1
|
Abbasifard M, Khorramdelazad H. Harmonizing hope: navigating the osteoarthritis melody through the CCL2/CCR2 axis for innovative therapeutic avenues. Front Immunol 2024; 15:1387651. [PMID: 39076996 PMCID: PMC11284107 DOI: 10.3389/fimmu.2024.1387651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
Osteoarthritis (OA) is characterized by a complex interplay of molecular signals orchestrated by the CCL2/CCR2 axis. The pathogenesis of OA has been revealed to be influenced by a multifaceted effect of CCL2/CCR2 signaling on inflammation, cartilage degradation, and joint homeostasis. The CCL2/CCR2 axis promotes immune cell recruitment and tips the balance toward degeneration by influencing chondrocyte behavior. Insights into these intricate pathways will offer novel therapeutic approaches, paving the way for targeted interventions that may redefine OA management in the future. This review article explores the molecular symphony through the lens of the CCL2/CCR2 axis, providing a harmonious blend of current knowledge and future directions on OA treatment. Furthermore, in this study, through a meticulous review of recent research, the key players and molecular mechanisms that amplify the catabolic cascade within the joint microenvironment are identified, and therapeutic approaches to targeting the CCL2/CCR axis are discussed.
Collapse
Affiliation(s)
- Mitra Abbasifard
- Department of Internal Medicine, School of Medicine, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
2
|
Li N, Oh JH, Suh JH, Jin SP, Lee DH, Lee Y, Chung JH. Impact of fucosyltransferase 1-mediated epidermal blood group antigen H on anti-inflammatory response in atopic dermatitis. Front Immunol 2024; 15:1365430. [PMID: 38840912 PMCID: PMC11151169 DOI: 10.3389/fimmu.2024.1365430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024] Open
Abstract
The presence of the blood group H2 antigen on the membrane of red blood cells determines blood type O in individuals and this H2 antigen serves as a precursor to the A and B antigens expressed in blood types A and B, respectively. However, the specific involvement of ABH antigens in skin diseases is unknown. Therefore, we aim to investigate the expression of ABH antigens in skin tissue of patients with atopic dermatitis (AD) and MC903-induced AD-like mice. We demonstrated that the expression of ABH antigen is primarily located in the granular and horny layers of the skin in healthy control individuals. However, in patients with AD, the expression of the ABH antigen was absent or diminished in these layers, while the H2 antigen expression increased in the spinous layers of the affected skin lesions. Then, we investigated the biological function of blood group H antigen mediated by fucosyltransferase 1 (Fut1) in the skin, utilizing an AD mouse model induced by MC903 in wild-type (WT) and Fut1-knockout mice. After the application of MC903, Fut1-deficient mice, with no H2 antigen expression on their skin, exhibited more severe clinical signs, increased ear swelling, and elevated serum IgE levels compared with those of WT mice. Additionally, the MC903-induced thickening of both the epidermis and dermis was more pronounced in Fut1-deficient mice than that in WT mice. Furthermore, Fut1-deficient mice showed a significantly higher production of interleukin-4 (IL-4) and IL-6 in skin lesions compared with that of their WT counterparts. The expression of chemokines, particularly Ccl2 and Ccl8, was notably higher in Fut1-deficient mice compared with those of WT mice. The infiltration of CD4+ T cells, eosinophils, and mast cells into the lesional skin was significantly elevated in Fut1-deficient mice compared with that in WT mice. These findings demonstrate the protective role of H2 antigen expression against AD-like inflammation and highlight its potential therapeutic impact on AD through the regulation of blood group antigens.
Collapse
Affiliation(s)
- Na Li
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Jang-Hee Oh
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Joong Heon Suh
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Seon-Pil Jin
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Youngae Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
- Institute on Aging, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Ma Y, Shi R, Li F, Chang H. Emerging strategies for treating autoimmune disease with genetically modified dendritic cells. Cell Commun Signal 2024; 22:262. [PMID: 38715122 PMCID: PMC11075321 DOI: 10.1186/s12964-024-01641-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/28/2024] [Indexed: 05/12/2024] Open
Abstract
Gene editing of living cells has become a crucial tool in medical research, enabling scientists to address fundamental biological questions and develop novel strategies for disease treatment. This technology has particularly revolutionized adoptive transfer cell therapy products, leading to significant advancements in tumor treatment and offering promising outcomes in managing transplant rejection, autoimmune disorders, and inflammatory diseases. While recent clinical trials have demonstrated the safety of tolerogenic dendritic cell (TolDC) immunotherapy, concerns remain regarding its effectiveness. This review aims to discuss the application of gene editing techniques to enhance the tolerance function of dendritic cells (DCs), with a particular focus on preclinical strategies that are currently being investigated to optimize the tolerogenic phenotype and function of DCs. We explore potential approaches for in vitro generation of TolDCs and provide an overview of emerging strategies for modifying DCs. Additionally, we highlight the primary challenges hindering the clinical adoption of TolDC therapeutics and propose future research directions in this field.
