51
|
Sylvester FA. Effects of Digestive Diseases on Bone Metabolism. PEDIATRIC GASTROINTESTINAL AND LIVER DISEASE 2021:1023-1031.e7. [DOI: 10.1016/b978-0-323-67293-1.00091-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
52
|
Maier I, Liu J, Ruegger PM, Deutschmann J, Patsch JM, Helbich TH, Borneman J, Schiestl RH. Intestinal bacterial indicator phylotypes associate with impaired DNA double-stranded break sensors but augmented skeletal bone micro-structure. Carcinogenesis 2020; 41:483-489. [PMID: 31840161 DOI: 10.1093/carcin/bgz204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 12/07/2019] [Accepted: 12/13/2019] [Indexed: 12/17/2022] Open
Abstract
Intestinal microbiota are considered a sensor for molecular pathways, which orchestrate energy balance, immune responses, and cell regeneration. We previously reported that microbiota restriction promoted higher levels of systemic radiation-induced genotoxicity, proliferative lymphocyte activation, and apoptotic polarization of metabolic pathways. Restricted intestinal microbiota (RM) that harbors increased abundance of Lactobacillus johnsonii (LBJ) has been investigated for bacterial communities that correlated radiation-induced genotoxicity. Indicator phylotypes were more abundant in RM mice and increased in prevalence after whole body irradiation in conventional microbiota (CM) mice, while none of the same ten most abundant phylotypes were different in abundance between CM mice before and after heavy ion irradiation. Muribaculum intestinale was detected highest in female small intestines in RM mice, which were lacking Ureaplasma felinum compared with males, and thus these bacteria could be contributing to the differential amounts of radiation-induced systemic genotoxicity between the CM and RM groups. Helicobacter rodentium and M.intestinale were found in colons in the radiation-resistant CM phenotype. While the expression of interferon-γ was elevated in the small intestine, and lower in blood in CM mice, high-linear energy transfer radiation reduced transforming growth factor-β with peripheral interleukin (IL)-17 in RM mice, particularly in females. We found that female RM mice showed improved micro-architectural bone structure and anti-inflammatory radiation response compared with CM mice at a delayed phase 6 weeks postexposure to particle radiation. However, microbiota restriction reduced inflammatory markers of tumor necrosis factor in marrow, when IL-17 was reduced by intraperitoneal injection of IL-17 neutralizing antibody.
Collapse
Affiliation(s)
- Irene Maier
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA
| | - Jared Liu
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA
| | - Paul M Ruegger
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| | - Julia Deutschmann
- Department for Radiologic Technology, University of Applied Sciences Wiener Neustadt for Business and Engineering Ltd., Lower Austria, Austria
| | - Janina M Patsch
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel,Vienna, Austria
| | - Thomas H Helbich
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel,Vienna, Austria
| | - James Borneman
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| | - Robert H Schiestl
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA.,Department of Pathology, University of California, Los Angeles, CA, USA
| |
Collapse
|
53
|
Dell'Aquila E, Armento G, Iuliani M, Simonetti S, D'Onofrio L, Zeppola T, Madaudo C, Russano M, Citarella F, Ribelli G, Pantano F, Vincenzi B, Tonini G, Santini D. Denosumab for cancer-related bone loss. Expert Opin Biol Ther 2020; 20:1261-1274. [PMID: 32835531 DOI: 10.1080/14712598.2020.1814731] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Prolonged use of anti-cancer treatments in breast and prostate tumors alters physiological bone turnover leading to adverse skeletal related events, such as osteoporosis, loss of bone mass, and increased risk of fractures. These complications known as cancer treatment-induced bone loss (CTIBL) should be managed with bone targeting agents such as the bisphosphonates and denosumab. The latter is a monoclonal antibody against the receptor activator of nuclear factor-kB ligand (RANKL) that suppresses osteoclasts function and survival increasing bone mass. AREAS COVERED This review will focus on the mechanisms associated with bone loss induced by cancer treatments and the most recent evidence about the use of denosumab as preventive and therapeutic strategy to protect bone health. Moreover, we will discuss several key aspects regarding the clinical practical use of denosumab to optimize the management of CTLIB in breast and prostate cancer. EXPERT OPINION Denosumab treatment strongly prevents cancer therapies-related skeletal issues in breast and prostate cancer with a good safety profile. Adjuvant six-monthly denosumab delays the time to first fracture onset in early stage breast cancer patients with normal or altered bone mineral density (BMD). Similarly, denosumab treatment is able to prevent fractures and BMD loss in nonmetastatic prostate cancer patients.
Collapse
Affiliation(s)
| | - Grazia Armento
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Michele Iuliani
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Sonia Simonetti
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Loretta D'Onofrio
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Tea Zeppola
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Cristina Madaudo
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Marco Russano
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Fabrizio Citarella
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Giulia Ribelli
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Francesco Pantano
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Bruno Vincenzi
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Giuseppe Tonini
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| | - Daniele Santini
- Medical Oncology Department, Campus Bio-Medico University of Rome , Rome, Itlay
| |
Collapse
|
54
|
Uehara IA, Soldi LR, Silva MJB. Current perspectives of osteoclastogenesis through estrogen modulated immune cell cytokines. Life Sci 2020; 256:117921. [DOI: 10.1016/j.lfs.2020.117921] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
|
55
|
Madel MB, Ibáñez L, Ciucci T, Halper J, Rouleau M, Boutin A, Hue C, Duroux-Richard I, Apparailly F, Garchon HJ, Wakkach A, Blin-Wakkach C. Dissecting the phenotypic and functional heterogeneity of mouse inflammatory osteoclasts by the expression of Cx3cr1. eLife 2020; 9:54493. [PMID: 32400390 PMCID: PMC7220377 DOI: 10.7554/elife.54493] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/26/2020] [Indexed: 12/19/2022] Open
Abstract
Bone destruction relies on interactions between bone and immune cells. Bone-resorbing osteoclasts (OCLs) were recently identified as innate immune cells activating T cells toward tolerance or inflammation. Thus, pathological bone destruction not only relies on increased osteoclast differentiation, but also on the presence of inflammatory OCLs (i-OCLs), part of which express Cx3cr1. Here, we investigated the contribution of mouse Cx3cr1+ and Cx3cr1neg i-OCLs to bone loss. We showed that Cx3cr1+ and Cx3cr1neg i-OCLs differ considerably in transcriptional and functional aspects. Cx3cr1neg i-OCLs have a high ability to resorb bone and activate inflammatory CD4+ T cells. Although Cx3cr1+ i-OCLs are associated with inflammation, they resorb less and have in vitro an immune-suppressive effect on Cx3cr1neg i-OCLs, mediated by PD-L1. Our results provide new insights into i-OCL heterogeneity. They also reveal that different i-OCL subsets may interact to regulate inflammation. This contributes to a better understanding and prevention of inflammatory bone destruction.
Collapse
Affiliation(s)
- Maria-Bernadette Madel
- Laboratoire de PhysioMédecine Moléculaire, CNRS, Nice, France.,Université Côte d'Azur, Nice, France
| | - Lidia Ibáñez
- Department of Pharmacy, Cardenal Herrera-CEU University, Valencia, Spain
| | - Thomas Ciucci
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Julia Halper
- Laboratoire de PhysioMédecine Moléculaire, CNRS, Nice, France.,Université Côte d'Azur, Nice, France
| | - Matthieu Rouleau
- Laboratoire de PhysioMédecine Moléculaire, CNRS, Nice, France.,Université Côte d'Azur, Nice, France
| | - Antoine Boutin
- Laboratoire de PhysioMédecine Moléculaire, CNRS, Nice, France.,Université Côte d'Azur, Nice, France
| | - Christophe Hue
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammation, Montigny-Le-Bretonneux, France
| | | | | | - Henri-Jean Garchon
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammation, Montigny-Le-Bretonneux, France.,Genetics division, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Abdelilah Wakkach
- Laboratoire de PhysioMédecine Moléculaire, CNRS, Nice, France.,Université Côte d'Azur, Nice, France
| | - Claudine Blin-Wakkach
- Laboratoire de PhysioMédecine Moléculaire, CNRS, Nice, France.,Université Côte d'Azur, Nice, France
| |
Collapse
|
56
|
Yao J, Gao R, Luo M, Li D, Guo L, Yu Z, Xiong F, Wei C, Wu B, Xu Z, Zhang D, Wang J, Wang L. miR-802 participates in the inflammatory process of inflammatory bowel disease by suppressing SOCS5. Biosci Rep 2020; 40:BSR20192257. [PMID: 32211804 PMCID: PMC7138906 DOI: 10.1042/bsr20192257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 01/01/2020] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
The present study aims to reveal the detailed molecular mechanism of microRNA (miR)-802 in the progression of inflammatory bowel disease (IBD). IBD tissues were obtained from IBD patients, followed by CD4+ cells isolation. Then, qRT-PCR and ELISA were used to detect the expression of miR-802, suppressor of cytokine signaling 5 (SOCS5), interleukin (IL)-17A and tumor necrosis factor (TNF)-α. Transfection of miR-802 mimics and miR-802 inhibitor in CD4+ cells was detected by Western blot. TargetScan and luciferase reporter assay were used to detect the relationship between SOCS5 and miR-802. Finally, colitis mice model was established to verify whether miR-802 inhibitor was involved in the protective effect of colonic mucosa. The miR-802 was highly expressed in inflamed mucosa and PBMC cells of IBD. The highest expression of miR-802 was observed in CD4+ T cells based on different immune cell subsets analysis. SOCS5 was the target gene of miR-802. The mice model experiments showed that blockade of miR-802 could alleviate mice colitis. Our study suggests that up-regulation of miR-802 plays an important role in inflammatory process of IBD via targeting SOCS5. Moreover, the differentiation of Th17 and secretion of TNF-α in IBD could be stimulated by miR-802.
Collapse
Affiliation(s)
- Jun Yao
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Ruoyu Gao
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Minghan Luo
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Defeng Li
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Liliangzi Guo
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Zichao Yu
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Feng Xiong
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Cheng Wei
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Benhua Wu
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Zhenglei Xu
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Dingguo Zhang
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Jianyao Wang
- Department of General Surgery, Shenzhen Children’s Hospital, No. 7019, Yitian Road Road, Shenzhen City, Guangdong Province 518026, China
| | - Lisheng Wang
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People’s Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| |
Collapse
|
57
|
Abstract
Gut microbiome refers to the microbes that live in human digestive tract and are symbiotic with the human body. They participate in the regulation of various physiological and pathological processes of the human body and are associated with various diseases. The pathological process of osteoporosis is affected by gut microbes. The molecular mechanisms of osteoporosis mainly include: 1) Intestinal barrier and nutrient absorption (involving SCFAs). 2) Immunoregulation (Th-17 and T-reg cells balance). 3) Regulation of intestinal-brain axis (involving 5-HT). Gut microbes can increase bone mass and improve osteoporosis by inhibiting osteoclast proliferation and differentiation, inducing apoptosis, reducing bone resorption, or promoting osteoblast proliferation and maturation. However, the therapeutic effect of gut microbes on osteoporosis remains to be further proven. At present, some of the findings on the impact of gut microbes on osteoporosis has been applied in clinical, including early diagnosis and intervention of osteoporosis and adjuvant therapy. In this article, we reviewed the molecular mechanisms underlying the regulatory effect of gut microbes on osteoporosis and the clinical practice of using gut microbes to improve bone health.
