, MD, PhD, Tytgat Institute for Liver and Intestinal Research, Department of Gastroenterology and Hepatology, Academic Medical Center, Meibergdreef 69-71, 1105 BK, Amsterdam, The Netherlands. fax: (020) 5669190.Tytgat Institute for Liver and Intestinal ResearchDepartment of Gastroenterology and HepatologyAcademic Medical CenterMeibergdreef 69-71, 1105 BKAmsterdamThe Netherlands
| |
Collapse
|
29
|
Takada H, Ibaragi S, Eguchi T, Okui T, Obata K, Masui M, Morisawa A, Takabatake K, Kawai H, Yoshioka N, Hassan NMM, Shimo T, Hu GF, Nagatsuka H, Sasaki A. Semaphorin 4D promotes bone invasion in head and neck squamous cell carcinoma. Int J Oncol 2017; 51:625-632. [PMID: 28656278 DOI: 10.3892/ijo.2017.4050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 06/15/2017] [Indexed: 12/11/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) frequently invade the bones of the facial skeleton. Semaphorin 4D (Sema4D) is an axon guidance molecule produced by oligodendrocytes. Sema4D was also identified in the bone microenvironment and many cancer tissues including HNSCC. To date, however, the role of Sema4D in cancer-associated bone disease is still unknown. This is the first study to demonstrate the role of Sema4D in bone invasion of cancer. In the clinical tissue samples of bone lesion of HNSCC, Sema4D was detected at high levels, and its expression was correlated with insulin-like growth factor-I (IGF-I) expression. In vitro experiments showed that IGF-I regulates Sema4D expression and Sema4D increased proliferation, migration and invasion in HNSCC cells. Sema4D also regulated the expression of receptor activator of nuclear factor κβ ligand (RANKL) in osteoblasts, and this stimulated osteoclastgenesis. Furthermore, knockdown of Sema4D in HNSCC cells inhibited tumor growth and decreased the number of osteoclasts in a mouse xenograft model. Taken together, IGF-I-driven production of Sema4D in HNSCCs promotes osteoclastogenesis and bone invasion.
Collapse
Affiliation(s)
- Hiroyuki Takada
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | - Soichiro Ibaragi
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | - Takanori Eguchi
- Department of Dental Pharmacology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | - Tatsuo Okui
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | - Kyoichi Obata
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | - Masanori Masui
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | - Ayaka Morisawa
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | - Kiyofumi Takabatake
- Departments of Oral Pathology and Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | - Hotaka Kawai
- Departments of Oral Pathology and Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | - Norie Yoshioka
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | | | - Tsuyoshi Shimo
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | - Guo-Fu Hu
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Hitoshi Nagatsuka
- Departments of Oral Pathology and Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | - Akira Sasaki
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| |
Collapse
|
30
|
Mirakaj V, Rosenberger P. Immunomodulatory Functions of Neuronal Guidance Proteins. Trends Immunol 2017; 38:444-456. [PMID: 28438491 DOI: 10.1016/j.it.2017.03.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023]
Abstract
Neuronal guidance proteins (NGPs) were originally identified for their role during the embryonic development of the nervous system. Recent years have seen the discovery of NGP functions during immune responses. In this context, NGPs were demonstrated to control leukocyte migration and the release of cytokines during conditions of acute inflammation, such as lung injury or sepsis. However, NGPs also display potent actions in the resolution of inflammation, chronic inflammatory conditions, the development of atherosclerosis, and during ischemia followed by reperfusion. Here, we provide an overview of the current state of knowledge about the role of NGPs in the immune system and describe their immunomodulatory function.
Collapse
Affiliation(s)
- Valbona Mirakaj
- Department of Anesthesia and Intensive Care Medicine, Tübingen University Hospital, Faculty of Medicine, Eberhard-Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany.
| | - Peter Rosenberger
- Department of Anesthesia and Intensive Care Medicine, Tübingen University Hospital, Faculty of Medicine, Eberhard-Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany.
| |
Collapse
|
31
|
Jongbloets BC, Lemstra S, Schellino R, Broekhoven MH, Parkash J, Hellemons AJCGM, Mao T, Giacobini P, van Praag H, De Marchis S, Ramakers GMJ, Pasterkamp RJ. Stage-specific functions of Semaphorin7A during adult hippocampal neurogenesis rely on distinct receptors. Nat Commun 2017; 8:14666. [PMID: 28281529 PMCID: PMC5353663 DOI: 10.1038/ncomms14666] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/23/2017] [Indexed: 02/06/2023] Open
Abstract
The guidance protein Semaphorin7A (Sema7A) is required for the proper development of the immune and nervous systems. Despite strong expression in the mature brain, the role of Sema7A in the adult remains poorly defined. Here we show that Sema7A utilizes different cell surface receptors to control the proliferation and differentiation of neural progenitors in the adult hippocampal dentate gyrus (DG), one of the select regions of the mature brain where neurogenesis occurs. PlexinC1 is selectively expressed in early neural progenitors in the adult mouse DG and mediates the inhibitory effects of Sema7A on progenitor proliferation. Subsequently, during differentiation of adult-born DG granule cells, Sema7A promotes dendrite growth, complexity and spine development through β1-subunit-containing integrin receptors. Our data identify Sema7A as a key regulator of adult hippocampal neurogenesis, providing an example of how differential receptor usage spatiotemporally controls and diversifies the effects of guidance cues in the adult brain.