Collapse
Affiliation(s)
- Yunhan Ma
- School of Medicine, Jiangsu University, Zhenjiang, 212000, China
| | - Ruobing Shi
- School of Medicine, Jiangsu University, Zhenjiang, 212000, China
| | - Fujun Li
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Haocai Chang
- MOE Key Laboratory of Laser Life Science, Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
4
|
Li C, Wang L, Zhang K, Wang Z, Li Z, Li Z, Chen L. Overcoming neutrophil-induced immunosuppression in postoperative cancer therapy: Combined sialic acid-modified liposomes with scaffold-based vaccines. Asian J Pharm Sci 2024; 19:100906. [PMID: 38595333 PMCID: PMC11002593 DOI: 10.1016/j.ajps.2024.100906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/13/2024] [Accepted: 02/17/2024] [Indexed: 04/11/2024] Open
Abstract
Immunotherapy is a promising approach for preventing postoperative tumor recurrence and metastasis. However, inflammatory neutrophils, recruited to the postoperative tumor site, have been shown to exacerbate tumor regeneration and limit the efficacy of cancer vaccines. Consequently, addressing postoperative immunosuppression caused by neutrophils is crucial for improving treatment outcomes. This study presents a combined chemoimmunotherapeutic strategy that employs a biocompatible macroporous scaffold-based cancer vaccine (S-CV) and a sialic acid (SA)-modified, doxorubicin (DOX)-loaded liposomal platform (DOX@SAL). The S-CV contains whole tumor lysates as antigens and imiquimod (R837, Toll-like receptor 7 activator)-loaded PLGA nanoparticles as immune adjuvants for cancer, which enhance dendritic cell activation and cytotoxic T cell proliferation upon localized implantation. When administered intravenously, DOX@SAL specifically targets and delivers drugs to activated neutrophils in vivo, mitigating neutrophil infiltration and suppressing postoperative inflammatory responses. In vivo and vitro experiments have demonstrated that S-CV plus DOX@SAL, a combined chemo-immunotherapeutic strategy, has a remarkable potential to inhibit postoperative local tumor recurrence and distant tumor progression, with minimal systemic toxicity, providing a new concept for postoperative treatment of tumors.
Collapse
Affiliation(s)
- Cong Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Lihong Wang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Kexin Zhang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Zeyu Wang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Zhihang Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Zehao Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Lijiang Chen
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| |
Collapse
|
5
|
Rodríguez Mesa XM, Contreras Bolaños LA, Modesti Costa G, Mejia AL, Santander González SP. A Bidens pilosa L. Non-Polar Extract Modulates the Polarization of Human Macrophages and Dendritic Cells into an Anti-Inflammatory Phenotype. Molecules 2023; 28:7094. [PMID: 37894572 PMCID: PMC10608814 DOI: 10.3390/molecules28207094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/03/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Different communities around the world traditionally use Bidens pilosa L. for medicinal purposes, mainly for its anti-inflammatory, antinociceptive, and antioxidant properties; it is used as an ingredient in teas or herbal medicines for the treatment of pain, inflammation, and immunological disorders. Several studies have been conducted that prove the immunomodulatory properties of this plant; however, it is not known whether the immunomodulatory properties of B. pilosa are mediated by its ability to modulate antigen-presenting cells (APCs) such as macrophages (MØs) and dendritic cells (DCs) (through polarization or the maturation state, respectively). Different polar and non-polar extracts and fractions were prepared from the aerial part of B. pilosa. Their cytotoxic and immunomodulatory effects were first tested on human peripheral blood mononuclear cells (PBMCs) and phytohemagglutinin (PHA)-stimulated PBMCs, respectively, via an MTT assay. Then, the non-cytotoxic plant extracts and fractions that showed the highest immunomodulatory activity were selected to evaluate their effects on human MØ polarization and DC maturation (cell surface phenotype and cytokine secretion) through multiparametric flow cytometry. Finally, the chemical compounds of the B. pilosa extract that showed the most significant immunomodulatory effects on human APCs were identified using gas chromatography coupled with mass spectrometry. The petroleum ether extract and the ethyl acetate and hydroalcoholic fractions obtained from B. pilosa showed low cytotoxicity and modulated the PHA-stimulated proliferation of PBMCs. Furthermore, the B. pilosa petroleum ether extract induced M2 polarization or a hybrid M1/M2 phenotype in MØs and a semi-mature status in DCs, regardless of exposure to a maturation stimulus. The immunomodulatory activity of the non-polar (petroleum ether) extract of B. pilosa on human PBMC proliferation, M2 polarization of MØs, and semi-mature status in DCs might be attributed to the low-medium polarity components in the extract, such as phytosterol terpenes and fatty acid esters.
Collapse
Affiliation(s)
| | | | - Geison Modesti Costa
- Phytochemistry Research Group (GIFUJ), Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Antonio Luis Mejia
- Phytoimmunomodulation Research Group, Juan N. Corpas University Foundation, Bogotá 111161, Colombia
| | | |
Collapse
|
6
|
Brown H, Komnick MR, Brigleb PH, Dermody TS, Esterházy D. Lymph node sharing between pancreas, gut, and liver leads to immune crosstalk and regulation of pancreatic autoimmunity. Immunity 2023; 56:2070-2085.e11. [PMID: 37557168 PMCID: PMC11040372 DOI: 10.1016/j.immuni.2023.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 05/03/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023]
Abstract
Lymph nodes (LNs) are critical sites for shaping tissue-specific adaptive immunity. However, the impact of LN sharing between multiple organs on such tailoring is less understood. Here, we describe the drainage hierarchy of the pancreas, liver, and the upper small intestine (duodenum) into three murine LNs. Migratory dendritic cells (migDCs), key in instructing adaptive immune outcome, exhibited stronger pro-inflammatory signatures when originating from the pancreas or liver than from the duodenum. Qualitatively different migDC mixing in each shared LN influenced pancreatic β-cell-reactive T cells to acquire gut-homing and tolerogenic phenotypes proportional to duodenal co-drainage. However, duodenal viral infections rendered non-intestinal migDCs and β-cell-reactive T cells more pro-inflammatory in all shared LNs, resulting in elevated pancreatic islet lymphocyte infiltration. Our study uncovers immune crosstalk through LN co-drainage as a powerful force regulating pancreatic autoimmunity.