Collapse
Affiliation(s)
- Kai Ding
- 1Department of Trauma Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China.,2Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, China.,3Department of Trauma Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Fei Hua
- 1Department of Trauma Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China.,2Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, China.,3Department of Trauma Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wenge Ding
- 1Department of Trauma Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China.,2Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, China.,3Department of Trauma Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
58
|
Hathaway-Schrader JD, Poulides NA, Carson MD, Kirkpatrick JE, Warner AJ, Swanson BA, Taylor EV, Chew ME, Reddy SV, Liu B, Westwater C, Novince CM. Specific Commensal Bacterium Critically Regulates Gut Microbiota Osteoimmunomodulatory Actions During Normal Postpubertal Skeletal Growth and Maturation. JBMR Plus 2020; 4:e10338. [PMID: 32161843 PMCID: PMC7059828 DOI: 10.1002/jbm4.10338] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 12/19/2022] Open
Abstract
The commensal gut microbiota critically regulates immunomodulatory processes that influence normal skeletal growth and maturation. However, the influence of specific microbes on commensal gut microbiota osteoimmunoregulatory actions is unknown. We have shown previously that the commensal gut microbiota enhances TH17/IL17A immune response effects in marrow and liver that have procatabolic/antianabolic actions in the skeleton. Segmented filamentous bacteria (SFB), a specific commensal gut bacterium within phylum Firmicutes, potently induces TH17/IL17A‐mediated immunity. The study purpose was to delineate the influence of SFB on commensal gut microbiota immunomodulatory actions regulating normal postpubertal skeletal development. Two murine models were utilized: SFB‐monoassociated mice versus germ‐free (GF) mice and specific‐pathogen‐free (SPF) mice +/− SFB. SFB colonization was validated by 16S rDNA analysis, and SFB‐induced TH17/IL17A immunity was confirmed by upregulation of Il17a in ileum and IL17A in serum. SFB‐colonized mice had an osteopenic trabecular bone phenotype, which was attributed to SFB actions suppressing osteoblastogenesis and enhancing osteoclastogenesis. Intriguingly, SFB‐colonized mice had increased expression of proinflammatory chemokines and acute‐phase reactants in the liver. Lipocalin‐2 (LCN2), an acute‐phase reactant and antimicrobial peptide, was substantially elevated in the liver and serum of SFB‐colonized mice, which supports the notion that SFB regulation of commensal gut microbiota osteoimmunomodulatory actions are mediated in part through a gut–liver–bone axis. Proinflammatory TH17 and TH1 cells were increased in liver‐draining lymph nodes of SFB‐colonized mice, which further substantiates that SFB osteoimmune‐response effects may be mediated through the liver. SFB‐induction of Il17a in the gut and Lcn2 in the liver resulted in increased circulating levels of IL17A and LCN2. Recognizing that IL17A and LCN2 support osteoclastogenesis/suppress osteoblastogenesis, SFB actions impairing postpubertal skeletal development appear to be mediated through immunomodulatory effects in both the gut and liver. This research reveals that specific microbes critically impact commensal gut microbiota immunomodulatory actions regulating normal postpubertal skeletal growth and maturation. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jessica D Hathaway-Schrader
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Nicole A Poulides
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Matthew D Carson
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Joy E Kirkpatrick
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Drug Discovery & Biomedical Sciences College of Pharmacy, Medical University of South Carolina Charleston SC USA
| | - Amy J Warner
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Brooks A Swanson
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Eliza V Taylor
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA
| | - Michael E Chew
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA
| | - Sakamuri V Reddy
- Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Bei Liu
- Department of Microbiology and Immunology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Caroline Westwater
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Microbiology and Immunology College of Medicine, Medical University of South Carolina Charleston SC USA
| | - Chad M Novince
- Department of Oral Health Sciences College of Dental Medicine, Medical University of South Carolina Charleston SC USA.,Department of Pediatrics-Division of Endocrinology College of Medicine, Medical University of South Carolina Charleston SC USA
| |
Collapse
|
59
|
Experimental arthritis and Porphyromonas gingivalis administration synergistically decrease bone regeneration in femoral cortical defects. Sci Rep 2019; 9:20031. [PMID: 31882624 PMCID: PMC6934576 DOI: 10.1038/s41598-019-56265-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 12/09/2019] [Indexed: 12/17/2022] Open
Abstract
Porphyromonas gingivalis infection can lead to periodontitis and dysbiosis, which are known risk factors for rheumatoid arthritis (RA). We investigated whether P. gingivalis administration affected bone regeneration in mice with or without arthritis. We administered P. gingivalis to male DBA/1 J mice that were or were not sensitised to type II collagen-induced arthritis (CIA). All mice underwent drilling of bilateral femurs. We histologically evaluated new bone regeneration (bone volume of the defect [BVd]/tissue volume of the defect [TVd]) using micro-computed tomography (micro-CT), osteoclast number/bone area, and active osteoblast surface/bone surface (Ob.S/BS). We measured serum cytokine levels and bone mineral density of the proximal tibia using micro-CT. CIA resulted in significantly reduced bone regeneration (BVd/TVd) at all time-points, whereas P. gingivalis administration showed similar effects at 2 weeks postoperatively. CIA resulted in higher osteoclast number/bone area and lower Ob.S/BS at 2 and 3 weeks postoperatively, respectively. However, P. gingivalis administration resulted in lower Ob.S/BS only at 2 weeks postoperatively. During later-stage bone regeneration, CIA and P. gingivalis administration synergistically decreased BVd/TVd, increased serum tumour necrosis factor-α, and resulted in the lowest bone mineral density. Therefore, RA and dysbiosis could be risk factors for prolonged fracture healing.
Collapse
|
60
|
Mechanisms Underlying Bone Loss Associated with Gut Inflammation. Int J Mol Sci 2019; 20:ijms20246323. [PMID: 31847438 PMCID: PMC6940820 DOI: 10.3390/ijms20246323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/29/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
Patients with gastrointestinal diseases frequently suffer from skeletal abnormality, characterized by reduced bone mineral density, increased fracture risk, and/or joint inflammation. This pathological process is characterized by altered immune cell activity and elevated inflammatory cytokines in the bone marrow microenvironment due to disrupted gut immune response. Gastrointestinal disease is recognized as an immune malfunction driven by multiple factors, including cytokines and signaling molecules. However, the mechanism by which intestinal inflammation magnified by gut-residing actors stimulates bone loss remains to be elucidated. In this article, we discuss the main risk factors potentially contributing to intestinal disease-associated bone loss, and summarize current animal models, illustrating gut-bone axis to bridge the gap between intestinal inflammation and skeletal disease.
Collapse
|
61
|
Brady RD, Wong KR, Robinson DL, Mychasiuk R, McDonald SJ, D'Cunha RA, Yamakawa GR, Sun M, Wark JD, Lee PVS, O'Brien TJ, Casillas-Espinosa PM, Shultz SR. Bone Health in Rats With Temporal Lobe Epilepsy in the Absence of Anti-Epileptic Drugs. Front Pharmacol 2019; 10:1278. [PMID: 31749702 PMCID: PMC6842946 DOI: 10.3389/fphar.2019.01278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/07/2019] [Indexed: 01/18/2023] Open
Abstract
Rationale: Epilepsy patients often exhibit reduced bone mineral density and are at an increased risk of bone fracture. Whether these bone abnormalities are due to the use of anti-epileptic drugs (AED’s) or the disease itself is unknown. For example, although decreased bone health in epilepsy patients is generally attributed to the use of AED’s, seizures can also trigger a number of physiological processes that have the potential to affect bone. Therefore, to assess whether bone abnormalities occur in epilepsy in the absence of AED’s, the current study investigated mechanical characteristics and trabecular bone morphology in rats with chronic temporal lobe epilepsy. Methods: Ten-week old male Wistar rats underwent kainic acid-induced status epilepticus (SE; n = 7) or a sham procedure (n = 9). Rats were implanted with EEG recording electrodes at nine weeks post-SE, and video-EEG was continuously recorded for one week at 10- and 22-weeks post-SE to confirm that SE rats had spontaneous seizures. Open-field testing to assess locomotion was conducted at 23-weeks post-SE. At 24-weeks post-SE, rats were euthanized and tibia were extracted to determine trabecular morphology by micro-computed tomography (µCT), while femurs were used to investigate mechanical properties via 3-point bending. Results: All post-SE rats had spontaneous seizures at 10- and 22-weeks post-SE, while none of the sham rats had seizures. µCT trabecular analysis of tibia revealed no differences in total volume, bone volume, bone volume fraction, trabecular number, or trabecular separation between post-SE or sham rats, although post-SE rats did have increased trabecular thickness. There were also no group differences in total distance travelled in the open field suggesting that activity levels did not account for the increased trabecular thickness. In addition, no differences in mechanical properties of femurs were observed between the two groups. Conclusion: There was a lack of overt bone abnormalities in rats with chronic temporal lobe epilepsy in the absence of AED treatment. Although further studies are still needed, these findings may have important implications towards understanding the source (e.g., AED treatments) of bone abnormalities in epilepsy patients.
Collapse
Affiliation(s)
- Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Ker Rui Wong
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Dale L Robinson
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Ryan A D'Cunha
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - John D Wark
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Peter Vee Sin Lee
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
62
|
Brylka LJ, Schinke T. Chemokines in Physiological and Pathological Bone Remodeling. Front Immunol 2019; 10:2182. [PMID: 31572390 PMCID: PMC6753917 DOI: 10.3389/fimmu.2019.02182] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/29/2019] [Indexed: 12/21/2022] Open
Abstract
The bone matrix is constantly remodeled by bone-resorbing osteoclasts and bone-forming osteoblasts. These two cell types are fundamentally different in terms of progenitor cells, mode of action and regulation by specific molecules, acting either systemically or locally. Importantly, there is increasing evidence for an impact of cell types or molecules of the adaptive and innate immune system on bone remodeling. Understanding these influences is the major goal of a novel research area termed osteoimmunology, which is of key relevance in the context of inflammation-induced bone loss, skeletal metastases, and diseases of impaired bone remodeling, such as osteoporosis. This review article aims at summarizing the current knowledge on one particular aspect of osteoimmunology, namely the impact of chemokines on skeletal cells in order to regulate bone remodeling under physiological and pathological conditions. Chemokines have key roles in the adaptive immune system by controlling migration, localization, and function of immune cells during inflammation. The vast majority of chemokines are divided into two subgroups based on the pattern of cysteine residues. More specifically, there are 27 known C-C-chemokines, binding to 10 different C-C receptors, and 17 known C-X-C-chemokines binding to seven different C-X-C receptors. Three additional chemokines do not fall into this category, and only one of them, i.e., CX3CL1, has been shown to influence bone remodeling cell types. There is a large amount of published studies demonstrating specific effects of certain chemokines on differentiation and function of osteoclasts and/or osteoblasts. Chemokine signaling by skeletal cells or by other cells of the bone marrow niche regulates bone formation and resorption through autocrine and paracrine mechanisms. In vivo evidence from mouse deficiency models strongly supports the role of certain chemokine signaling pathways in bone remodeling. We will summarize these data in the present review with a special focus on the most established subsets of chemokines. In combination with the other review articles of this issue, the knowledge presented here confirms that there is a physiologically relevant crosstalk between the innate immune system and bone remodeling cell types, whose molecular understanding is of high clinical relevance.
Collapse
Affiliation(s)
- Laura J Brylka
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
63
|
Sgambato D, Gimigliano F, De Musis C, Moretti A, Toro G, Ferrante E, Miranda A, De Mauro D, Romano L, Iolascon G, Romano M. Bone alterations in inflammatory bowel diseases. World J Clin Cases 2019; 7:1908-1925. [PMID: 31423424 PMCID: PMC6695530 DOI: 10.12998/wjcc.v7.i15.1908] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/14/2019] [Accepted: 06/26/2019] [Indexed: 02/05/2023] Open
Abstract
Inflammatory bowel diseases (IBDs) are characterized by a multifactorial partially unknown etiology that involves genetic, immunological and environmental factors. Up to 50% of IBD patients experience at least one extraintestinal manifestation; among them is the involvement of bone density which is referred to as metabolic bone disease (MBD), including osteopenia and osteoporosis. Bone alterations in IBDs population appear to have a multifactorial etiology: Decreased physical activity, inflammation-related bone resorption, multiple intestinal resections, dietary malabsorption of minerals and vitamin D deficiency, genetic factors, gut-bone immune signaling interaction, steroid treatment, microbiota and pathogenic micro-organisms interaction, and dietary malabsorption of minerals, that, all together or individually, may contribute to the alteration of bone mineral density. This review aims to summarize the prevalence and pathophysiology of metabolic bone alterations in IBD subjects outlining the main risk factors of bone fragility. We also want to underline the role of the screening and prophylaxis of bone alterations in Crohn's disease and ulcerative colitis patients and the importance of treating appropriately MBD.