Collapse
Affiliation(s)
- Bart C. Jongbloets
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Suzanne Lemstra
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Roberta Schellino
- Dipartimento di Scienze della Vita e Biologia dei Sistemi and Neuroscience Institute Cavalieri Ottolenghi, University of Torino, 10100 Torino, Italy
| | - Mark H. Broekhoven
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Jyoti Parkash
- Centre for Animal Sciences, School of Basic and Applied Sciences, Central University Punjab, City Campus, Mansa Road, Bathinda 151001, India
| | - Anita J. C. G. M. Hellemons
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Tianyi Mao
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, 59045 Lille, France
- University of Lille, 59045 Lille, France
| | - Henriette van Praag
- Neuroplasticity and Behavior Unit, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Silvia De Marchis
- Dipartimento di Scienze della Vita e Biologia dei Sistemi and Neuroscience Institute Cavalieri Ottolenghi, University of Torino, 10100 Torino, Italy
| | - Geert M. J. Ramakers
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - R. Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
32
|
He M, Bian Z. Expression of hypoxia-induced semaphorin 7A correlates with the severity of inflammation and osteoclastogenesis in experimentally induced periapical lesions. Arch Oral Biol 2017; 75:114-119. [DOI: 10.1016/j.archoralbio.2016.10.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 10/19/2016] [Accepted: 10/25/2016] [Indexed: 11/29/2022]
|
33
|
Semaphorin 7A as a potential immune regulator and promising therapeutic target in rheumatoid arthritis. Arthritis Res Ther 2017; 19:10. [PMID: 28109308 PMCID: PMC5251212 DOI: 10.1186/s13075-016-1217-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/30/2016] [Indexed: 12/23/2022] Open
Abstract
Background Semaphorin 7A (Sema7A) is expressed by several different classes of lymphoid and myeloid cells and is a potent immunomodulator. We examined the role of Sema7A in modulating cellular immune responses and to provide experimental data validating the therapeutic potential of Sema7A in rheumatoid arthritis (RA). Methods Soluble Sema7A (sSema7A) levels in the serum and synovial fluid from patients with RA or osteoarthritis, as well as cytokine secretions, were analyzed with an enzyme-linked immunosorbent assay. The cell surface levels and transcripts of Sema7A were evaluated in T cells and monocytes from patients with RA. The effect of Sema7A on the functions of primary T cells isolated from the peripheral blood of healthy donors was observed. Detection of the activation of the signal mediator focal adhesion kinase was performed by Western blotting. Shedding of sSema7A was evaluated in monocytes. The introduction of anti-Sema7A antibody to mice with collagen-induced arthritis (CIA) was observed in vivo. Results Upregulation of sSema7A levels in both the serum and synovial fluid of patients with RA was correlated with disease activity markers. sSema7A markedly increased Th1/Th17 cytokine secretion and induced evident upregulation of T-bet and retinoic acid receptor-related orphan nuclear receptor γt levels in T cells. Cell surface Sema7A was cleaved by a disintegrin and metalloprotease 17 (ADAM17) in monocytes. Interleukin-6 and tumor necrosis factor-α stimulated ADAM17 secretion in synovial macrophages. Blocking of β1-integrin abrogated the Sema7A-mediated cytokine secretion. Treatment with an anti-Sema7A antibody significantly attenuated CIA. Conclusions These findings indicate that Sema7A as a potent activator of T cells and monocytes in the immune response contributes to the inflammation and progression of RA, suggesting its therapeutic potential in the treatment of RA.
Collapse
|
34
|
Esnault S, Torr EE, Bernau K, Johansson MW, Kelly EA, Sandbo N, Jarjour NN. Endogenous Semaphorin-7A Impedes Human Lung Fibroblast Differentiation. PLoS One 2017; 12:e0170207. [PMID: 28095470 PMCID: PMC5240965 DOI: 10.1371/journal.pone.0170207] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/31/2016] [Indexed: 12/20/2022] Open
Abstract
Semaphorin-7A is a glycosylphosphatidylinositol-anchored protein, initially characterized as an axon guidance protein. Semaphorin-7A also contributes to immune cell regulation and may be an essential pro-fibrotic factor when expressed by non-fibroblast cell types (exogenous). In mouse models, semaphorin-7A was shown to be important for TGF-ß1-induced pulmonary fibrosis characterized by myofibroblast accumulation and extracellular matrix deposition, but the cell-specific role of semaphorin-7A was not examined in fibroblasts. The purpose of this study is to determine semaphorin-7A expression by fibroblasts and to investigate the function of endogenously expressed semaphorin-7A in primary human lung fibroblasts (HLF). Herein, we show that non-fibrotic HLF expressed high levels of cell surface semaphorin-7A with little dependence on the percentage of serum or recombinant TGF-ß1. Semaphorin-7A siRNA strongly decreased semaphorin-7A mRNA expression and reduced cell surface semaphorin-7A. Reduction of semaphorin-7A induced increased proliferation and migration of non-fibrotic HLF. Also, independent of the presence of TGF-ß1, the decline of semaphorin-7A by siRNA was associated with increased α-smooth muscle actin production and gene expression of periostin, fibronectin, laminin, and serum response factor (SRF), indicating differentiation into a myofibroblast. Conversely, overexpression of semaphorin-7A in the NIH3T3 fibroblast cell line reduced the production of pro-fibrotic markers. The inverse association between semaphorin-7A and pro-fibrotic fibroblast markers was further analyzed using HLF from idiopathic pulmonary fibrosis (IPF) (n = 6) and non-fibrotic (n = 7) lungs. Using these 13 fibroblast lines, we observed that semaphorin-7A and periostin expression were inversely correlated. In conclusion, our study indicates that endogenous semaphorin-7A in HLF plays a role in maintaining fibroblast homeostasis by preventing up-regulation of pro-fibrotic genes. Therefore, endogenous and exogenous semaphorin-7A may have opposite effects on the fibroblast phenotype.