Collapse
Affiliation(s)
- Hailey Brown
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Macy R Komnick
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Pamela H Brigleb
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Terence S Dermody
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Daria Esterházy
- Department of Pathology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
7
|
ElGindi M, Sapudom J, Garcia Sabate A, Chesney Quartey B, Alatoom A, Al-Sayegh M, Li R, Chen W, Teo J. Effects of an aged tissue niche on the immune potency of dendritic cells using simulated microgravity. NPJ AGING 2023; 9:14. [PMID: 37393393 DOI: 10.1038/s41514-023-00111-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/18/2023] [Indexed: 07/03/2023]
Abstract
Microgravity accelerates the aging of various physiological systems, and it is well acknowledged that aged individuals and astronauts both have increased susceptibility to infections and poor response to vaccination. Immunologically, dendritic cells (DCs) are the key players in linking innate and adaptive immune responses. Their distinct and optimized differentiation and maturation phases play a critical role in presenting antigens and mounting effective lymphocyte responses for long-term immunity. Despite their importance, no studies to date have effectively investigated the effects of microgravity on DCs in their native microenvironment, which is primarily located within tissues. Here, we address a significantly outstanding research gap by examining the effects of simulated microgravity via a random positioning machine on both immature and mature DCs cultured in biomimetic collagen hydrogels, a surrogate for tissue matrices. Furthermore, we explored the effects of loose and dense tissues via differences in collagen concentration. Under these various environmental conditions, the DC phenotype was characterized using surface markers, cytokines, function, and transcriptomic profiles. Our data indicate that aged or loose tissue and exposure to RPM-induced simulated microgravity both independently alter the immunogenicity of immature and mature DCs. Interestingly, cells cultured in denser matrices experience fewer effects of simulated microgravity at the transcriptome level. Our findings are a step forward to better facilitate healthier future space travel and enhance our understanding of the aging immune system on Earth.
Collapse
Affiliation(s)
- Mei ElGindi
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Anna Garcia Sabate
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Brian Chesney Quartey
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Aseel Alatoom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Rui Li
- Department of Biomedical Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| | - Weiqiang Chen
- Department of Biomedical Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
- Department of Mechanical and Aerospace Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| | - Jeremy Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates.
- Department of Biomedical Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA.
- Department of Mechanical and Aerospace Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA.
| |
Collapse
|
8
|
Uwazie CC, Faircloth TU, Parr RN, Reddy YU, Hematti P, Rajan D, Chinnadurai R. Contrariety of Human Bone Marrow Mesenchymal Stromal Cell Functionality in Modulating Circulatory Myeloid and Plasmacytoid Dendritic Cell Subsets. BIOLOGY 2023; 12:biology12050725. [PMID: 37237538 DOI: 10.3390/biology12050725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023]
Abstract
Mesenchymal Stromal Cells (MSCs) derived from bone marrow are widely tested in clinical trials as a cellular therapy for potential inflammatory disorders. The mechanism of action of MSCs in mediating immune modulation is of wide interest. In the present study, we investigated the effect of human bone-marrow-derived MSCs in modulating the circulating peripheral blood dendritic cell responses through flow cytometry and multiplex secretome technology upon their coculture ex vivo. Our results demonstrated that MSCs do not significantly modulate the responses of plasmacytoid dendritic cells. However, MSCs dose-dependently promote the maturation of myeloid dendritic cells. Mechanistic analysis showed that dendritic cell licensing cues (Lipopolysaccharide and Interferon-gamma) stimulate MSCs to secret an array of dendritic cell maturation-associated secretory factors. We also identified that MSC-mediated upregulation of myeloid dendritic cell maturation is associated with the unique predictive secretome signature. Overall, the present study demonstrated the dichotomy of MSC functionality in modulating myeloid and plasmacytoid dendritic cells. This study provides clues that clinical trials need to investigate if circulating dendritic cell subsets in MSC therapy can serve as potency biomarkers.