Collapse
Affiliation(s)
- Dolores Sgambato
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ‘‘Luigi Vanvitelli’’ and University Hospital, Naples 80131, Italy
| | - Francesca Gimigliano
- Department of Physical and Mental Health, University of Campania “Luigi Vanvitelli”, Naples 80131, Italy
| | - Cristiana De Musis
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ‘‘Luigi Vanvitelli’’ and University Hospital, Naples 80131, Italy
| | - Antimo Moretti
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, Naples 80131, Italy
| | - Giuseppe Toro
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, Naples 80131, Italy
| | - Emanuele Ferrante
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ‘‘Luigi Vanvitelli’’ and University Hospital, Naples 80131, Italy
| | - Agnese Miranda
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ‘‘Luigi Vanvitelli’’ and University Hospital, Naples 80131, Italy
| | - Domenico De Mauro
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ‘‘Luigi Vanvitelli’’ and University Hospital, Naples 80131, Italy
| | - Lorenzo Romano
- Surgical Digestive Endoscopy, Department of Clinical Medicine and Surgery, Federico II University, Naples 80131, Italy
| | - Giovanni Iolascon
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, Naples 80131, Italy
| | - Marco Romano
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ‘‘Luigi Vanvitelli’’ and University Hospital, Naples 80131, Italy
| |
Collapse
|
64
|
Madel MB, Ibáñez L, Wakkach A, de Vries TJ, Teti A, Apparailly F, Blin-Wakkach C. Immune Function and Diversity of Osteoclasts in Normal and Pathological Conditions. Front Immunol 2019; 10:1408. [PMID: 31275328 PMCID: PMC6594198 DOI: 10.3389/fimmu.2019.01408] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/04/2019] [Indexed: 12/31/2022] Open
Abstract
Osteoclasts (OCLs) are key players in controlling bone remodeling. Modifications in their differentiation or bone resorbing activity are associated with a number of pathologies ranging from osteopetrosis to osteoporosis, chronic inflammation and cancer, that are all characterized by immunological alterations. Therefore, the 2000s were marked by the emergence of osteoimmunology and by a growing number of studies focused on the control of OCL differentiation and function by the immune system. At the same time, it was discovered that OCLs are much more than bone resorbing cells. As monocytic lineage-derived cells, they belong to a family of cells that displays a wide heterogeneity and plasticity and that is involved in phagocytosis and innate immune responses. However, while OCLs have been extensively studied for their bone resorption capacity, their implication as immune cells was neglected for a long time. In recent years, new evidence pointed out that OCLs play important roles in the modulation of immune responses toward immune suppression or inflammation. They unlocked their capacity to modulate T cell activation, to efficiently process and present antigens as well as their ability to activate T cell responses in an antigen-dependent manner. Moreover, similar to other monocytic lineage cells such as macrophages, monocytes and dendritic cells, OCLs display a phenotypic and functional plasticity participating to their anti-inflammatory or pro-inflammatory effect depending on their cell origin and environment. This review will address this novel vision of the OCL, not only as a phagocyte specialized in bone resorption, but also as innate immune cell participating in the control of immune responses.
Collapse
Affiliation(s)
- Maria-Bernadette Madel
- CNRS, Laboratoire de PhysioMédecine Moléculaire, Faculté de Médecine, UMR7370, Nice, France.,Faculé de Médecine, Université Côte d'Azur, Nice, France
| | - Lidia Ibáñez
- Department of Pharmacy, Cardenal Herrera-CEU University, València, Spain
| | - Abdelilah Wakkach
- CNRS, Laboratoire de PhysioMédecine Moléculaire, Faculté de Médecine, UMR7370, Nice, France.,Faculé de Médecine, Université Côte d'Azur, Nice, France
| | - Teun J de Vries
- Department of Periodontology, Academic Centre of Dentistry Amsterdam, University of Amsterdam and Vrije Univeristeit, Amsterdam, Netherlands
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | | - Claudine Blin-Wakkach
- CNRS, Laboratoire de PhysioMédecine Moléculaire, Faculté de Médecine, UMR7370, Nice, France.,Faculé de Médecine, Université Côte d'Azur, Nice, France
| |
Collapse
|
65
|
Osteoimmunology: evolving concepts in bone-immune interactions in health and disease. Nat Rev Immunol 2019; 19:626-642. [PMID: 31186549 DOI: 10.1038/s41577-019-0178-8] [Citation(s) in RCA: 461] [Impact Index Per Article: 76.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2019] [Indexed: 12/14/2022]
Abstract
In terrestrial vertebrates, bone tissue constitutes the 'osteoimmune' system, which functions as a locomotor organ and a mineral reservoir as well as a primary lymphoid organ where haematopoietic stem cells are maintained. Bone and mineral metabolism is maintained by the balanced action of bone cells such as osteoclasts, osteoblasts and osteocytes, yet subverted by aberrant and/or prolonged immune responses under pathological conditions. However, osteoimmune interactions are not restricted to the unidirectional effect of the immune system on bone metabolism. In recent years, we have witnessed the discovery of effects of bone cells on immune regulation, including the function of osteoprogenitor cells in haematopoietic stem cell regulation and osteoblast-mediated suppression of haematopoietic malignancies. Moreover, the dynamic reciprocal interactions between bone and malignancies in remote organs have attracted attention, extending the horizon of osteoimmunology. Here, we discuss emerging concepts in the osteoimmune dialogue in health and disease.
Collapse
|
66
|
Cheng C, Hua J, Tan J, Qian W, Zhang L, Hou X. Identification of differentially expressed genes, associated functional terms pathways, and candidate diagnostic biomarkers in inflammatory bowel diseases by bioinformatics analysis. Exp Ther Med 2019; 18:278-288. [PMID: 31258663 PMCID: PMC6566124 DOI: 10.3892/etm.2019.7541] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 03/29/2019] [Indexed: 12/20/2022] Open
Abstract
Inflammatory bowel diseases (IBDs), including ulcerative colitis (UC) and Crohn's disease (CD), are chronic inflammatory disorders caused by genetic influences, the immune system and environmental factors. However, the underlying pathogenesis of IBDs and the pivotal molecular interactions remain to be fully elucidated. The aim of the present study was to identify genetic signatures in patients with IBDs and elucidate the potential molecular mechanisms underlying IBD subtypes. The gene expression profiles of the GSE75214 datasets were obtained from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified in UC and CD patients compared with controls using the GEO2R tool. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of DEGs were performed using DAVID. Furthermore, protein-protein interaction (PPI) networks of the DEGs were constructed using Cytoscape software. Subsequently, significant modules were selected and the hub genes were identified. In the GO and KEGG pathway analysis, the top enriched pathways in UC and CD included Staphylococcus aureus infection, rheumatoid arthritis, complement and coagulation cascades, PI3K/Akt signaling pathway and osteoclast differentiation. In addition, the GO terms in the category biological process significantly enriched by these genes were inflammatory response, immune response, leukocyte migration, cell adhesion, response to molecules of bacterial origin and extracellular matrix (ECM) organization. However, several other biological processes (GO terms) and pathways (e.g., ‘chemotaxis’, ‘collagen catabolic process’ and ‘ECM-receptor interaction’) exhibited significant differences between the two subtypes of IBD. The top 10 hub genes were identified from the PPI network using respective DEGs. Of note, the hub genes G protein subunit gamma 11 (GNG11), G protein subunit beta 4 (GNB4), Angiotensinogen (AGT), Phosphoinositide-3-kinase regulatory subunit 3 (PIK3R3) and C-C motif chemokine receptor 7 (CCR7) are disease-specific and may be used as biomarkers for differentiating UC from CD. Furthermore, module analysis further confirmed that common significant pathways involved in the pathogenesis of IBD subtypes were associated with chemokine-induced inflammation, innate immunity, adapted immunity and infectious microbes. In conclusion, the present study identified DEGs, key target genes, functional pathways and enrichment analysis of IBDs, enhancing the understanding of the pathogenesis of IBDs and also advancing the clarification of the underlying molecular mechanisms of UC and CD. Furthermore, these results may provide potential molecular targets and diagnostic biomarkers for UC and CD.
Collapse
Affiliation(s)
- Chunwei Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Juan Hua
- Department of Cardiology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, Hubei 430015, P.R. China
| | - Jun Tan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wei Qian
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lei Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
67
|
Schmidt T, Schwinge D, Rolvien T, Jeschke A, Schmidt C, Neven M, Butscheidt S, Kriz M, Kunzmann L, Mussawy H, Hubert J, Hawellek T, Rüther W, Oheim R, Barvencik F, Lohse AW, Schramm C, Schinke T, Amling M. Th17 cell frequency is associated with low bone mass in primary sclerosing cholangitis. J Hepatol 2019; 70:941-953. [PMID: 30641095 DOI: 10.1016/j.jhep.2018.12.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/19/2018] [Accepted: 12/26/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND & AIMS Osteoporotic fractures are a major cause of morbidity and reduced quality of life in patients with primary sclerosing cholangitis (PSC), a progressive bile duct disease of unknown origin. Although it is generally assumed that this pathology is a consequence of impaired calcium homeostasis and malabsorption, the cellular and molecular causes of PSC-associated osteoporosis are unknown. METHODS We determined bone mineral density by dual-X-ray absorptiometry and assessed bone microstructure by high-resolution peripheral quantitative computed tomography in patients with PSC. Laboratory markers of liver and bone metabolism were measured, and liver stiffness was assessed by FibroScan. We determined the frequency of Th17 cells by the ex vivo stimulation of peripheral blood mononuclear cells in a subgroup of 40 patients with PSC. To investigate the potential involvement of IL-17 in PSC-associated bone loss, we analyzed the skeletal phenotype of mice lacking Abcb4 and/or Il-17. RESULTS Unlike in patients with primary biliary cholangitis, bone loss in patients with PSC was not associated with disease duration or liver fibrosis. However, we observed a significant negative correlation between the bone resorption biomarker deoxypyridinoline and bone mineral density in the PSC cohort, indicating increased bone resorption. Importantly, the frequency of Th17 cells in peripheral blood was positively correlated with the urinary deoxypyridinoline level and negatively correlated with bone mass. We observed that Abcb4-deficient mice displayed a low-bone-mass phenotype, which was corrected by an additional Il-17 deficiency or anti-IL-17 treatment, whereas the liver pathology was unaffected. CONCLUSIONS Our findings demonstrate that an increased frequency of Th17 cells is associated with bone resorption in PSC. Whether antibody-based IL-17 blockade is beneficial against bone loss in patients with PSC should be addressed in future studies. LAY SUMMARY Primary sclerosing cholangitis (PSC) is a cholestatic liver disease characterized by progressive bile duct destruction. One serious complication of PSC is reduced bone mass resulting in increased fracture risk. Herein, we demonstrate that Th17 cells mediate bone loss in PSC by inducing bone resorption, which suggests that antibody-based IL-17 blockade might be beneficial for the treatment of bone loss in affected patients.
Collapse
Affiliation(s)
- Tobias Schmidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Dorothee Schwinge
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Anke Jeschke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Constantin Schmidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Mona Neven
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Sebastian Butscheidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Marvin Kriz
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Lilly Kunzmann
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Haider Mussawy
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Jan Hubert
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Thelonius Hawellek
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Wolfgang Rüther
- Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Florian Barvencik
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Ansgar W Lohse
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Christoph Schramm
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.
| |
Collapse
|
68
|
Cao WT, Fan YH, Lv B. Intestinal microbial markers for diagnosis of inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2019; 27:190-196. [DOI: 10.11569/wcjd.v27.i3.190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is caused by immune, genetic, and environmental factors together. It is difficult to accurately diagnose IBD early because it is characterized by atypical symptoms at early stage and diverse lesions at late stage. The diagnosis is currently dependent on endoscopic and imaging examinations, but patients often have poor compliance. The characteristic change of gut microbiota in IBD suggests that it may become a new biomarker. In recent years, a number of clinical studies have focused on studying the role of characteristic changes of gut microbiota in the differential diagnosis, disease activity, and extra-intestinal manifestations of IBD. These studies have established a gut microbiota based diagnostic model with high sensitivity and specificity, but the model is susceptible to be influenced by external factors and needs to be further improved. This paper reviews the diagnostic value and clinical significance of gut microbiota in IBD.