Collapse
Affiliation(s)
- Stephane Esnault
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, the University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
- * E-mail:
| | - Elizabeth E. Torr
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, the University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Ksenija Bernau
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, the University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Mats W. Johansson
- Department of Biomolecular Chemistry, the University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Elizabeth A. Kelly
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, the University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Nathan Sandbo
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, the University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Nizar N. Jarjour
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, the University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| |
Collapse
|
35
|
Yokota-Nakatsuma A, Ohoka Y, Takeuchi H, Song SY, Iwata M. Beta 1-integrin ligation and TLR ligation enhance GM-CSF-induced ALDH1A2 expression in dendritic cells, but differentially regulate their anti-inflammatory properties. Sci Rep 2016; 6:37914. [PMID: 27897208 PMCID: PMC5126582 DOI: 10.1038/srep37914] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 11/03/2016] [Indexed: 12/11/2022] Open
Abstract
Retinoic acid (RA)–producing CD103+ mature dendritic cells (DCs) in mesenteric lymph nodes (MLNs) play crucial roles in gut immunity. GM-CSF and RA contribute to the expression of the RA-producing enzyme ALDH1A2. However, additional signals appeared to be required for inducing ALDH1A2high mature DCs from immature DCs. We found here that TLR ligands (Ls) and immobilized E-cadherin could provide such signals in FLT3-L–generated bone marrow (BM)–derived DCs after treatment with GM-CSF and the RA receptor agonist Am80. The TLR-L-treated DCs produced proinflammatory cytokines unlike normal ALDH1A2high MLN-DCs, whereas the E-cadherin-treated DCs did not. Immobilized VCAM-1 and semaphorin 7 A exerted effects similar to those of E-cadherin. Soluble anti-integrin β1 antibodies or inhibitors of integrin signaling molecules suppressed the effects of these immobilized proteins, whereas immobilized anti-integrin β1 antibodies enhanced the GM-CSF/Am80-induced ALDH1A2 expression without inducing proinflammatory cytokines. Sequential stimulation of splenic pre-DCs with GM-CSF/Am80 and immobilized E-cadherin or anti-integrin β1 antibody also induced differentiation to mature DCs with high ALDH activity. The E-cadherin-treated BM-DCs induced gut-tropic Foxp3+ T cells and alleviated DSS–induced colitis, whereas the TLR-L-treated DCs aggravated DSS–induced colitis. The results suggest that integrin β1-mediated signals contribute to the differentiation and maturation of RA-producing anti-inflammatory DCs.
Collapse
Affiliation(s)
- Aya Yokota-Nakatsuma
- Laboratory of Immunology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido, Sanuki-shi, Kagawa, Japan.,Japan Science and Technology Agency, CREST, Chiyoda-ku, Tokyo, Japan
| | - Yoshiharu Ohoka
- Laboratory of Immunology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido, Sanuki-shi, Kagawa, Japan.,Japan Science and Technology Agency, CREST, Chiyoda-ku, Tokyo, Japan
| | - Hajime Takeuchi
- Laboratory of Immunology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido, Sanuki-shi, Kagawa, Japan.,Japan Science and Technology Agency, CREST, Chiyoda-ku, Tokyo, Japan
| | - Si-Young Song
- Japan Science and Technology Agency, CREST, Chiyoda-ku, Tokyo, Japan.,Institute of Neuroscience, Tokushima Bunri University, Shido, Sanuki-shi, Kagawa, Japan
| | - Makoto Iwata
- Laboratory of Immunology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido, Sanuki-shi, Kagawa, Japan.,Japan Science and Technology Agency, CREST, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
36
|
Peng X, Moore M, Mathur A, Zhou Y, Sun H, Gan Y, Herazo-Maya JD, Kaminski N, Hu X, Pan H, Ryu C, Osafo-Addo A, Homer RJ, Feghali-Bostwick C, Fares WH, Gulati M, Hu B, Lee CG, Elias JA, Herzog EL. Plexin C1 deficiency permits synaptotagmin 7-mediated macrophage migration and enhances mammalian lung fibrosis. FASEB J 2016; 30:4056-4070. [PMID: 27609773 DOI: 10.1096/fj.201600373r] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/15/2016] [Indexed: 12/11/2022]
Abstract
Pulmonary fibrosis is a progressive and often fatal condition that is believed to be partially orchestrated by macrophages. Mechanisms that control migration of these cells into and within the lung remain undefined. We evaluated the contributions of the semaphorin receptor, plexin C1 (PLXNC1), and the exocytic calcium sensor, synaptotagmin 7 (Syt7), in these processes. We evaluated the role of PLXNC1 in macrophage migration by using Boyden chambers and scratch tests, characterized its contribution to experimentally induced lung fibrosis in mice, and defined the mechanism for our observations. Our findings reveal that relative to control participants, patients with idiopathic pulmonary fibrosis demonstrate excessive monocyte migration and underexpression of PLXNC1 in the lungs and circulation, a finding that is recapitulated in the setting of scleroderma-related interstitial lung disease. Relative to wild type, PLXNC1-/- mouse macrophages are excessively migratory, and PLXNC1-/- mice show exacerbated collagen accumulation in response to either inhaled bleomycin or inducible lung targeted TGF-β1 overexpression. These findings are ameliorated by replacement of PLXNC1 on bone marrow-derived cells or by genetic deletion of Syt7. These data demonstrate the previously unrecognized observation that PLXNC1 deficiency permits Syt7-mediated macrophage migration and enhances mammalian lung fibrosis.-Peng, X., Moore, M., Mathur, A., Zhou, Y., Sun, H., Gan, Y., Herazo-Maya, J. D., Kaminski, N., Hu, X., Pan, H., Ryu, C., Osafo-Addo, A., Homer, R. J., Feghali-Bostwick, C., Fares, W. H., Gulati, M., Hu, B., Lee, C.-G., Elias, J. A., Herzog, E. L. Plexin C1 deficiency permits synaptotagmin 7-mediated macrophage migration and enhances mammalian lung fibrosis.