Collapse
Affiliation(s)
- Crystal C Uwazie
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, USA
| | - Tyler U Faircloth
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, USA
| | - Rhett N Parr
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, USA
| | - Yenamala U Reddy
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, USA
| | - Peiman Hematti
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Devi Rajan
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, USA
| | - Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, USA
| |
Collapse
|
9
|
Lin Z, Shi JL, Chen M, Zheng ZM, Li MQ, Shao J. CCL2: An important cytokine in normal and pathological pregnancies: A review. Front Immunol 2023; 13:1053457. [PMID: 36685497 PMCID: PMC9852914 DOI: 10.3389/fimmu.2022.1053457] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
C-C motif ligand 2 (CCL2), also known as monocytic chemotactic protein 1 (MCP-1), is an integral chemotactic factor which recruits macrophages for the immune response. Together with its receptors (e.g., CCR2, ACKR1, and ACKR2), they exert noticeable influences on various diseases of different systems. At the maternal-fetal interface, CCL2 is detected to be expressed in trophoblasts, decidual tissue, the myometrium, and others. Meanwhile, existing reports have determined a series of physiological regulators of CCL2, which functions in maintaining normal recruitment of immunocytes, tissue remodeling, and angiogenesis. However, abnormal levels of CCL2 have also been reported to be associated with adverse pregnancy outcomes such as spontaneous abortion, preeclampsia and preterm labor. In this review, we concentrate on CCL2 expression at the maternal-fetal interface, as well as its precise regulatory mechanisms and classic signaling pathways, to reveal the multidimensional aspects of CCL2 in pregnancy.
Collapse
Affiliation(s)
- Zhi Lin
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jia-Lu Shi
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Min Chen
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Zi-Meng Zheng
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
- National Health Commision (NHC) Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Jun Shao
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Johnson CS, Cook LM. Osteoid cell-derived chemokines drive bone-metastatic prostate cancer. Front Oncol 2023; 13:1100585. [PMID: 37025604 PMCID: PMC10070788 DOI: 10.3389/fonc.2023.1100585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
One of the greatest challenges in improving prostate cancer (PCa) survival is in designing new therapies to effectively target bone metastases. PCa regulation of the bone environment has been well characterized; however, bone-targeted therapies have little impact on patient survival, demonstrating a need for understanding the complexities of the tumor-bone environment. Many factors contribute to creating a favorable microenvironment for prostate tumors in bone, including cell signaling proteins produced by osteoid cells. Specifically, there has been extensive evidence from both past and recent studies that emphasize the importance of chemokine signaling in promoting PCa progression in the bone environment. Chemokine-focused strategies present promising therapeutic options for treating bone metastasis. These signaling pathways are complex, with many being produced by (and exerting effects on) a plethora of different cell types, including stromal and tumor cells of the prostate tumor-bone microenvironment. This review highlights an underappreciated molecular family that should be interrogated for treatment of bone metastatic prostate cancer (BM-PCa).
Collapse
Affiliation(s)
- Catherine S. Johnson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE, United States
| | - Leah M. Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Leah M. Cook,
| |
Collapse
|
11
|
Che Y, He J, Li X, Wu D, Zhang Y, Yuan G. Overexpression of microRNA-381-3p ameliorates hypoxia/ischemia-induced neuronal damage and microglial inflammation via regulating the C-C chemokine receptor type 2 /nuclear transcription factor-kappa B axis. Bioengineered 2022; 13:6839-6855. [PMID: 35246016 PMCID: PMC8973660 DOI: 10.1080/21655979.2022.2038448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
microRNAs, as small endogenous RNAs, influence umpteen sophisticated cellular biological functions regarding neurodegenerative and cerebrovascular diseases. Here, we interrogated miR-381-3p’s influence on BV2 activation and neurotoxicity in ischemic and hypoxic environment. Oxygen-glucose deprivation (OGD) was adopted to induce microglial activation and HT-22 neuron damage. Quantitative polymerase chain reaction (qRT-PCR) was taken to check miR-381-3p expression in OGD-elicited BV2 cells and HT-22 neurons. It transpired that miR-381-3p expression was lowered in BV2 cells and HT-22 cells elicited by OGD. miR-381-3p up-regulation remarkably hampered inflammatory mediator expression in BV2 cells induced by OGD and weakened HT22 neuron apoptosis. In vivo, miR-381-3p expression was abated in HI rats’ ischemic lesions, and miR-381-3p up-regulation could ameliorate inflammation and neuron apoptosis in their brain. C-C chemokine receptor type 2 (CCR2) was identified as the downstream target of miR-381-3p, and miR-381-3p suppressed the CCR2/NF-κB pathway to mitigate microglial activation and neurotoxicity. Therefore, we believed that miR-381-3p overexpression exerts anti-inflammation and anti-apoptosis in ischemic brain injury by targeting CCR2
Collapse
Affiliation(s)
- Yuanmei Che
- Department of Infection, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianglong He
- Department of Infection, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaopeng Li
- Department of Infection, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Daxian Wu
- Department of Infection, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yi Zhang
- Department of Infection, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guicai Yuan
- Department of Infection, The Second Affiliated Hospital of Yichun University, Yichun, China
| |
Collapse
|
12
|