Collapse
Affiliation(s)
- Wan-Ting Cao
- Department of Gastroenterology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Yi-Hong Fan
- Department of Gastroenterology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Bin Lv
- Department of Gastroenterology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| |
Collapse
|
69
|
Ge X, Chen Z, Xu Z, Lv F, Zhang K, Yang Y. The effects of dihydroartemisinin on inflammatory bowel disease-related bone loss in a rat model. Exp Biol Med (Maywood) 2019; 243:715-724. [PMID: 29763384 DOI: 10.1177/1535370218769420] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bone loss is one of the important extra-intestinal manifestations in patients with inflammatory bowel diseases (IBDs). Compounds derived from natural products have been used to treat IBDs. However, the role of natural products on IBD-induced bone loss is not completely clarified. In the present study, we observed the effects of dihydroartemisinin (DHA), an antimalaria drug, on IBD and IBD-induced bone loss in a rat model. Chronic IBD model was established in Sprague-Dawley rats by giving them 2.5% dextran sodium sulfate in drinking water. DHA was given by intraperitoneal injection. Blood, colon, and bone samples were collected for biomarker assay and histological analysis. There was an obvious increase in tumor necrotic factor (TNF) α and receptor activator of nuclear factor (NF)-kB ligand (RANKL), and decrease in procollagen type 1 N-terminal propeptide (P1NP) level in IBD groups compared with the normal control (p < 0.05). The disease activity score of IBD rats was significantly higher than the control (p < 0.01). Obvious decrease in disease activity score, TNFα, and RANKL level and increase in P1NP were observed in DHA-treated IBD rats. Bone loss, shown as the decrease in bone mineral density, bone volume fraction, and trabecular number and increase in trabecular separation were observed in IBD rats compared with control (p < 0.01). DHA treatment obviously abolished the bone loss, in particular in the high-dose group (p < 0.05). DHA treatment also inhibited the excessive osteoclast formation; RANKL protein expression; and RANK, TRAF6, Fra-1, NFATc1 mRNA expression induced by IBD. Our data indicated that DHA may be a potential therapeutic agent for IBD and IBD-induced bone loss. Impact statement Bone loss is one of the important extra-intestinal manifestations in patients with inflammatory bowel diseases (IBDs). Studies have shown that compounds derived from natural products are useful in the treatment of IBDs. However, few studies have investigated the role of compounds derived from natural products in treatment of osteoporosis in IBDs. The current study aimed to show the effects of dihydroartemisinin (DHA), antimalaria drug, on bone loss in a rat model of IBD. The findings showed that DHA intervention dose dependently protected against bone loss in IBD rats by inhibiting tumor necrotic factor α production and osteoclast formation. These findings highlights that DHA may be beneficial for bone health in those patients with IBD.
Collapse
Affiliation(s)
- Xingtao Ge
- 1 Department of Orthopedics, Rizhao People's Hospital, Rizhao city 276800, China
| | - Zhijian Chen
- 2 Department of Nuclear Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Zhenjie Xu
- 3 Department of Clinical Laboratory, Rizhao People's Hospital, Rizhao 276800, China
| | - Fang Lv
- 4 Department of Rheumatology and Immunology, Rizhao People's Hospital, Rizhao 276800, China
| | - Kewei Zhang
- 5 Department of Nephrology, Fuyang Traditional Chinese Medicine Hospital of Hangzhou, Hangzhou 311400, China
| | - Yu Yang
- 6 Department of Geriatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| |
Collapse
|
70
|
Madel MB, Ibáñez L, Rouleau M, Wakkach A, Blin-Wakkach C. A Novel Reliable and Efficient Procedure for Purification of Mature Osteoclasts Allowing Functional Assays in Mouse Cells. Front Immunol 2018; 9:2567. [PMID: 30450105 PMCID: PMC6224441 DOI: 10.3389/fimmu.2018.02567] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/17/2018] [Indexed: 01/07/2023] Open
Abstract
Osteoclasts (OCLs) are multinucleated phagocytes of monocytic origin responsible for physiological and pathological bone resorption including aging processes, chronic inflammation and cancer. Besides bone resorption, they are also involved in the modulation of immune responses and the regulation of hematopoietic niches. Accordingly, OCLs are the subject of an increasing number of studies. Due to their rarity and the difficulty to isolate them directly ex vivo, analyses on OCLs are usually performed on in vitro differentiated cells. In this state, however, OCLs represent a minority of differentiated cells. Since up to date a reliable purification procedure is still lacking for mature OCLs, all cells present in the culture are analyzed collectively to answer OCL-specific questions. With the development of in-depth transcriptomic and proteomic analyses, such global analyses on unsorted cells can induce severe bias effects in further results. In addition, for instance, analysis on OCL immune function requires working on purified OCLs to avoid contamination effects of monocytic precursors that may persist during the culture. This clearly highlights the need for a reliable OCL purification procedure. Here, we describe a novel and reliable method to sort OCLs based on cell multinucleation while preserving cell viability. Using this method, we successfully purified multinucleated murine cells. We showed that they expressed high levels of OCL markers and retained a high capacity of bone resorption, demonstrating that these are mature OCLs. The same approach was equally applied for the purification of human mature OCLs. Comparison of purified OCLs with mononucleated cells or unsorted cells revealed significant differences in the expression of OCL-specific markers at RNA and/or protein level. This exemplifies that substantially better outcomes for OCLs are achieved after the exclusion of mononucleated cells. Our results clearly demonstrate that the in here presented procedure for the analysis and sorting of pure OCLs represents a novel, robust and reliable method for the detailed examination of bona fide mature OCLs in a range that was previously impossible. Noteworthy, this procedure will open new perspectives into the biology of osteoclasts and osteoclast-related diseases.
Collapse
Affiliation(s)
- Maria-Bernadette Madel
- CNRS UMR7370, Laboratoire de PhysioMédecine Moléculaire, Faculty of Medicine, Nice, France.,University Nice Sophia Antipolis, Nice, France
| | - Lidia Ibáñez
- CNRS UMR7370, Laboratoire de PhysioMédecine Moléculaire, Faculty of Medicine, Nice, France.,University Nice Sophia Antipolis, Nice, France
| | - Matthieu Rouleau
- CNRS UMR7370, Laboratoire de PhysioMédecine Moléculaire, Faculty of Medicine, Nice, France.,University Nice Sophia Antipolis, Nice, France
| | - Abdelilah Wakkach
- CNRS UMR7370, Laboratoire de PhysioMédecine Moléculaire, Faculty of Medicine, Nice, France.,University Nice Sophia Antipolis, Nice, France
| | - Claudine Blin-Wakkach
- CNRS UMR7370, Laboratoire de PhysioMédecine Moléculaire, Faculty of Medicine, Nice, France.,University Nice Sophia Antipolis, Nice, France
| |
Collapse
|
71
|
Brunetti G, Rizzi R, Storlino G, Bortolotti S, Colaianni G, Sanesi L, Lippo L, Faienza MF, Mestice A, Curci P, Specchia G, Grano M, Colucci S. LIGHT/TNFSF14 as a New Biomarker of Bone Disease in Multiple Myeloma Patients Experiencing Therapeutic Regimens. Front Immunol 2018; 9:2459. [PMID: 30405638 PMCID: PMC6206078 DOI: 10.3389/fimmu.2018.02459] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 10/04/2018] [Indexed: 11/13/2022] Open
Abstract
We have previously shown that through the production of high LIGHT levels, immune cells contribute to both osteoclastogenesis and bone destruction in Multiple Myeloma (MM)-related bone disease. With the aim of further exploring the mechanisms underlying the development of MM-related bone disease, here we focused on a possible role of LIGHT in MM patients with active bone disease despite the treatment received. We detected LIGHT over-expression by circulating CD14+ monocytes from MM patients still showing active bone disease, despite the treatment. In addition, we found over-expression of receptor activator of nuclear factor kappa-B ligand (RANKL), whose pro-osteoclastogenic role is well-known, in T-lymphocytes isolated from the same patients. Although the percentages of circulating osteoclast progenitors, CD14+CD16+ monocytes, were higher in all the MM patients than in the controls spontaneous osteoclastogenesis occurred only in the cultures derived from PBMCs of MM patients with unresponsive bone disease. Of note, in the same cultures osteoclastogenesis was partially or completely inhibited, in a dose-dependent manner, by the addition of RANK-Fc or anti-LIGHT neutralizing antibody, demonstrating the contribution of both LIGHT and RANKL to the enhanced osteoclast formation observed. In addition, high serum levels of TRAP5b and CTX, the two markers of osteoclast activity, were detected in MM patients with bone disease not responsive to treatment. In conclusion, our study indicates a prominent role of LIGHT in the crosstalk among osteoclasts and immune cells, co-involved together with RANKL in the pathophysiological mechanisms leading to MM-related bone disease. This TNF superfamily member may thus be a possible new therapeutic target in MM-related bone disease.
Collapse
Affiliation(s)
- Giacomina Brunetti
- Section of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, School of Medicine, University of Bari, Bari, Italy
| | - Rita Rizzi
- Section of Hematology, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | - Giuseppina Storlino
- Section of Human Anatomy and Histology, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | - Sara Bortolotti
- Section of Human Anatomy and Histology, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | - Graziana Colaianni
- Section of Human Anatomy and Histology, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | - Lorenzo Sanesi
- Section of Human Anatomy and Histology, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | - Luciana Lippo
- Section of Human Anatomy and Histology, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | - Maria Felicia Faienza
- Paediatric Unit, Department of Biomedical Science and Human Oncology, University of Bari, Bari, Italy
| | - Anna Mestice
- Section of Hematology, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | - Paola Curci
- Section of Hematology, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | - Giorgina Specchia
- Section of Hematology, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | - Maria Grano
- Section of Human Anatomy and Histology, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | - Silvia Colucci
- Section of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, School of Medicine, University of Bari, Bari, Italy
| |
Collapse
|
72
|
Sylvester FA. Inflammatory Bowel Disease: Effects on Bone and Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1033:133-150. [PMID: 29101654 DOI: 10.1007/978-3-319-66653-2_7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammatory bowel disease (IBD) is associated with decreased bone mass and alterations in bone geometry from the time of diagnosis, before anti-inflammatory therapy is instituted. Deficits in bone mass can persist despite absence of symptoms of active IBD. The effects of IBD on the skeleton are complex. Protein-calorie malnutrition, inactivity, hypogonadism, deficits in calcium intake and vitamin D consumption and synthesis, stunted growth in children, decreased skeletal muscle mass, and inflammation all likely play a role. Preliminary studies suggest that the dysbiotic intestinal microbial flora present in IBD may also affect bone at a distance. Several mechanisms are possible. T cells activated by the gut microbiota may serve as "inflammatory shuttles" between the intestine and bone. Microbe-associated molecular patterns leaked into the circulation in IBD may activate immune responses in the bone marrow by immune cells and by osteocytes, osteoblasts, and osteoclasts that lead to decreased bone formation and increased resorption. Finally, intestinal microbial metabolites such as H2S may also affect bone cell function. Uncovering these mechanisms will enable the design of microbial cocktails to help restore bone mass in patients with IBD.
Collapse
Affiliation(s)
- Francisco A Sylvester
- Division Chief of Pediatric Gastroenterology, The University of North Carolina at Chapel Hil, 333 South Columbia Street, MacNider Hall 247, Chapel Hill, NC, 27599-7229, USA.
| |
Collapse
|
73
|
Liu TH, Tsai TY, Pan TM. The Anti-Periodontitis Effects of Ethanol Extract Prepared Using Lactobacillus paracasei subsp. paracasei NTU 101. Nutrients 2018; 10:nu10040472. [PMID: 29649103 PMCID: PMC5946257 DOI: 10.3390/nu10040472] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 11/21/2022] Open
Abstract
Poor oral health and related diseases, including caries, periodontal disease, and oral cancer, are highly prevalent across the world, particularly in the elderly. This study aimed to investigate the anti-periodontitis activity of fermented skim milk produced using the promising probiotic Lactobacillus paracasei subsp. paracasei NTU 101 (NTU101FM). An initial analysis found that an ethanol extract of NTU101FM displayed anti-oxidative activities. Further investigation of pathogen growth inhibition zones, minimum inhibitory concentrations (MICs), and minimum bactericidal concentrations (MBCs) revealed that the NTU101FM ethanol extract also had anti-periodontal pathogen activities. In addition, the NTU101FM ethanol extract significantly decreased the release of pro-inflammatory cytokines induced by lipopolysaccharide (LPS) in RAW 264.7 macrophage cells. Finally, the NTU101FM ethanol extract was found to inhibit receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation by reducing tartrate-resistant acid phosphatase (TRAP) activity and the number of TRAP-positive multinucleated osteoclasts. In summary, our study demonstrated that ethanol extract prepared from NTU101FM has potential use as an anti-periodontitis agent.