Collapse
Affiliation(s)
- Xueyan Peng
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Meagan Moore
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Aditi Mathur
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Yang Zhou
- Department of Molecular Microbiology and Immunology, Warren Alpert School of Medicine, Brown University, Providence, Rhode Island, USA
| | - Huanxing Sun
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ye Gan
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jose D Herazo-Maya
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Naftali Kaminski
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Xinyuan Hu
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Hongyi Pan
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Changwan Ryu
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Awo Osafo-Addo
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Robert J Homer
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA; and
| | - Carol Feghali-Bostwick
- Department of Medicine, Medical University of South Carolina School of Medicine, Charleston, South Carolina, USA
| | - Wassim H Fares
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Mridu Gulati
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Buqu Hu
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Chun-Geun Lee
- Department of Molecular Microbiology and Immunology, Warren Alpert School of Medicine, Brown University, Providence, Rhode Island, USA
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Warren Alpert School of Medicine, Brown University, Providence, Rhode Island, USA
| | - Erica L Herzog
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA;
| |
Collapse
|
37
|
|
38
|
A novel podocyte gene, semaphorin 3G, protects glomerular podocyte from lipopolysaccharide-induced inflammation. Sci Rep 2016; 6:25955. [PMID: 27180624 PMCID: PMC4867620 DOI: 10.1038/srep25955] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/26/2016] [Indexed: 12/12/2022] Open
Abstract
Kidney diseases including diabetic nephropathy have become huge medical problems, although its precise mechanisms are still far from understood. In order to increase our knowledge about the patho-physiology of kidney, we have previously identified >300 kidney glomerulus-enriched transcripts through large-scale sequencing and microarray profiling of the mouse glomerular transcriptome. One of the glomerulus-specific transcripts identified was semaphorin 3G (Sema3G) which belongs to the semaphorin family. The aim of this study was to analyze both the in vivo and in vitro functions of Sema3G in the kidney. Sema3G was expressed in glomerular podocytes. Although Sema3G knockout mice did not show obvious glomerular defects, ultrastructural analyses revealed partially aberrant podocyte foot processes structures. When these mice were injected with lipopolysaccharide to induce acute inflammation or streptozotocin to induce diabetes, the lack of Sema3G resulted in increased albuminuria. The lack of Sema3G in podocytes also enhanced the expression of inflammatory cytokines including chemokine ligand 2 and interleukin 6. On the other hand, the presence of Sema3G attenuated their expression through the inhibition of lipopolysaccharide-induced Toll like receptor 4 signaling. Taken together, our results surmise that the Sema3G protein is secreted by podocytes and protects podocytes from inflammatory kidney diseases and diabetic nephropathy.
Collapse
|
39
|
Kato R, Takahashi K. Gene Expression of Semaphorin 7A During Osteogenic Differentiation in Human Dental Follicle Cells. J HARD TISSUE BIOL 2016. [DOI: 10.2485/jhtb.25.263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Ryoichi Kato
- Department of Maxillofacial Surgery, Nihon University School of Dentistry at Matsudo
| | - Kosuke Takahashi
- Department of Maxillofacial Surgery, Nihon University School of Dentistry at Matsudo
- Research Institute of Oral Science
| |
Collapse
|
40
|
Gross M, Salame TM, Jung S. Guardians of the Gut - Murine Intestinal Macrophages and Dendritic Cells. Front Immunol 2015; 6:254. [PMID: 26082775 PMCID: PMC4451680 DOI: 10.3389/fimmu.2015.00254] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/07/2015] [Indexed: 12/14/2022] Open
Abstract
Intestinal mononuclear phagocytes find themselves in a unique environment, most prominently characterized by its constant exposure to commensal microbiota and food antigens. This anatomic setting has resulted in a number of specializations of the intestinal mononuclear phagocyte compartment that collectively contribute the unique steady state immune landscape of the healthy gut, including homeostatic innate lymphoid cells, B, and T cell compartments. As in other organs, macrophages and dendritic cells (DCs) orchestrate in addition the immune defense against pathogens, both in lymph nodes and mucosa-associated lymphoid tissue. Here, we will discuss origins and functions of intestinal DCs and macrophages and their respective subsets, focusing largely on the mouse and cells residing in the lamina propria.