Song YC, Liu CT, Lee HJ, Yen HR. Cordycepin prevents and ameliorates experimental autoimmune encephalomyelitis by inhibiting leukocyte infiltration and reducing neuroinflammation. Biochem Pharmacol 2022; 197:114918. [DOI: 10.1016/j.bcp.2022.114918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/30/2022]
|
13
|
To KKW, Sridhar S, Chiu KHY, Hung DLL, Li X, Hung IFN, Tam AR, Chung TWH, Chan JFW, Zhang AJX, Cheng VCC, Yuen KY. Lessons learned 1 year after SARS-CoV-2 emergence leading to COVID-19 pandemic. Emerg Microbes Infect 2021; 10:507-535. [PMID: 33666147 PMCID: PMC8006950 DOI: 10.1080/22221751.2021.1898291] [Citation(s) in RCA: 170] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 02/26/2021] [Accepted: 02/28/2021] [Indexed: 02/06/2023]
Abstract
Without modern medical management and vaccines, the severity of the Coronavirus Disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2) might approach the magnitude of 1894-plague (12 million deaths) and 1918-A(H1N1) influenza (50 million deaths) pandemics. The COVID-19 pandemic was heralded by the 2003 SARS epidemic which led to the discovery of human and civet SARS-CoV-1, bat SARS-related-CoVs, Middle East respiratory syndrome (MERS)-related bat CoV HKU4 and HKU5, and other novel animal coronaviruses. The suspected animal-to-human jumping of 4 betacoronaviruses including the human coronaviruses OC43(1890), SARS-CoV-1(2003), MERS-CoV(2012), and SARS-CoV-2(2019) indicates their significant pandemic potential. The presence of a large reservoir of coronaviruses in bats and other wild mammals, culture of mixing and selling them in urban markets with suboptimal hygiene, habit of eating exotic mammals in highly populated areas, and the rapid and frequent air travels from these areas are perfect ingredients for brewing rapidly exploding epidemics. The possibility of emergence of a hypothetical SARS-CoV-3 or other novel viruses from animals or laboratories, and therefore needs for global preparedness should not be ignored. We reviewed representative publications on the epidemiology, virology, clinical manifestations, pathology, laboratory diagnostics, treatment, vaccination, and infection control of COVID-19 as of 20 January 2021, which is 1 year after person-to-person transmission of SARS-CoV-2 was announced. The difficulties of mass testing, labour-intensive contact tracing, importance of compliance to universal masking, low efficacy of antiviral treatment for severe disease, possibilities of vaccine or antiviral-resistant virus variants and SARS-CoV-2 becoming another common cold coronavirus are discussed.
Collapse
Affiliation(s)
- Kelvin Kai-Wang To
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Siddharth Sridhar
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Kelvin Hei-Yeung Chiu
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Derek Ling-Lung Hung
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Xin Li
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Anthony Raymond Tam
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Tom Wai-Hin Chung
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Anna Jian-Xia Zhang
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Vincent Chi-Chung Cheng
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| |
Collapse
|
14
|
Lauerma A, Werner P, Wisgrill L, Fyhrquist N. New Key Players in Irritant Contact Dermatitis: Residential Skin Cells and Neutrophils Drive Inflammation. J Invest Dermatol 2021; 142:509-512. [PMID: 34749986 DOI: 10.1016/j.jid.2021.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 11/29/2022]
Abstract
The chemokine CCL2 is a potential biomarker for progression of inflammatory skin disease. In a new article of the Journal of Investigative Dermatology, Shibuya et al. (2021) use murine experimental models to show that CCL2‒CCR2‒dependent IL-1β secretion by local skin cells and skin-infiltrating neutrophils are key drivers of skin irritation.
Collapse
Affiliation(s)
- Antti Lauerma
- Department of Dermatology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.
| | - Paulina Werner
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lukas Wisgrill
- Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescence Medicine, Medical University of Vienna, Vienna, Austria
| | - Nanna Fyhrquist
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Pallazola AM, Rao JX, Mengistu DT, Morcos MS, Toma MS, Stolberg VR, Tretyakova A, McCloskey L, Curtis JL, Freeman CM. Human lung cDC1 drive increased perforin-mediated NK cytotoxicity in Chronic Obstructive Pulmonary Disease. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1183-L1193. [PMID: 34704847 PMCID: PMC8715029 DOI: 10.1152/ajplung.00322.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
In chronic obstructive pulmonary disease (COPD), lung natural killer cells (NKs) lyse autologous lung epithelial cells in vitro, but underlying mechanisms and their relationship to epithelial cell apoptosis in vivo are undefined. Although this cytolytic capacity of lung NKs depends on priming by dendritic cells (DC), whether priming correlates with DC maturation or is limited to a specific DC subset are also unknown. We recruited ever-smokers (≥10 pack-years) (n=96) undergoing clinically-indicated lung resections. We analyzed lung NKs for cytotoxic molecule transcripts and for cytotoxicity, which we correlated with in situ detection of activated Caspase-3/7+ airway epithelial cells. To investigate DC priming, we measured lung DC expression of CCR2, CCR7, and CX3CR1, and co-cultured peripheral blood NKs with autologous lung DC, either matured using LPS (non-obstructed smokers) or separated into conventional DC type-1 (cDC1) versus cDC type-2 (cDC2) (COPD). Lung NKs in COPD expressed more perforin (p<0.02) and granzyme B (p<0.03) transcripts; inhibiting perforin blocked in vitro killing by lung NKs. Cytotoxicity in vitro correlated significantly (Sr=0.68, p=0.0043) with numbers of apoptotic epithelial cells per airway. In non-obstructed smokers, LPS-induced maturation enhanced DC-mediated priming of blood NKs, reflected by greater epithelial cell death. Although CCR7 expression was greater in COPD in both cDC1 (p<0.03) and cDC2 (p=0.009), only lung cDC1 primed NK killing. Thus, rather than being intrinsic to those with COPD, NK priming is a capacity of human lung DC that is inducible by recognition of bacterial (and possibly other) danger signals and restricted to the cDC1 subset.