Collapse
Affiliation(s)
- Te-Hua Liu
- Department of Biochemical Science & Technology, National Taiwan University, Taipei 10617, Taiwan.
| | - Tsung-Yu Tsai
- Department of Food Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan.
| | - Tzu-Ming Pan
- Department of Biochemical Science & Technology, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
74
|
Ibáñez L, Rouleau M, Wakkach A, Blin-Wakkach C. Gut microbiome and bone. Joint Bone Spine 2018; 86:43-47. [PMID: 29654948 DOI: 10.1016/j.jbspin.2018.02.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2018] [Indexed: 12/19/2022]
Abstract
The gut microbiome is now viewed as a tissue that interacts bidirectionally with the gastrointestinal, immune, endocrine and nervous systems, affecting the cellular responses in numerous organs. Evidence is accumulating of gut microbiome involvement in a growing number of pathophysiological processes, many of which are linked to inflammatory responses. More specifically, data acquired over the last decade point to effects of the gut microbiome on bone mass regulation and on the development of bone diseases (such as osteoporosis) and of inflammatory joint diseases characterized by bone loss. Mice lacking a gut microbiome have bone mass alteration that can be reversed by gut recolonization. Changes in the gut microbiome composition have been reported in mice with estrogen-deficiency osteoporosis and have also been found in a few studies in humans. Probiotic therapy decreases bone loss in estrogen-deficient animals. The effect of the gut microbiome on bone tissue involves complex mechanisms including modulation of CD4+T cell activation, control of osteoclastogenic cytokine production and modifications in hormone levels. This complexity may contribute to explain the discrepancies observed betwwen some studies whose results vary depending on the age, gender, genetic background and treatment duration. Further elucidation of the mechanisms involved is needed. However, the available data hold promise that gut microbiome manipulation may prove of interest in the management of bone diseases.
Collapse
Affiliation(s)
- Lidia Ibáñez
- CNRS UMR7370, LP2M, faculté de médecine, 28, avenue de Valombrose, 06107 Nice cedex 2, France; Université Nice-Sophia-Antipolis, 06100 Nice, France; Department of Pharmacy, Cardenal Herrera-CEU University, 46115 Alfara del Patriarca, València, Spain
| | - Matthieu Rouleau
- CNRS UMR7370, LP2M, faculté de médecine, 28, avenue de Valombrose, 06107 Nice cedex 2, France; Université Nice-Sophia-Antipolis, 06100 Nice, France
| | - Abdelilah Wakkach
- CNRS UMR7370, LP2M, faculté de médecine, 28, avenue de Valombrose, 06107 Nice cedex 2, France; Université Nice-Sophia-Antipolis, 06100 Nice, France
| | - Claudine Blin-Wakkach
- CNRS UMR7370, LP2M, faculté de médecine, 28, avenue de Valombrose, 06107 Nice cedex 2, France; Université Nice-Sophia-Antipolis, 06100 Nice, France.
| |
Collapse
|
75
|
Srivastava RK, Dar HY, Mishra PK. Immunoporosis: Immunology of Osteoporosis-Role of T Cells. Front Immunol 2018; 9:657. [PMID: 29675022 PMCID: PMC5895643 DOI: 10.3389/fimmu.2018.00657] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/16/2018] [Indexed: 12/28/2022] Open
Abstract
The role of immune system in various bone pathologies, such as osteoporosis, osteoarthritis, and rheumatoid arthritis is now well established. This had led to the emergence of a modern field of systems biology called as osteoimmunology, an integrated research between fields of immunology and bone biology under one umbrella. Osteoporosis is one of the most common inflammatory bone loss condition with more than 200 million individuals affected worldwide. T helper (Th) cells along with various other immune cells are major players involved in bone homeostasis. In the present review, we specifically discuss the role of various defined T lymphocyte subsets (Th cells comprising Th1, Th2, Th9, Th17, Th22, regulatory T cells, follicular helper T cells, natural killer T cells, γδ T cells, and CD8+ T cells) in the pathophysiology of osteoporosis. The study of the specific role of immune system in osteoporosis has now been proposed by our group as "immunoporosis: the immunology of osteoporosis" with special emphasis on the role of various subsets of T lymphocytes. The establishment of this new field had been need of the hour due to the emergence of novel roles of various T cell lymphocytes in accelerated bone loss observed during osteoporosis. Activated T cells either directly or indirectly through the secretion of various cytokines and factors modulate bone health and thereby regulate bone remodeling. Several studies have summarized the role of inflammation in pathogenesis of osteoporosis but very few reports had delineated the precise role of various T cell subsets in the pathobiology of osteoporosis. The present review thus for the first time clearly highlights and summarizes the role of various T lymphocytes in the development and pathophysiology of osteoporosis, giving birth to a new field of biology termed as "immunoporosis". This novel field will thus provide an overview of the nexus between the cellular components of both bone and immune systems, responsible for the observed bone loss in osteoporosis. A molecular insight into the upcoming and novel field of immunoporosis would thus leads to development of innovative approaches for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Rupesh K. Srivastava
- Department of Zoology, School of Biological Sciences, Dr. Hari Singh Gour University, Sagar, India
- Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Hamid Y. Dar
- Department of Zoology, School of Biological Sciences, Dr. Hari Singh Gour University, Sagar, India
| | - Pradyumna K. Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| |
Collapse
|
76
|
McCabe LR, Parameswaran N. Advances in Probiotic Regulation of Bone and Mineral Metabolism. Calcif Tissue Int 2018; 102:480-488. [PMID: 29453726 PMCID: PMC5849527 DOI: 10.1007/s00223-018-0403-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/08/2018] [Indexed: 12/19/2022]
Abstract
Probiotics have been consumed by humans for thousands of years because they are beneficial for long-term storage of foods and promote the health of their host. Ingested probiotics reside in the gastrointestinal tract where they have many effects including modifying the microbiota composition, intestinal barrier function, and the immune system which result in systemic benefits to the host, including bone health. Probiotics benefit bone growth, density, and structure under conditions of dysbiosis, intestinal permeability, and inflammation (recognized mediators of bone loss and osteoporosis). It is likely that multiple mechanisms are involved in mediating probiotic signals from the gut to the bone. Studies indicate a role for the microbiota (composition and activity), intestinal barrier function, and immune cells in the signaling process. These mechanisms are not mutually exclusive, but rather, may synergize to provide benefits to the skeletal system of the host and serve as a starting point for investigation. Given that probiotics hold great promise for supporting bone health and are generally regarded as safe, future studies identifying mechanisms are warranted.
Collapse
Affiliation(s)
- Laura R McCabe
- Department of Physiology, Michigan State University, East Lansing, MI, USA.
- Department of Radiology, Michigan State University, East Lansing, MI, USA.
- Biomedical Imaging Research Center, Michigan State University, East Lansing, MI, USA.
| | | |
Collapse
|
77
|
Li Y, Yu Q, Zhang Z, Wang J, Li S, Zhang J, Liu G. TH9 cell differentiation, transcriptional control and function in inflammation, autoimmune diseases and cancer. Oncotarget 2018; 7:71001-71012. [PMID: 27589682 PMCID: PMC5342605 DOI: 10.18632/oncotarget.11681] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/26/2016] [Indexed: 12/31/2022] Open
Abstract
Naïve CD4+T cells differentiate into various T cell subsets depending on the specific cytokine environment. TH9 cells are less well-characterized than other T cell subsets, and factors that control their development and function have only recently been identified. It is now clear that TH9 cells play critical roles in immune-mediated diseases, including allergic airway, autoimmune and inflammatory bowel diseases, and cancer. Thus, the promotion or suppression of TH9 cell differentiation, transcriptional control and function may provide novel treatments for clinical inflammation, autoimmune diseases and tumors.
Collapse
Affiliation(s)
- Yan Li
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China.,Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Yu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Zhengguo Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China.,Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jian Wang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China.,Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Simin Li
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jiangyuan Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China.,Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
78
|
Wang Y, Lin F, Zhu X, Leone VA, Dalal S, Tao Y, Messer JS, Chang EB. Distinct roles of intracellular heat shock protein 70 in maintaining gastrointestinal homeostasis. Am J Physiol Gastrointest Liver Physiol 2018; 314:G164-G178. [PMID: 29051186 PMCID: PMC5866418 DOI: 10.1152/ajpgi.00208.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 01/31/2023]
Abstract
The inducible heat shock protein 70 (Hsp70) is both cytoprotective and immunomodulatory, potentially accounting for its critical role in maintaining gastrointestinal homeostasis. When levels are reduced in conditions like inflammatory bowel diseases (IBD), loss of function contributes to the severity and chronicity of these diseases, although through which cell types and mechanisms remains unclear. Here, the role of Hsp70-mediated intestinal epithelial protection and immune regulation in experimental colitis was examined by using a villin promoter-driven Hsp70 transgene in the 2,4,6-trinitrobenzene sulfonic acid (TNBS) and dextran sodium sulfate (DSS) models and in IL-10/Hsp70 double knockout (IL10-/-/Hsp70-/-) mice. In addition, Hsp70-mediated IL-10 production and immune protection were investigated using a CD45RBhigh transfer model and measuring colonic and immune cell cytokine expression during colitis. We found that the epithelial-specific expression of Hsp70 transgene attenuated DSS-induced colitis in Hsp70-/- mice by protecting tight junctions (TJ) and their interaction with the TJ-associated protein ZO-1. In the TNBS colitis and CD45RBhigh model, Hsp70 carried out its intracellular anti-inflammatory function by maintaining IL-10 production. Impaired ERK phosphorylation, but not p38 or JNK phosphorylation pathways, was associated with decreased IL-10 production in Hsp70-deficient cells. Together, these actions can be leveraged in the context of cellular specificity to develop complementary strategies that can lead to reduction in mucosal injury and immune activation in colonic colitis development. NEW & NOTEWORTHY Using four different experimental colitis models, we filled an important gap in knowledge by defining essential roles of intracellular heat shock protein 70 in different cell types in maintaining intestinal integrity and immune regulation. These findings are relevant to human inflammatory bowel diseases and represent potential avenues for developing therapeutic strategies, not only to counter the destructive processes of inflammation but also to promote tissue healing and prevent complications frequently associated with chronic intestinal inflammation.
Collapse
Affiliation(s)
- Yunwei Wang
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago , Chicago, Illinois
| | - Fanfei Lin
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago , Chicago, Illinois
| | - Xiaorong Zhu
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago , Chicago, Illinois
| | - Vanessa A Leone
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago , Chicago, Illinois
| | - Sushila Dalal
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago , Chicago, Illinois
| | - Yun Tao
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago , Chicago, Illinois
| | - Jeannette S Messer
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago , Chicago, Illinois
| | - Eugene B Chang
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago , Chicago, Illinois
| |
Collapse
|
79
|
Roux C, Saviane G, Pini J, Belaïd N, Dhib G, Voha C, Ibáñez L, Boutin A, Mazure NM, Wakkach A, Blin-Wakkach C, Rouleau M. Immunosuppressive Mesenchymal Stromal Cells Derived from Human-Induced Pluripotent Stem Cells Induce Human Regulatory T Cells In Vitro and In Vivo. Front Immunol 2018; 8:1991. [PMID: 29422893 PMCID: PMC5788894 DOI: 10.3389/fimmu.2017.01991] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/21/2017] [Indexed: 12/18/2022] Open
Abstract
Despite mesenchymal stromal cells (MSCs) are considered as a promising source of cells to modulate immune functions on cells from innate and adaptive immune systems, their clinical use remains restricted (few number, limited in vitro expansion, absence of a full phenotypic characterization, few insights on their in vivo fate). Standardized MSCs derived in vitro from human-induced pluripotent stem (huIPS) cells, remediating part of these issues, are considered as well as a valuable tool for therapeutic approaches, but their functions remained to be fully characterized. We generated multipotent MSCs derived from huiPS cells (huiPS-MSCs), and focusing on their immunosuppressive activity, we showed that human T-cell activation in coculture with huiPS-MSCs was significantly reduced. We also observed the generation of functional CD4+ FoxP3+ regulatory T (Treg) cells. Further tested in vivo in a model of human T-cell expansion in immune-deficient NSG mice, huiPS-MSCs immunosuppressive activity prevented the circulation and the accumulation of activated human T cells. Intracytoplasmic labeling of cytokines produced by the recovered T cells showed reduced percentages of human-differentiated T cells producing Th1 inflammatory cytokines. By contrast, T cells producing IL-10 and FoxP3+-Treg cells, absent in non-treated animals, were detected in huiPS-MSCs treated mice. For the first time, these results highlight the immunosuppressive activity of the huiPS-MSCs on human T-cell stimulation with a concomitant generation of human Treg cells in vivo. They may favor the development of new tools and strategies based on the use of huiPS cells and their derivatives for the induction of immune tolerance.