Collapse
Affiliation(s)
- Mor Gross
- Department of Immunology, Weizmann Institute of Science , Rehovot , Israel ; Biological Services, Weizmann Institute of Science , Rehovot , Israel
| | - Tomer-Meir Salame
- Department of Immunology, Weizmann Institute of Science , Rehovot , Israel ; Biological Services, Weizmann Institute of Science , Rehovot , Israel
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science , Rehovot , Israel ; Biological Services, Weizmann Institute of Science , Rehovot , Israel
| |
Collapse
|
41
|
Viral semaphorin inhibits dendritic cell phagocytosis and migration but is not essential for gammaherpesvirus-induced lymphoproliferation in malignant catarrhal fever. J Virol 2015; 89:3630-47. [PMID: 25589653 DOI: 10.1128/jvi.03634-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
UNLABELLED Viral semaphorins are semaphorin 7A (sema7A) mimics found in pox- and herpesviruses. Among herpesviruses, semaphorins are encoded by gammaherpesviruses of the Macavirus genus only. Alcelaphine herpesvirus 1 (AlHV-1) is a macavirus that persistently infects wildebeest asymptomatically but induces malignant catarrhal fever (MCF) when transmitted to several species of susceptible ruminants and the rabbit model. MCF is caused by the activation/proliferation of latently infected T lymphocytes. Viral semaphorins have been suggested to mediate immune evasion mechanisms and/or directly alter host T cell function. We studied AlHV-sema, the viral semaphorin encoded by the A3 gene of AlHV-1. Phylogenetic analyses revealed independent acquisition of pox- and herpesvirus semaphorins, suggesting that these proteins might have distinct functions. AlHV-sema showed a predicted three-dimensional structure very similar to sema7A and conserved key residues in sema7A-plexinC1 interaction. Expression analyses revealed that AlHV-sema is a secreted 93-kDa glycoprotein expressed during the early phase of virus replication. Purified AlHV-sema was able to bind to fibroblasts and dendritic cells and induce F-actin condensation and cell retraction through a plexinC1 and Rho/cofilin-dependent mechanism. Cytoskeleton rearrangement was further associated with inhibition of phagocytosis by dendritic cells and migration to the draining lymph node. Next, we generated recombinant viruses and demonstrated that the lack of A3 did not significantly affect virus growth in vitro and did not impair MCF induction and associated lymphoproliferative lesions. In conclusion, AlHV-sema has immune evasion functions through mechanisms similar to poxvirus semaphorin but is not directly involved in host T cell activation during MCF. IMPORTANCE Whereas most poxviruses encode viral semaphorins, semaphorin-like genes have only been identified in few gammaherpesviruses belonging to the Macavirus genus. Alcelaphine herpesvirus 1 (AlHV-1) is a macavirus carried asymptomatically by wildebeest but induces a latency-associated lymphoproliferative disease of T lymphocytes in various ruminant species, namely, malignant catarrhal fever (MCF). Viral semaphorins have been hypothesized to have immune evasion functions and/or be involved in activating latently infected T cells. We present evidence that the viral semaphorin AlHV-sema inhibits dendritic cell phagocytosis and migration to the draining lymph node, both being indispensable mechanisms for protective antiviral responses. Next, we engineered recombinant viruses unable to express AlHV-sema and demonstrated that this protein is dispensable for the induction of MCF. In conclusion, this study suggests that herpesvirus and poxvirus semaphorins have independently evolved similar functions to thwart the immune system of the host while AlHV-sema is not directly involved in MCF-associated T-cell activation.
Collapse
|
42
|
Ma B, Herzog EL, Lee CG, Peng X, Lee CM, Chen X, Rockwell S, Koo JS, Kluger H, Herbst RS, Sznol M, Elias JA. Role of chitinase 3-like-1 and semaphorin 7a in pulmonary melanoma metastasis. Cancer Res 2014; 75:487-96. [PMID: 25511377 DOI: 10.1158/0008-5472.can-13-3339] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The prototypic chitinase-like protein Chi3l1 is induced in cancers and portends a poor prognosis, but whether it contributes to cancer progression is unknown. To address this gap in knowledge, we investigated the production of Chi3l1 in melanoma lung metastases. We found that Chi3l1 was induced during pulmonary melanoma metastasis and that this induction was regulated by the semaphorin Sema7a, interacting in stimulatory or inhibitory ways with its β1 integrin or Plexin C1 receptors, respectively. In mouse strains with genetic deletions of Chi3l1 or Sema7a, there was a significant reduction in pulmonary metastasis. Notably, antiserum raised against Chi3l1 or Sema7a phenocopied the reduction produced by genetic deletions. Melanoma lung metastasis was also decreased in the absence of IL13Rα2, a recently identified receptor for Chi3l1, consistent with a key role for Chi3l1 in melanoma spread. We confirmed roles for Sema7a and Chi3l1 in pulmonary metastasis of EMT6 breast cancer cells. Taken together, our studies establish a novel pathway through which Sem7a and its receptors regulate Chi3l1, revealing a host axis involving IL13Rα2 that plays a critical role in generating a pulmonary microenvironment that is critical to license metastasis.