Collapse
Affiliation(s)
- Alexander M Pallazola
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School and Michigan Medicine, Ann Arbor, MI, United States
| | - Jessica X Rao
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School and Michigan Medicine, Ann Arbor, MI, United States
| | - Dawit T Mengistu
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, United States
| | - Maria S Morcos
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School and Michigan Medicine, Ann Arbor, MI, United States
| | - Mariam S Toma
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School and Michigan Medicine, Ann Arbor, MI, United States
| | - Valerie R Stolberg
- Research Service, VA Ann Arbor Healthcare System, Ann Arbor, MI, United States
| | - Alexandra Tretyakova
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School and Michigan Medicine, Ann Arbor, MI, United States
| | - Lisa McCloskey
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School and Michigan Medicine, Ann Arbor, MI, United States
| | - Jeffrey L Curtis
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School and Michigan Medicine, Ann Arbor, MI, United States.,Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, United States.,Pulmonary and Critical Care Medicine Section, VA Ann Arbor Healthcare System, Ann Arbor, MI, United States
| | - Christine M Freeman
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School and Michigan Medicine, Ann Arbor, MI, United States.,Research Service, VA Ann Arbor Healthcare System, Ann Arbor, MI, United States.,Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
16
|
Mysore V, Cullere X, Mears J, Rosetti F, Okubo K, Liew PX, Zhang F, Madera-Salcedo I, Rosenbauer F, Stone RM, Aster JC, von Andrian UH, Lichtman AH, Raychaudhuri S, Mayadas TN. FcγR engagement reprograms neutrophils into antigen cross-presenting cells that elicit acquired anti-tumor immunity. Nat Commun 2021; 12:4791. [PMID: 34373452 PMCID: PMC8352912 DOI: 10.1038/s41467-021-24591-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Classical dendritic cells (cDC) are professional antigen-presenting cells (APC) that regulate immunity and tolerance. Neutrophil-derived cells with properties of DCs (nAPC) are observed in human diseases and after culture of neutrophils with cytokines. Here we show that FcγR-mediated endocytosis of antibody-antigen complexes or an anti-FcγRIIIB-antigen conjugate converts neutrophils into nAPCs that, in contrast to those generated with cytokines alone, activate T cells to levels observed with cDCs and elicit CD8+ T cell-dependent anti-tumor immunity in mice. Single cell transcript analyses and validation studies implicate the transcription factor PU.1 in neutrophil to nAPC conversion. In humans, blood nAPC frequency in lupus patients correlates with disease. Moreover, anti-FcγRIIIB-antigen conjugate treatment induces nAPCs that can activate autologous T cells when using neutrophils from individuals with myeloid neoplasms that harbor neoantigens or those vaccinated against bacterial toxins. Thus, anti-FcγRIIIB-antigen conjugate-induced conversion of neutrophils to immunogenic nAPCs may represent a possible immunotherapy for cancer and infectious diseases.
Collapse
Affiliation(s)
- Vijayashree Mysore
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Xavier Cullere
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Joseph Mears
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Immunology, Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Florencia Rosetti
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Koshu Okubo
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Pei X Liew
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Fan Zhang
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Immunology, Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Iris Madera-Salcedo
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Frank Rosenbauer
- Institute of Molecular Tumor Biology, University of Muenster, Muenster, Germany
| | - Richard M Stone
- Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Jon C Aster
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ulrich H von Andrian
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Immunology, Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
| | - Tanya N Mayadas
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Li S, Yao JC, Li JT, Schmidt AP, Link DC. TLR7/8 agonist treatment induces an increase in bone marrow resident dendritic cells and hematopoietic progenitor expansion and mobilization. Exp Hematol 2021; 96:35-43.e7. [PMID: 33556431 PMCID: PMC9900459 DOI: 10.1016/j.exphem.2021.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 02/08/2023]
Abstract
There is accumulating evidence suggesting that toll-like receptor (TLR) signals play an important role in the regulation of hematopoietic stem/progenitor cells (HSPCs). TLR7/8 stimulation induces the myeloid differentiation of normal HSPCs and acute myeloid leukemia cells. However, the in vivo effect of TLR7/8 agonists on hematopoiesis is largely unknown. Here, we show that, similar to TLR4 and TLR2, treatment with the TLR7/8 agonist R848 induces an expansion of phenotypic hematopoietic stem cells (HSCs) with reduced repopulating potential and HSPC mobilization. In contrast to chronic TLR4 stimulation, treatment with R848 for 5 days did not induce a significant increase in myeloid-biased HSCs. Treatment with R848 results in a significant increase in classic dendritic cells (DCs) in the bone marrow, but a decrease in common dendritic cell progenitors and pre-DCs. Phenotypic analysis of DCs revealed that R848 treatment is associated with altered expression of certain chemokines, activation markers, and migratory receptors. Together, these data indicate that systemic administration of a TLR7/8 agonist has unique effects on hematopoiesis, including the expansion of DCs in the bone marrow, that might have clinical relevance to augment responses to certain immunotherapies, such as cancer vaccines and immune checkpoint blockade.