Collapse
Affiliation(s)
- Clémence Roux
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France.,Service d'Hématologie Clinique, CHU de Nice, Hôpital de l'Archet, Nice, France
| | - Gaëlle Saviane
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Jonathan Pini
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Nourhène Belaïd
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Gihen Dhib
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Christine Voha
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France.,Pôle d'Odontologie, CHU de Nice, Hôpital Saint-Roch, Nice, France
| | - Lidia Ibáñez
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Antoine Boutin
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Nathalie M Mazure
- Université Nice-Sophia Antipolis, Nice, France.,Institute for Research on Cancer and Aging of Nice, CNRS-UMR 7284-INSERM U108, Centre Antoine Lacassagne, Nice, France
| | - Abdelilah Wakkach
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Claudine Blin-Wakkach
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| | - Matthieu Rouleau
- LP2M, CNRS-UMR 7370, Faculty of Medicine, Nice, France.,Université Nice-Sophia Antipolis, Nice, France
| |
Collapse
|
80
|
A STAT3-dependent transcriptional circuitry inhibits cytotoxic gene expression in T cells. Proc Natl Acad Sci U S A 2017; 114:13236-13241. [PMID: 29180433 DOI: 10.1073/pnas.1711160114] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
CD8+ T cells are preprogrammed for cytotoxic differentiation in the thymus as they acquire expression of the transcription factor Runx3. However, a subset of effector CD8+ T cells (Tc17) produce IL-17 and fail to express cytotoxic genes. Here, we show that the transcription factors directing IL-17 production, STAT3 and RORγt, inhibit cytotoxicity despite persistent Runx3 expression. Cytotoxic gene repression did not require the transcription factor Thpok, which in CD4+ T cells restrains Runx3 functions and cytotoxicity; and STAT3 restrained cytotoxic gene expression in CD8+ T cells responding to viral infection in vivo. STAT3-induced RORγt represses cytotoxic genes by inhibiting the functions but not the expression of the "cytotoxic" transcription factors T-bet and Eomesodermin. Thus, the transcriptional circuitry directing IL-17 expression inhibits cytotoxic functions. However, by allowing expression of activators of the cytotoxic program, this inhibitory mechanism contributes to the instability of IL-17-producing T cells.
Collapse
|
81
|
Cho KA, Park M, Kim YH, Ryu KH, Woo SY. Mesenchymal stem cells inhibit RANK-RANKL interactions between osteoclasts and Th17 cells via osteoprotegerin activity. Oncotarget 2017; 8:83419-83431. [PMID: 29137353 PMCID: PMC5663525 DOI: 10.18632/oncotarget.21379] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 09/05/2017] [Indexed: 11/25/2022] Open
Abstract
Th17 cells play a critical role in several autoimmune diseases, including psoriasis and psoriatic arthritis (PsA). Psoriasis is a chronic inflammatory skin disease associated with systemic inflammation and comorbidities, such as PsA. PsA develops in nearly 70% of patients with psoriasis, and osteoclasts associated bone erosion is a hallmark of the disease. Thus far, the effect of Th17 cells on osteoclastogenesis via direct cell-to-cell interactions is less understood. In this study, we observed that Th17 cells directly promote osteoclast differentiation and maturation via expression of receptor activator of nuclear factor-κ β ligand (RANKL) in vitro. We investigated the impact of conditioned medium obtained from human palatine tonsil-derived mesenchymal stem cells (T-CM) on the interactions between osteoclasts and Th17 cells. T-CM effectively blunted the RANK-RANKL interaction between the osteoclast precursor cell line RAW 264.7 and Th17 cells via osteoprotegerin (OPG) activity. The frequency of tartrate-resistant acid phosphatase (TRAP)-positive cells in the bone marrow of an imiquimod (IMQ)-induced psoriasis mouse model was decreased following T-CM injection. Therefore, our data provide novel insight into the therapeutic potential of tonsil-derived mesenchymal stem cell-mediated therapy (via OPG production) for the treatment of pathophysiologic processes induced by osteoclasts under chronic inflammatory conditions such as psoriasis.
Collapse
Affiliation(s)
- Kyung-Ah Cho
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Minhwa Park
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Yu-Hee Kim
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - So-Youn Woo
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
82
|
Jafarnejad S, Djafarian K, Fazeli MR, Yekaninejad MS, Rostamian A, Keshavarz SA. Effects of a Multispecies Probiotic Supplement on Bone Health in Osteopenic Postmenopausal Women: A Randomized, Double-blind, Controlled Trial. J Am Coll Nutr 2017; 36:497-506. [PMID: 28628374 DOI: 10.1080/07315724.2017.1318724] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE The development of alternative approaches to prevent and/or treat osteoporosis, as a chronic progressive bone disease, is being considered currently. Among dietary supplements, probiotics may have favorable effects on bone metabolism. Therefore, the aim of this study was to evaluate the effects of a multispecies probiotic supplementation on bone biomarkers and bone density in osteopenic postmenopausal women. METHODS This randomized double-blind placebo-controlled clinical trial was performed on 50 patients with osteopenia aged 50-72 years. Participants were randomly assigned to take either a multispecies probiotic supplement (GeriLact; n = 25) or placebo (n = 25) for 6 months. GeriLact contains 7 probiotic bacteria species. Participants received 500 mg Ca plus 200 IU vitamin D daily. Bone mineral density (BMD) of lumbar spine and total hip and blood biomarkers including bone-specific alkaline phosphatase (BALP), osteocalcin (OC), collagen type 1 cross-linked C-telopeptide (CTX), deoxypyridinoline (DPD), parathyroid hormone (PTH), 25-OH vitamin D, and serum pro-inflammatory cytokines (tumor necrosis factor [TNF]-α and interleukin [IL]-1β) were assessed at baseline and at the end of the study. RESULTS The multispecies probiotic significantly decreased BALP (p = 0.03) and CTX (p = 0.04) levels in comparison with the control group but had no effect on BMD of the spine and total hip. Moreover, there was a statistically significant decrease in serum PTH (p = 0.01) and TNF-α (p = 0.02) in the intervention group compared to the placebo group. CONCLUSIONS These results may suggest the favorable effects of the multispecies probiotic supplementation for 6 months on bone health in postmenopausal women due to slowing down the rate of bone turnover.
Collapse
Affiliation(s)
- Sadegh Jafarnejad
- a Department of Clinical Nutrition , School of Nutritional Sciences and Dietetic, Tehran University of Medical Sciences , Tehran , Iran
| | - Kurosh Djafarian
- a Department of Clinical Nutrition , School of Nutritional Sciences and Dietetic, Tehran University of Medical Sciences , Tehran , Iran
| | - Mohammad Reza Fazeli
- b Department of Drug & Food Control , Faculty of Pharmacy and Pharmaceutical Quality Assurance Research Centre, Tehran University of Medical Sciences , Tehran , Iran
| | - Mir Saeed Yekaninejad
- c Department of Epidemiology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Abdolrahman Rostamian
- d Department of Rheumatology , Tehran University of Medical Sciences , Tehran , Iran
| | - Seyed Ali Keshavarz
- a Department of Clinical Nutrition , School of Nutritional Sciences and Dietetic, Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
83
|
Mansour A, Wakkach A, Blin-Wakkach C. Emerging Roles of Osteoclasts in the Modulation of Bone Microenvironment and Immune Suppression in Multiple Myeloma. Front Immunol 2017; 8:954. [PMID: 28848556 PMCID: PMC5554508 DOI: 10.3389/fimmu.2017.00954] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 07/26/2017] [Indexed: 12/26/2022] Open
Abstract
Multiple myeloma (MM) is one of the most common forms of hematologic malignancy resulting from cancerous proliferation of mature malignant plasma cells (MPCs). But despite the real improvement in therapeutics in the past years, it remains largely incurable. MM is the most frequent cancer to involve bone due to the stimulation of osteoclast (OCL) differentiation and activity. OCLs have a unique capacity to resorb bone. However, recent studies reveal that they are not restrained to this sole function. They participate in the control of angiogenesis, medullary niches, and immune responses, including in MM. Therefore, therapeutic approaches targeting OCLs probably affect not only bone resorption but also many other functions, and OCLs should not be considered anymore only as targets to improve the bone phenotype but also to modulate bone microenvironment. In this review, we explore these novel contributions of OCLs to MM which reveal their strong implication in the MM physiopathology. We also underline the therapeutic interest of targeting OCLs not only to overcome bone lesions, but also to improve bone microenvironment and anti-tumoral immune responses.
Collapse
Affiliation(s)
- Anna Mansour
- CNRS, UMR7370, LP2M, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France.,Faculté de Médecine, Université Aix-Marseille, Marseille, France
| | - Abdelilah Wakkach
- CNRS, UMR7370, LP2M, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Claudine Blin-Wakkach
- CNRS, UMR7370, LP2M, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| |
Collapse
|
84
|
Novince CM, Whittow CR, Aartun JD, Hathaway JD, Poulides N, Chavez MB, Steinkamp HM, Kirkwood KA, Huang E, Westwater C, Kirkwood KL. Commensal Gut Microbiota Immunomodulatory Actions in Bone Marrow and Liver have Catabolic Effects on Skeletal Homeostasis in Health. Sci Rep 2017; 7:5747. [PMID: 28720797 PMCID: PMC5515851 DOI: 10.1038/s41598-017-06126-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 06/08/2017] [Indexed: 12/11/2022] Open
Abstract
Despite knowledge the gut microbiota regulates bone mass, mechanisms governing the normal gut microbiota’s osteoimmunomodulatory effects on skeletal remodeling and homeostasis are unclear in the healthy adult skeleton. Young adult specific-pathogen-free and germ-free mice were used to delineate the commensal microbiota’s immunoregulatory effects on osteoblastogenesis, osteoclastogenesis, marrow T-cell hematopoiesis, and extra-skeletal endocrine organ function. We report the commensal microbiota has anti-anabolic effects suppressing osteoblastogenesis and pro-catabolic effects enhancing osteoclastogenesis, which drive bone loss in health. Suppression of Sp7(Osterix) and Igf1 in bone, and serum IGF1, in specific-pathogen-free mice suggest the commensal microbiota’s anti-osteoblastic actions are mediated via local disruption of IGF1-signaling. Differences in the RANKL/OPG Axis in vivo, and RANKL-induced maturation of osteoclast-precursors in vitro, indicate the commensal microbiota induces sustained changes in RANKL-mediated osteoclastogenesis. Candidate mechanisms mediating commensal microbiota’s pro-osteoclastic actions include altered marrow effector CD4+T-cells and a novel Gut-Liver-Bone Axis. The previously unidentified Gut-Liver-Bone Axis intriguingly implies the normal gut microbiota’s osteoimmunomodulatory actions are partly mediated via immunostimulatory effects in the liver. The molecular underpinnings defining commensal gut microbiota immunomodulatory actions on physiologic bone remodeling are highly relevant in advancing the understanding of normal osteoimmunological processes, having implications for the prevention of skeletal deterioration in health and disease.