Collapse
Affiliation(s)
- Bing Ma
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Erica L Herzog
- Section of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Xueyan Peng
- Section of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Xiaosong Chen
- Section of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Sara Rockwell
- Department of Therapeutic Radiology, Yale Cancer Center, New Haven, Connecticut
| | - Ja Seok Koo
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Harriet Kluger
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Roy S Herbst
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Mario Sznol
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island. Section of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
43
|
Takata K, Tomita T, Okuno T, Kinoshita M, Koda T, Honorat JA, Takei M, Hagihara K, Sugimoto T, Mochizuki H, Sakoda S, Nakatsuji Y. Dietary Yeasts Reduce Inflammation in Central Nerve System via Microflora. Ann Clin Transl Neurol 2014; 2:56-66. [PMID: 25642435 PMCID: PMC4301675 DOI: 10.1002/acn3.153] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/28/2014] [Accepted: 11/02/2014] [Indexed: 12/20/2022] Open
Abstract
Objectives The intestinal microflora affects the pathogenesis of several autoimmune diseases by influencing immune system function. Some bacteria, such as lactic acid bacteria, have been reported to have beneficial effects on immune function. However, little is known about the effects of yeasts. Here, we aimed to investigate the effects of various dietary yeasts contained in fermented foods on experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), and to elucidate the mechanisms underlying these effects. Methods The effects of eight yeasts selected from 18 types of yeasts contained in fermented foods were examined using an EAE model. Of these, Candida kefyr was investigated by analyzing the intestinal microflora and its effects on intestinal and systemic immune states. Results Administration of C. kefyr ameliorated the severity of EAE. Reduced numbers of Th17 cells, suppressed interleukin (IL)-6 production by intestinal explants, and increased Tregs and CD103-positive regulatory dendritic cells in mesenteric lymph nodes (MLNs) were observed. Analysis of 16s-rDNA from feces of C. kefyr-treated mice demonstrated increased Lactobacillales and decreased Bacteroides compared to control flora. Transfer of intestinal microbiota also resulted in decreased Bacteroides and ameliorated symptoms of EAE. Thus, oral administration of C. kefyr ameliorated EAE by altering the microflora, accompanied by increased Tregs and CD103-positive regulatory dendritic cells in MLNs and decreased Th17 cells in the intestinal lamina propria. Interpretation Oral ingestion of C. kefyr may have beneficial effects on MS by modifying microflora. In addition, our findings also suggested the potential health benefits of dietary yeasts.
Collapse
Affiliation(s)
- Kazushiro Takata
- Department of Neurology, Osaka University Graduate School of Medicine D4 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Takayuki Tomita
- Discovery Research Laboratories, Kyorin Pharmaceutical Co., ltd. 2399-1, Nogi, Nogi-machi, Shimotsuga-gun, Tochigi, 329-0114, Japan
| | - Tatsusada Okuno
- Department of Neurology, Osaka University Graduate School of Medicine D4 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Makoto Kinoshita
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Osaka University Graduate School of Medicine C6 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Toru Koda
- Department of Neurology, Osaka University Graduate School of Medicine D4 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Josephe A Honorat
- Department of Neurology, Osaka University Graduate School of Medicine D4 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Masaya Takei
- Discovery Research Laboratories, Kyorin Pharmaceutical Co., ltd. 2399-1, Nogi, Nogi-machi, Shimotsuga-gun, Tochigi, 329-0114, Japan
| | - Kouichiro Hagihara
- Discovery Research Laboratories, Kyorin Pharmaceutical Co., ltd. 2399-1, Nogi, Nogi-machi, Shimotsuga-gun, Tochigi, 329-0114, Japan
| | - Tomoyuki Sugimoto
- Research Division, Hirosaki University Graduate School of Science and Technology 3-bunkyocho, Hirosaki, Aomori, 036-8560, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine D4 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Saburo Sakoda
- Department of Neurology, National Hospital Organization Toneyama 5-5-1 Toneyama, Toyonaka, Osaka, 560-8552, Japan
| | - Yuji Nakatsuji
- Department of Neurology, Osaka University Graduate School of Medicine D4 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
44
|
|
45
|
Garcia-Areas R, Libreros S, Iragavarapu-Charyulu V. Semaphorin7A: branching beyond axonal guidance and into immunity. Immunol Res 2014; 57:81-5. [PMID: 24222277 DOI: 10.1007/s12026-013-8460-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Semaphorins are a family of proteins that were originally described for their role in axonal guidance. Studies now show that semaphorins encompass many physiological functions outside of the nervous system, including immune responses. Semaphorin7A (SEMA7A) belongs to the "immune" semaphorin group and has been shown to play a crucial role in regulating immune responses. In this review, we discuss the structure and function of SEMA7A as well as its role in innate and adaptive immunity [corrected].We further describe SEMA7A's involvement in inflammatory disease and its emergent role in cancer.