Collapse
Affiliation(s)
- Sidan Li
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.,Hematology Oncology Center, Beijing Children’s Hospital, National Center for Children’s Health, Capital Medial University, Beijing, China
| | - Juo-Chin Yao
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Justin T. Li
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Amy P. Schmidt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Daniel C. Link
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
18
|
An X, Bai Q, Bing Z, Liu H, Zhang Q, Liu H, Yao X. Revealing the Positive Binding Cooperativity Mechanism between the Orthosteric and the Allosteric Antagonists of CCR2 by Metadynamics and Gaussian Accelerated Molecular Dynamics Simulations. ACS Chem Neurosci 2020; 11:628-637. [PMID: 31968162 DOI: 10.1021/acschemneuro.9b00630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
CC chemokine receptor 2 (CCR2) and its endogenous CC chemokine ligands are associated with numerous inflammatory, neurodegenerative diseases, and cancer. CCR2 is becoming an attractive target in the treatment of autoimmune disease and neurodegenerative diseases. The orthosteric antagonist BMS-681 and allosteric antagonist CCR2-RA-[R] of CCR2 show positive binding cooperativity. We performed well-tempered metadynamics simulations and Gaussian accelerated MD simulations to reveal the influence of the orthosteric antagonist on the unbinding of allosteric antagonist of CCR2. We revealed different unbinding pathways of CCR2-RA-[R] in binary complex CCR2-VT5 and ternary complex CCR2-73R-VT5. The different unbinding pathways of CCR2-RA-[R] are due to the conformational dynamics of TM6. We obtained the significant conformational differences of the intracellular side of TM6 upon CCR2 binding to different ligands by GaMD simulation. The conformational dynamics of TM6 are consistent with the unbinding pathway analysis. GaMD simulations indicate that BMS-681 binding restricts the bend of intracellular side of TM6 by stabilizing the extracellular sides of TM6 and TM7. The charged residues Arg2065.43 of TM5 and Glu2917.39 of TM7 play key roles in stabling TM7 and TM6. TM6 and TM7 are crucial components in the orthosteric and allosteric binding sites. Our results illustrate the conformational details about the effect of the orthosteric antagonist on the allosteric antagonist of CCR2. The conformational dynamics of CCR2 upon binding to different ligands can provide a rational basis for development of allosteric ligands of CCR2.
Collapse
Affiliation(s)
- Xiaoli An
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Qifeng Bai
- School of Basic Medical Science, Lanzhou University, Lanzhou 730000, China
| | - Zhitong Bing
- School of Basic Medical Science, Lanzhou University, Lanzhou 730000, China
- Institute of Modern Physics of Chinese Academy of Sciences, Lanzhou, Gansu 730000, China
| | - Hongli Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P. R. China
| | - Qianqian Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P. R. China
| | - Huanxiang Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P. R. China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou 730000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| |
Collapse
|
19
|
Joshi U, Pearson A, Evans JE, Langlois H, Saltiel N, Ojo J, Klimas N, Sullivan K, Keegan AP, Oberlin S, Darcey T, Cseresznye A, Raya B, Paris D, Hammock B, Vasylieva N, Hongsibsong S, Stern LJ, Crawford F, Mullan M, Abdullah L. A permethrin metabolite is associated with adaptive immune responses in Gulf War Illness. Brain Behav Immun 2019; 81:545-559. [PMID: 31325531 PMCID: PMC7155744 DOI: 10.1016/j.bbi.2019.07.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/17/2019] [Accepted: 07/11/2019] [Indexed: 10/31/2022] Open
Abstract
Gulf War Illness (GWI), affecting 30% of veterans from the 1991 Gulf War (GW), is a multi-symptom illness with features similar to those of patients with autoimmune diseases. The objective of the current work is to determine if exposure to GW-related pesticides, such as permethrin (PER), activates peripheral and central nervous system (CNS) adaptive immune responses. In the current study, we focused on a PER metabolite, 3-phenoxybenzoic acid (3-PBA), as this is a common metabolite previously shown to form adducts with endogenous proteins. We observed the presence of 3-PBA and 3-PBA modified lysine of protein peptides in the brain, blood and liver of pyridostigmine bromide (PB) and PER (PB+PER) exposed mice at acute and chronic post-exposure timepoints. We tested whether 3-PBA-haptenated albumin (3-PBA-albumin) can activate immune cells since it is known that chemically haptenated proteins can stimulate immune responses. We detected autoantibodies against 3-PBA-albumin in plasma from PB + PER exposed mice and veterans with GWI at chronic post-exposure timepoints. We also observed that in vitro treatment of blood with 3-PBA-albumin resulted in the activation of B- and T-helper lymphocytes and that these immune cells were also increased in blood of PB + PER exposed mice and veterans with GWI. These immune changes corresponded with elevated levels of infiltrating monocytes in the brain and blood of PB + PER exposed mice which coincided with alterations in the markers of blood-brain barrier disruption, brain macrophages and neuroinflammation. These studies suggest that pesticide exposure associated with GWI may have resulted in the activation of the peripheral and CNS adaptive immune responses, possibly contributing to an autoimmune-type phenotype in veterans with GWI.