Collapse
Affiliation(s)
- Chad M Novince
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA.
| | - Carolyn R Whittow
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Johannes D Aartun
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Jessica D Hathaway
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Nicole Poulides
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Michael B Chavez
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Heidi M Steinkamp
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Kaeleigh A Kirkwood
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Emily Huang
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Caroline Westwater
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA.,Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Keith L Kirkwood
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA.,Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| |
Collapse
|
85
|
Collins FL, Schepper JD, Rios-Arce ND, Steury MD, Kang HJ, Mallin H, Schoenherr D, Camfield G, Chishti S, McCabe LR, Parameswaran N. Immunology of Gut-Bone Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1033:59-94. [PMID: 29101652 PMCID: PMC5749247 DOI: 10.1007/978-3-319-66653-2_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent years a link between the gastrointestinal tract and bone health has started to gain significant attention. Dysbiosis of the intestinal microbiota has been linked to the pathology of a number of diseases which are associated with bone loss. In addition modulation of the intestinal microbiota with probiotic bacteria has revealed to have both beneficial local and systemic effects. In the present chapter, we discuss the intestinal and bone immune systems, explore how intestinal disease affects the immune system, and examine how these pathologic changes could adversely impact bone health.
Collapse
Affiliation(s)
- Fraser L Collins
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | | | - Naiomy Deliz Rios-Arce
- Department of Physiology, Michigan State University, East Lansing, MI, USA
- Comparative Medicine and Integrative Biology Program, East Lansing, MI, USA
| | - Michael D Steury
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Ho Jun Kang
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Heather Mallin
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Daniel Schoenherr
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Glen Camfield
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Saima Chishti
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Laura R McCabe
- Department of Physiology and Department of Radiology, Biomedical Imaging Research Centre, Michigan State University, East Lansing, MI, USA.
| | - Narayanan Parameswaran
- Department of Physiology, Michigan State University, East Lansing, MI, USA.
- Comparative Medicine and Integrative Biology Program, East Lansing, MI, USA.
| |
Collapse
|
86
|
Di Rosa F, Watts TH. Editorial: Bone Marrow T Cells at the Center Stage in Immunological Memory. Front Immunol 2016; 7:596. [PMID: 28018359 PMCID: PMC5155117 DOI: 10.3389/fimmu.2016.00596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/30/2016] [Indexed: 01/09/2023] Open
Affiliation(s)
- Francesca Di Rosa
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, c/o Department of Molecular Medicine, Sapienza University , Rome , Italy
| | - Tania H Watts
- Department of Immunology, University of Toronto , Toronto, ON , Canada
| |
Collapse
|
87
|
Abstract
Appreciation of the role of the gut microbiome in regulating vertebrate metabolism has exploded recently. However, the effects of gut microbiota on skeletal growth and homeostasis have only recently begun to be explored. Here, we report that colonization of sexually mature germ-free (GF) mice with conventional specific pathogen-free (SPF) gut microbiota increases both bone formation and resorption, with the net effect of colonization varying with the duration of colonization. Although colonization of adult mice acutely reduces bone mass, in long-term colonized mice, an increase in bone formation and growth plate activity predominates, resulting in equalization of bone mass and increased longitudinal and radial bone growth. Serum levels of insulin-like growth factor 1 (IGF-1), a hormone with known actions on skeletal growth, are substantially increased in response to microbial colonization, with significant increases in liver and adipose tissue IGF-1 production. Antibiotic treatment of conventional mice, in contrast, decreases serum IGF-1 and inhibits bone formation. Supplementation of antibiotic-treated mice with short-chain fatty acids (SCFAs), products of microbial metabolism, restores IGF-1 and bone mass to levels seen in nonantibiotic-treated mice. Thus, SCFA production may be one mechanism by which microbiota increase serum IGF-1. Our study demonstrates that gut microbiota provide a net anabolic stimulus to the skeleton, which is likely mediated by IGF-1. Manipulation of the microbiome or its metabolites may afford opportunities to optimize bone health and growth.
Collapse
|
88
|
Brady RD, Shultz SR, Sun M, Romano T, van der Poel C, Wright DK, Wark JD, O'Brien TJ, Grills BL, McDonald SJ. Experimental Traumatic Brain Injury Induces Bone Loss in Rats. J Neurotrauma 2016; 33:2154-2160. [PMID: 25686841 DOI: 10.1089/neu.2014.3836] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Few studies have investigated the influence of traumatic brain injury (TBI) on bone homeostasis; however, pathophysiological mechanisms involved in TBI have potential to be detrimental to bone. The current study assessed the effect of experimental TBI in rats on the quantity and quality of two different weight-bearing bones, the femur and humerus. Rats were randomly assigned into either sham or lateral fluid percussion injury (FPI) groups. Open-field testing to assess locomotion was conducted at 1, 4, and 12 weeks post-injury, with the rats killed at 1 and 12 weeks post-injury. Bones were analyzed using peripheral quantitative computed tomography (pQCT), histomorphometric analysis, and three-point bending. pQCT analysis revealed that at 1 and 12 weeks post-injury, the distal metaphyseal region of femora from FPI rats had reduced cortical content (10% decrease at 1 week, 8% decrease at 12 weeks; p < 0.01) and cortical thickness (10% decrease at 1 week, 11% decrease at 12 weeks p < 0.001). There was also a 23% reduction in trabecular bone volume ratio at 1 week post-injury and a 27% reduction at 12 weeks post-injury in FPI rats compared to sham (p < 0.001). There were no differences in bone quantity and mechanical properties of the femoral midshaft between sham and TBI animals. There were no differences in locomotor outcomes, which suggested that post-TBI changes in bone were not attributed to immobility. Taken together, these findings indicate that this rat model of TBI was detrimental to bone and suggests a link between TBI and altered bone remodeling.
Collapse
Affiliation(s)
- Rhys D Brady
- 1 Department of Physiology, Anatomy and Microbiology, La Trobe University , Bundoora, VIC, Australia
| | - Sandy R Shultz
- 2 Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne , Parkville, VIC, Australia
| | - Mujun Sun
- 2 Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne , Parkville, VIC, Australia
| | - Tania Romano
- 1 Department of Physiology, Anatomy and Microbiology, La Trobe University , Bundoora, VIC, Australia
| | - Chris van der Poel
- 1 Department of Physiology, Anatomy and Microbiology, La Trobe University , Bundoora, VIC, Australia
| | - David K Wright
- 3 Anatomy and Neuroscience, The University of Melbourne , Parkville, VIC, Australia .,4 The Florey Institute of Neuroscience and Mental Health, The University of Melbourne , Parkville, VIC, Australia
| | - John D Wark
- 2 Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne , Parkville, VIC, Australia
| | - Terence J O'Brien
- 2 Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne , Parkville, VIC, Australia
| | - Brian L Grills
- 1 Department of Physiology, Anatomy and Microbiology, La Trobe University , Bundoora, VIC, Australia
| | - Stuart J McDonald
- 1 Department of Physiology, Anatomy and Microbiology, La Trobe University , Bundoora, VIC, Australia
| |
Collapse
|
89
|
Ibáñez L, Abou-Ezzi G, Ciucci T, Amiot V, Belaïd N, Obino D, Mansour A, Rouleau M, Wakkach A, Blin-Wakkach C. Inflammatory Osteoclasts Prime TNFα-Producing CD4 + T Cells and Express CX 3 CR1. J Bone Miner Res 2016; 31:1899-1908. [PMID: 27161765 DOI: 10.1002/jbmr.2868] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 04/19/2016] [Accepted: 05/06/2016] [Indexed: 11/07/2022]
Abstract
Bone destruction is a hallmark of chronic rheumatic diseases. Although the role of osteoclasts in bone loss is clearly established, their implication in the inflammatory response has not been investigated despite their monocytic origin. Moreover, specific markers are lacking to characterize osteoclasts generated in inflammatory conditions. Here, we have explored the phenotype of inflammatory osteoclasts and their effect on CD4+ T cell responses in the context of bone destruction associated with inflammatory bowel disease. We used the well-characterized model of colitis induced by transfer of naive CD4+ T cells into Rag1-/- mice, which is associated with severe bone destruction. We set up a novel procedure to sort pure osteoclasts generated in vitro to analyze their phenotype and specific immune responses by FACS and qPCR. We demonstrated that osteoclasts generated from colitic mice induced the emergence of TNFα-producing CD4+ T cells, whereas those generated from healthy mice induced CD4+ FoxP3+ regulatory T cells, in an antigen-dependent manner. This difference is related to the osteoclast origin from monocytes or dendritic cells, to their cytokine expression pattern, and their environment. We identified CX3 CR1 as a marker of inflammatory osteoclasts and we demonstrated that the differentiation of CX3 CR1+ osteoclasts is controlled by IL-17 in vitro. This work is the first demonstration that, in addition to participating to bone destruction, osteoclasts also induce immunogenic CD4+ T cell responses upon inflammation. They highlight CX3 CR1 as a novel dual target for antiresorptive and anti-inflammatory treatment in inflammatory chronic diseases. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Lidia Ibáñez
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Grazia Abou-Ezzi
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Thomas Ciucci
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Vanessa Amiot
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Nourhène Belaïd
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Dorian Obino
- INSERM-U932 Immunité et Cancer, Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | - Anna Mansour
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Matthieu Rouleau
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Abdelilah Wakkach
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Claudine Blin-Wakkach
- CNRS, LP2M, UMR7370, Faculté de Médecine, Nice, France. .,Université Nice Sophia Antipolis, Nice, France.
| |
Collapse
|
90
|
Hernandez CJ, Guss JD, Luna M, Goldring SR. Links Between the Microbiome and Bone. J Bone Miner Res 2016; 31:1638-46. [PMID: 27317164 PMCID: PMC5434873 DOI: 10.1002/jbmr.2887] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 12/28/2022]
Abstract
The human microbiome has been shown to influence a number of chronic conditions associated with impaired bone mass and bone quality, including obesity, diabetes, and inflammatory bowel disease. The connection between the microbiome and bone health, however, has not been well studied. The few studies available demonstrate that the microbiome can have a large effect on bone remodeling and bone mass. The gut microbiome is the largest reservoir of microbial organisms in the body and consists of more than a thousand different species interacting with one another in a stable, dynamic equilibrium. How the microbiome can affect organs distant from the gut is not well understood but is believed to occur through regulation of nutrition, regulation of the immune system, and/or translocation of bacterial products across the gut endothelial barrier. Here we review each of these mechanisms and discuss their potential effect on bone remodeling and bone mass. We discuss how preclinical studies of bone-microbiome interactions are challenging because the microbiome is sensitive to genetic background, housing environment, and vendor source. Additionally, although the microbiome exhibits a robust response to external stimuli, it rapidly returns to its original steady state after a disturbance, making it difficult to sustain controlled changes in the microbiome over time periods required to detect alterations in bone remodeling, mass, or structure. Despite these challenges, an understanding of the mechanisms by which the gut microbiome affects bone has the potential to provide insights into the dissociation between fracture risk and bone mineral density in patients including those with obesity, diabetes, or inflammatory bowel disease. In addition, alteration of the gut microbiome has the potential to serve as a biomarker of bone metabolic activity as well as a target for therapies to improve bone structure and quality using pharmaceutical agents or pre- or probiotics. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Christopher J Hernandez
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.,Hospital for Special Surgery, New York, NY, USA
| | - Jason D Guss
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Marysol Luna
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
91
|
Lapérine O, Blin-Wakkach C, Guicheux J, Beck-Cormier S, Lesclous P. Dendritic-cell-derived osteoclasts: a new game changer in bone-resorption-associated diseases. Drug Discov Today 2016; 21:1345-1354. [PMID: 27151158 DOI: 10.1016/j.drudis.2016.04.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/24/2016] [Accepted: 04/26/2016] [Indexed: 12/23/2022]
Abstract
Bone-resorbing cells, osteoclasts (OCs), and antigen-presenting cells, dendritic cells (DCs), share several features. They are derived from a common hematopoietic precursor, exhibit phagocytic activities and their functions are dependent upon receptor activator of nuclear factor κB ligand (RANKL). Upon inflammatory conditions, DCs can transdifferentiate toward functional OCs in the presence of RANKL. It has then been assumed that the increase in proinflammatory cytokines could provide a supportive environment for this transdifferentiation. In this review, we emphasize the molecular mechanisms underlying the potential for DCs to give rise to resorbing OCs in the context of bone-destruction-associated diseases upon inflammatory conditions. Whether these mechanisms reveal new strategies for the discovery of therapeutic targets and drugs is discussed extensively.