Collapse
Affiliation(s)
- Ramon Garcia-Areas
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Road, Boca Raton, FL, 33431-0991, USA
| | | | | |
Collapse
|
46
|
Than BLN, Goos JACM, Sarver AL, O’Sullivan MG, Rod A, Starr TK, Fijneman RJA, Meijer GA, Zhao L, Zhang Y, Largaespada DA, Scott PM, Cormier RT. The role of KCNQ1 in mouse and human gastrointestinal cancers. Oncogene 2014; 33:3861-3868. [PMID: 23975432 PMCID: PMC3935979 DOI: 10.1038/onc.2013.350] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 06/07/2013] [Accepted: 07/04/2013] [Indexed: 12/19/2022]
Abstract
Kcnq1, which encodes for the pore-forming α-subunit of a voltage-gated potassium channel, was identified as a gastrointestinal (GI) tract cancer susceptibility gene in multiple Sleeping Beauty DNA transposon-based forward genetic screens in mice. To confirm that Kcnq1 has a functional role in GI tract cancer, we created Apc(Min) mice that carried a targeted deletion mutation in Kcnq1. Results demonstrated that Kcnq1 is a tumor suppressor gene as Kcnq1 mutant mice developed significantly more intestinal tumors, especially in the proximal small intestine and colon, and some of these tumors progressed to become aggressive adenocarcinomas. Gross tissue abnormalities were also observed in the rectum, pancreas and stomach. Colon organoid formation was significantly increased in organoids created from Kcnq1 mutant mice compared with wild-type littermate controls, suggesting a role for Kcnq1 in the regulation of the intestinal crypt stem cell compartment. To identify gene expression changes due to loss of Kcnq1, we carried out microarray studies in the colon and proximal small intestine. We identified altered genes involved in innate immune responses, goblet and Paneth cell function, ion channels, intestinal stem cells, epidermal growth factor receptor and other growth regulatory signaling pathways. We also found genes implicated in inflammation and in cellular detoxification. Pathway analysis using Ingenuity Pathway Analysis and Gene Set Enrichment Analysis confirmed the importance of these gene clusters and further identified significant overlap with genes regulated by MUC2 and CFTR, two important regulators of intestinal homeostasis. To investigate the role of KCNQ1 in human colorectal cancer (CRC), we measured protein levels of KCNQ1 by immunohistochemistry in tissue microarrays containing samples from CRC patients with liver metastases who had undergone hepatic resection. Results showed that low expression of KCNQ1 expression was significantly associated with poor overall survival.
Collapse
Affiliation(s)
- B. L. N Than
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812
- Toxicology Graduate Program, University of Minnesota, Duluth, MN 55812
| | - J. A. C. M. Goos
- Dept. of Pathology, VU University Medical Center, 1081 HV, Amsterdam, Netherlands
| | - A. L. Sarver
- Department of Biostatistics and Informatics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
| | - M. G. O’Sullivan
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - A. Rod
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812
| | - T. K. Starr
- Department of Genetics, Cell Biology & Development, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
- Department of Obstetrics, Gynecology & Women’s Health, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455
| | - R. J. A. Fijneman
- Dept. of Pathology, VU University Medical Center, 1081 HV, Amsterdam, Netherlands
| | - G. A. Meijer
- Dept. of Pathology, VU University Medical Center, 1081 HV, Amsterdam, Netherlands
| | - L Zhao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812
| | - Y Zhang
- University of Minnesota Supercomputing Institute, Minneapolis, MN 55455
| | - D. A. Largaespada
- Department of Genetics, Cell Biology & Development, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
| | - P. M. Scott
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812
| | - R. T. Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812
| |
Collapse
|
47
|
Granja T, Köhler D, Mirakaj V, Nelson E, König K, Rosenberger P. Crucial role of Plexin C1 for pulmonary inflammation and survival during lung injury. Mucosal Immunol 2014; 7:879-91. [PMID: 24345803 DOI: 10.1038/mi.2013.104] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/25/2013] [Accepted: 10/26/2013] [Indexed: 02/04/2023]
Abstract
Acute pulmonary inflammation during lung injury is initiated by the migration of neutrophils into the alveolar space. The severity of these inflammatory changes within the pulmonary tissue determines the severity of lung injury and ultimately patient outcome. Recent work has demonstrated that the guidance protein Semaphorin 7A propagates the infiltration of neutrophils into an hypoxic tissue site, yet the role of its target receptor Plexin C1 (PLXNC1) during lung injury is to date unknown. We demonstrate here that PLXNC1(+) neutrophils are present within the alveolar space and that PLXNC1 is induced in vitro and in vivo during lung injury. In a model of high-pressure ventilation PLXNC1(-/-) animals show decreased signs of alveolar inflammation and improved survival compared with wild-type controls. Studies employing chimeric animals identified the hematopoietic expression of PLXNC1 to be of crucial importance for the observed results. Functional inhibition of PLXNC1 resulted in improved survival and ameliorated the signs of inflammation within the lung. Furthermore, the injection of a peptide binding to PLXNC1 resulted in improved survival and attenuated pulmonary inflammation. As such we demonstrate here, that previously unknown PLXNC1 holds significant importance for degree of pulmonary inflammation and determines outcome during experimental lung injury.