Collapse
Affiliation(s)
- Olivier Lapérine
- INSERM, U791, LIOAD, Nantes F-44042, France; Université de Nantes, UMR-S 791, LIOAD, UFR Odontologie, Nantes F-44042, France; ONIRIS, UMR-S 791, LIOAD, Nantes F-44307, France
| | - Claudine Blin-Wakkach
- CNRS, UMR 7370, LP2M, Faculté de médecine, Nice, France; Université Nice Sophia Antipolis, Nice, France
| | - Jérôme Guicheux
- INSERM, U791, LIOAD, Nantes F-44042, France; Université de Nantes, UMR-S 791, LIOAD, UFR Odontologie, Nantes F-44042, France; ONIRIS, UMR-S 791, LIOAD, Nantes F-44307, France; CHU Nantes, PHU 4 OTONN, Nantes F-44042, France.
| | - Sarah Beck-Cormier
- INSERM, U791, LIOAD, Nantes F-44042, France; Université de Nantes, UMR-S 791, LIOAD, UFR Odontologie, Nantes F-44042, France; ONIRIS, UMR-S 791, LIOAD, Nantes F-44307, France
| | - Philippe Lesclous
- INSERM, U791, LIOAD, Nantes F-44042, France; Université de Nantes, UMR-S 791, LIOAD, UFR Odontologie, Nantes F-44042, France; ONIRIS, UMR-S 791, LIOAD, Nantes F-44307, France; CHU Nantes, PHU 4 OTONN, Nantes F-44042, France
| |
Collapse
|
92
|
Di Rosa F, Gebhardt T. Bone Marrow T Cells and the Integrated Functions of Recirculating and Tissue-Resident Memory T Cells. Front Immunol 2016; 7:51. [PMID: 26909081 PMCID: PMC4754413 DOI: 10.3389/fimmu.2016.00051] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/01/2016] [Indexed: 12/15/2022] Open
Abstract
Changes in T cell trafficking accompany the naive to memory T cell antigen-driven differentiation, which remains an incompletely defined developmental step. Upon priming, each naive T cell encounters essential signals – i.e., antigen, co-stimuli and cytokines – in a secondary lymphoid organ; nevertheless, its daughter effector and memory T cells recirculate and receive further signals during their migration through various lymphoid and non-lymphoid organs. These additional signals from tissue microenvironments have an impact on immune response features, including T cell effector function, expansion and contraction, memory differentiation, long-term maintenance, and recruitment upon antigenic rechallenge into local and/or systemic responses. The critical role of T cell trafficking in providing efficient T cell memory has long been a focus of interest. It is now well recognized that naive and memory T cells have different migratory pathways, and that memory T cells are heterogeneous with respect to their trafficking. We and others have observed that, long time after priming, memory T cells are preferentially found in certain niches such as the bone marrow (BM) or at the skin/mucosal site of pathogen entry, even in the absence of residual antigen. The different underlying mechanisms and peculiarities of resulting immunity are currently under study. In this review, we summarize key findings on BM and tissue-resident memory (TRM) T cells and revisit some issues in memory T cell maintenance within such niches. Moreover, we discuss BM seeding by memory T cells in the context of migration patterns and protective functions of either recirculating or TRM T cells.
Collapse
Affiliation(s)
- Francesca Di Rosa
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, c/o Department of Molecular Medicine Sapienza University , Rome , Italy
| | - Thomas Gebhardt
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne, VIC , Australia
| |
Collapse
|
93
|
Sorting nexin 10 acting as a novel regulator of macrophage polarization mediates inflammatory response in experimental mouse colitis. Sci Rep 2016; 6:20630. [PMID: 26856241 PMCID: PMC4746623 DOI: 10.1038/srep20630] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/06/2016] [Indexed: 02/07/2023] Open
Abstract
In response to changes in microenvironment, macrophages polarize into functionally distinct phenotypes, playing a crucial role in the pathogenesis of inflammatory bowel disease (IBD). Here, we investigated the effects of sorting nexin 10 (SNX10), a protein involved in endosomal trafficking and osteoclast maturation, on regulation of macrophage polarization and progression of mouse colitis. Our results revealed that SNX10 deficiency increased the population of M2-type monocytes/macrophages, and protected against colonic inflammation and pathological damage induced by dextran sulfate sodium (DSS). By in vitro study, we showed that deficiency of SNX10 polarized macrophages derived from mouse bone marrow or human peripheral blood mononuclear cells (PBMCs) towards an anti-inflammatory M2 phenotype, which partially reversed by SNX10 plasmid transfection. Adoptive transfer of SNX10−/− macrophages ameliorated colitis in WT mice. However, transfer of WT macrophages exacerbated colitis in SNX10−/− mice. Our data disclose a crucial role and novel function for SNX10 in macrophage polarization. Loss of SNX10 function may be a potential promising therapeutic strategy for IBD.
Collapse
|
94
|
Sugimoto K, Ikeya K, Iida T, Kawasaki S, Arai O, Umehara K, Watanabe F, Tani S, Oishi S, Osawa S, Yamamoto T, Hanai H. An Increased Serum N-Terminal Telopeptide of Type I Collagen, a Biochemical Marker of Increased Bone Resorption, Is Associated with Infliximab Therapy in Patients with Crohn's Disease. Dig Dis Sci 2016; 61:99-106. [PMID: 26254083 DOI: 10.1007/s10620-015-3838-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 07/30/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Osteopenia and osteoporosis are considered to be extra-intestinal manifestations of inflammatory bowel disease (IBD). Anti-tumor necrosis factor (TNF)-α biologics have been introduced as novel medications for an active IBD. However, it is still not well documented whether anti-TNF-α affects the frequency of bone loss or abnormality of bone mineral markers among patients with IBD. AIMS This study was to investigate the biochemical basis of low bone mineral density (BMD) and increased turnover in IBD during infliximab (IFX) therapy. METHODS Forty patients with Crohn's disease (CD), 80 patients with ulcerative colitis (UC), and 65 age- and gender-matched controls were included. BMD was measured with dual-energy X-ray absorptiometry, and vitamins K and D were measured as serum undercarboxylated osteocalcin (ucOC) and 1,25-(OH)2D, respectively. Bone formation and resorption were based on measuring bone-specific alkaline phosphatase (BAP) and serum N-terminal telopeptide of type I collagen (NTx), respectively. RESULTS Significantly lower BMD was found in patients with UC and CD as compared to controls (P < 0.05). BAP, 1,25-(OH)2D, ucOC, and NTx were significantly higher in CD patients, but not in UC patients as compared to controls (P < 0.05). Further, serum NTx level was significantly higher in CD patients who were receiving IFX as compared to CD patients who were not receiving IFX (P < 0.01). CONCLUSIONS A lower BMD and higher bone metabolism markers were found in CD patients as compared to controls or UC patients. A significant increased serum level of NTx, a biochemical marker of increased bone resorption, was observed in CD patients during IFX therapy.
Collapse
Affiliation(s)
- Ken Sugimoto
- First Department of Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan.
| | - Kentaro Ikeya
- Centre for Gastroenterology and Inflammatory Bowel Disease Research, Hamamatsu South Hospital, Hamamatsu, 430-0846, Japan
| | - Takayuki Iida
- Centre for Gastroenterology and Inflammatory Bowel Disease Research, Hamamatsu South Hospital, Hamamatsu, 430-0846, Japan
| | - Shinsuke Kawasaki
- Centre for Gastroenterology and Inflammatory Bowel Disease Research, Hamamatsu South Hospital, Hamamatsu, 430-0846, Japan
| | - Osamu Arai
- Centre for Gastroenterology and Inflammatory Bowel Disease Research, Hamamatsu South Hospital, Hamamatsu, 430-0846, Japan
| | - Keita Umehara
- Orthopedics Unit, Hamamatsu South Hospital, Hamamatsu, 430-0846, Japan
| | - Fumitoshi Watanabe
- Centre for Gastroenterology and Inflammatory Bowel Disease Research, Hamamatsu South Hospital, Hamamatsu, 430-0846, Japan
| | - Shinya Tani
- First Department of Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Shinji Oishi
- First Department of Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Satoshi Osawa
- Department of Endoscopic and Photodynamic Medicine, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Takayuki Yamamoto
- Inflammatory Bowel Disease Centre, Yokkaichi Hazu Medical Centre, Mie, 510-0016, Japan
| | - Hiroyuki Hanai
- Centre for Gastroenterology and Inflammatory Bowel Disease Research, Hamamatsu South Hospital, Hamamatsu, 430-0846, Japan
| |
Collapse
|
95
|
Wakkach A, Rouleau M, Blin-Wakkach C. Osteoimmune Interactions in Inflammatory Bowel Disease: Central Role of Bone Marrow Th17 TNFα Cells in Osteoclastogenesis. Front Immunol 2015; 6:640. [PMID: 26734007 PMCID: PMC4683185 DOI: 10.3389/fimmu.2015.00640] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/04/2015] [Indexed: 12/18/2022] Open
Abstract
Osteoimmunology is an interdisciplinary research field dedicated to the study of the crosstalk between the immune and bone systems. CD4+ T cells are central players in this crosstalk. There is an emerging understanding that CD4+ T cells play an important role in the bone marrow (BM) under physiological and pathological conditions and modulate the differentiation of bone-resorbing osteoclasts. However, identification of the mechanisms that maintain CD4+ T cells in the BM is still a matter of investigation. This article describes the CD4+ T cell populations of the BM and reviews their role as osteoclastogenic population in inflammatory bowel disease.
Collapse
Affiliation(s)
- Abdelilah Wakkach
- CNRS, UMR 7370, Laboratoire de PhysioMédecine Moléculaire (LP2M), Faculté de Médecine, Nice, France; University Nice Sophia Antipolis, Nice, France
| | - Matthieu Rouleau
- CNRS, UMR 7370, Laboratoire de PhysioMédecine Moléculaire (LP2M), Faculté de Médecine, Nice, France; University Nice Sophia Antipolis, Nice, France
| | - Claudine Blin-Wakkach
- CNRS, UMR 7370, Laboratoire de PhysioMédecine Moléculaire (LP2M), Faculté de Médecine, Nice, France; University Nice Sophia Antipolis, Nice, France
| |
Collapse
|
96
|
Syrbe U, Siegmund B. Bone marrow Th17 TNFα cells induce osteoclast differentiation and link bone destruction to IBD. Gut 2015; 64:1011-2. [PMID: 25391836 DOI: 10.1136/gutjnl-2014-308436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 10/28/2014] [Indexed: 02/01/2023]
Affiliation(s)
- Uta Syrbe
- Department of Medicine (Gastroenterology, Infectious Diseases, Rheumatology), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Britta Siegmund
- Department of Medicine (Gastroenterology, Infectious Diseases, Rheumatology), Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
97
|
Bone Loss Triggered by the Cytokine Network in Inflammatory Autoimmune Diseases. J Immunol Res 2015; 2015:832127. [PMID: 26065006 PMCID: PMC4434203 DOI: 10.1155/2015/832127] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 12/24/2014] [Accepted: 12/26/2014] [Indexed: 01/14/2023] Open
Abstract
Bone remodeling is a lifelong process in vertebrates that relies on the correct balance between bone resorption by osteoclasts and bone formation by osteoblasts. Bone loss and fracture risk are implicated in inflammatory autoimmune diseases such as rheumatoid arthritis, ankylosing spondylitis, inflammatory bowel disease, and systemic lupus erythematosus. The network of inflammatory cytokines produced during chronic inflammation induces an uncoupling of bone formation and resorption, resulting in significant bone loss in patients with inflammatory autoimmune diseases. Here, we review and discuss the involvement of the inflammatory cytokine network in the pathophysiological aspects and the therapeutic advances in inflammatory autoimmune diseases.
Collapse
|