Collapse
Affiliation(s)
- T Granja
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital; Eberhard-Karls University, Tübingen, Germany
| | - D Köhler
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital; Eberhard-Karls University, Tübingen, Germany
| | - V Mirakaj
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital; Eberhard-Karls University, Tübingen, Germany
| | - E Nelson
- Department of Anesthesiology and Perioperative Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - K König
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital; Eberhard-Karls University, Tübingen, Germany
| | - P Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital; Eberhard-Karls University, Tübingen, Germany
| |
Collapse
|
48
|
König K, Marth L, Roissant J, Granja T, Jennewein C, Devanathan V, Schneider M, Köhler D, Zarbock A, Rosenberger P. The plexin C1 receptor promotes acute inflammation. Eur J Immunol 2014; 44:2648-58. [DOI: 10.1002/eji.201343968] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 05/02/2014] [Accepted: 05/27/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Klemens König
- Department of Anaesthesiology and Intensive Care Medicine; Tübingen University Hospital, Eberhard-Karls University Tübingen; Tübingen Germany
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy; University Hospital Frankfurt am Main; Frankfurt Germany
| | - Linda Marth
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy; University Hospital Frankfurt am Main; Frankfurt Germany
| | - Jan Roissant
- Department of Anesthesiology, Intensive Care and Pain Medicine; University of Münster, Max-Planck Institute for Molecular Biomedicine; Münster Germany
| | - Tiago Granja
- Department of Anaesthesiology and Intensive Care Medicine; Tübingen University Hospital, Eberhard-Karls University Tübingen; Tübingen Germany
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy; University Hospital Frankfurt am Main; Frankfurt Germany
| | - Carla Jennewein
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy; University Hospital Frankfurt am Main; Frankfurt Germany
| | - Vasudharani Devanathan
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology; Tübingen University Hospital, University Tübingen; Tübingen Germany
| | - Mariella Schneider
- Department of Anaesthesiology and Intensive Care Medicine; Tübingen University Hospital, Eberhard-Karls University Tübingen; Tübingen Germany
| | - David Köhler
- Department of Anaesthesiology and Intensive Care Medicine; Tübingen University Hospital, Eberhard-Karls University Tübingen; Tübingen Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine; University of Münster, Max-Planck Institute for Molecular Biomedicine; Münster Germany
| | - Peter Rosenberger
- Department of Anaesthesiology and Intensive Care Medicine; Tübingen University Hospital, Eberhard-Karls University Tübingen; Tübingen Germany
| |
Collapse
|
49
|
Fujita N, Oritani K, Ichii M, Yokota T, Saitoh N, Okuzaki D, Sekine Y, Kon S, Muromoto R, Saitoh K, Yoshimura A, Matsuda T, Kanakura Y. Signal-transducing adaptor protein-2 regulates macrophage migration into inflammatory sites during dextran sodium sulfate induced colitis. Eur J Immunol 2014; 44:1791-801. [PMID: 24733425 DOI: 10.1002/eji.201344239] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/16/2014] [Accepted: 03/13/2014] [Indexed: 12/31/2022]
Abstract
Signal-transducing adaptor protein-2 (STAP-2) was cloned as a c-fms/M-CSF receptor interacting protein. STAP-2 is an adaptor protein carrying pleckstrin homology and Src homology 2 like domains, as well as a YXXQ motif. STAP-2 has been indicated to have an ability to bind and modulate a variety of signaling and transcriptional molecules. Especially, our previous in vitro studies showed that STAP-2 is crucial for immune and/or inflammatory responses. Here, we have investigated the role of STAP-2 in intestinal inflammation in vivo. The disruption of STAP-2 attenuates dextran sodium sulfate induced colitis via inhibition of macrophage recruitment. To study whether hematopoietic or epithelial cell derived STAP-2 is required for this phenomenon, we generated BM chimeric mice. STAP-2-deficient macrophages impair the ability of CXCL12-induced migration. Intriguingly, STAP-2 also regulates production of proinflammatory chemokines and cytokines such as CXCL1 and TNF-α from intestinal epithelial cells. Therefore, STAP-2 has a potential to regulate plural molecular events during pathological inflammatory responses. Furthermore, our findings not only indicate that STAP-2 is important in regulating intestinal inflammation, but also provide new insights toward the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Natsuko Fujita
- Department of Hematology and Oncology, Osaka University Graduate School of medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Peterson LW, Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol 2014; 14:141-53. [PMID: 24566914 DOI: 10.1038/nri3608] [Citation(s) in RCA: 2062] [Impact Index Per Article: 187.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The abundance of innate and adaptive immune cells that reside together with trillions of beneficial commensal microorganisms in the mammalian gastrointestinal tract requires barrier and regulatory mechanisms that conserve host-microbial interactions and tissue homeostasis. This homeostasis depends on the diverse functions of intestinal epithelial cells (IECs), which include the physical segregation of commensal bacteria and the integration of microbial signals. Hence, IECs are crucial mediators of intestinal homeostasis that enable the establishment of an immunological environment permissive to colonization by commensal bacteria. In this Review, we provide a comprehensive overview of how IECs maintain host-commensal microbial relationships and immune cell homeostasis in the intestine.
Collapse
Affiliation(s)
- Lance W Peterson
- Department of Microbiology and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania
| | - David Artis
- 1] Department of Microbiology and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania. [2] Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|