Collapse
Affiliation(s)
- Utsav Joshi
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Andrew Pearson
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - James E. Evans
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Heather Langlois
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Nicole Saltiel
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Joseph Ojo
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Nancy Klimas
- NOVA Southeastern University, Ft. Lauderdale, FL, USA,Miami VAMC, Miami, FL, USA
| | | | | | - Sarah Oberlin
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Teresa Darcey
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Adam Cseresznye
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Balaram Raya
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,James A. Haley VA Hospital, Tampa, FL, USA
| | - Daniel Paris
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Bruce Hammock
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Natalia Vasylieva
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Surat Hongsibsong
- Environment and Health Research Unit, Research Institute for Health Science, Chiang Mai University, Chiang, Thailand
| | - Lawrence J. Stern
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA,Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Fiona Crawford
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Michael Mullan
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA,Open University, Milton Keynes, UK,James A. Haley VA Hospital, Tampa, FL, USA
| | - Laila Abdullah
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL, USA; Open University, Milton Keynes, UK; James A. Haley VA Hospital, Tampa, FL, USA.
| |
Collapse
|
20
|
Gomes AC, Mohsen MO, Mueller JE, Leoratti FMS, Cabral-Miranda G, Bachmann MF. Early Transcriptional Signature in Dendritic Cells and the Induction of Protective T Cell Responses Upon Immunization With VLPs Containing TLR Ligands-A Role for CCL2. Front Immunol 2019; 10:1679. [PMID: 31428084 PMCID: PMC6687836 DOI: 10.3389/fimmu.2019.01679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 07/04/2019] [Indexed: 11/13/2022] Open
Abstract
Inducing T cell responses by therapeutic vaccination requires appropriate activation of antigen presenting cells (APCs). The use of virus-like particles (VLPs) containing Toll-like receptor (TLR) ligands has demonstrated remarkable potential in activating APCs and modulating the immune response both for prophylactic vaccines as well as immunotherapy. Here, we employed VLPs associated to TLR ligands as tools to modulate cytotoxic response mediated by CD8+ T cells and provide further insight in the development of T cell-based immunotherapy. We have investigated the in vivo transcriptional signature in dendritic cells (DCs) from mice immunized with VLPs containing distinct classes of nucleic acid and correlated the expression patterns with the efficiency of induced T cell responses. We identified key pathways activated in DCs that are involved in the appropriated induction of T cell responses and show evidence for the modulatory effect of CCL2 in CD8+ T cells responses. These insights shed light on immune networks that are pivotal for the induction of potent cytotoxic T cell responses and identify key genes for appropriate DC activation and subsequent modulation of the adaptive immune response.
Collapse
Affiliation(s)
- Ariane C. Gomes
- The Jenner Institute, Oxford University, Oxford, United Kingdom
| | - Mona O. Mohsen
- The Jenner Institute, Oxford University, Oxford, United Kingdom
| | | | | | | | - Martin F. Bachmann
- The Jenner Institute, Oxford University, Oxford, United Kingdom
- Immunology, Inselspital, Bern, Switzerland
| |
Collapse
|
21
|
Yashiro T, Takeuchi H, Nakamura S, Tanabe A, Hara M, Uchida K, Okumura K, Kasakura K, Nishiyama C. PU.1 plays a pivotal role in dendritic cell migration from the periphery to secondary lymphoid organs via regulating CCR7 expression. FASEB J 2019; 33:11481-11491. [PMID: 31314592 DOI: 10.1096/fj.201900379rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
C-C chemokine receptor type 7 (CCR7) is essential for migration of dendritic cells (DCs) to draining lymph nodes. PU.1/Spi1 is a transcription factor playing a critical role in the gene regulation of DCs. PU.1 knockdown decreased the expression of CCR7 in bone marrow-derived DCs and subsequently attenuated migration in vitro and in vivo. Reporter assays, EMSA, and chromatin immunoprecipitation assays revealed that PU.1 binds to the most proximal Ets motif of the Ccr7 promoter, which is involved in transcriptional activation. The CCR7 expression level, which was higher in the programmed cell death 1 ligand 2 (PD-L2)+ population than in the PD-L2- population and was markedly suppressed by TGF-β treatment, coincided with the binding level of PU.1 to the Ccr7 promoter. The PU.1 binding level in CCR7high mesenteric lymph nodes DCs was higher than in other DC subtypes. The involvement of PU.1 in the expression of the CCR7 gene was also observed in human DCs. We conclude that PU.1 plays a pivotal role in DC migration by transactivating the CCR7 gene via the Ets motif in the promoter in both humans and mice.-Yashiro, T., Takeuchi, H., Nakamura, S., Tanabe, A., Hara, M., Uchida, K., Okumura, K., Kasakura, K., Nishiyama, C. PU.1 plays a pivotal role in dendritic cell migration from the periphery to secondary lymphoid organs via regulating CCR7 expression.
Collapse
Affiliation(s)
- Takuya Yashiro
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan.,Atopy Research Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiromi Takeuchi
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Shusuke Nakamura
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Atsushi Tanabe
- Atopy Research Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Mutsuko Hara
- Atopy Research Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Koichiro Uchida
- Atopy Research Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Atopy Research Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazumi Kasakura
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan.,Atopy Research Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Chiharu Nishiyama
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan.,Atopy Research Center, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Xu L, Sharkey D, Cantley LG. Tubular GM-CSF Promotes Late MCP-1/CCR2-Mediated Fibrosis and Inflammation after Ischemia/Reperfusion Injury. J Am Soc Nephrol 2019; 30:1825-1840. [PMID: 31315923 DOI: 10.1681/asn.2019010068] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/22/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND After bilateral kidney ischemia/reperfusion injury (IRI), monocytes infiltrate the kidney and differentiate into proinflammatory macrophages in response to the initial kidney damage, and then transition to a form that promotes kidney repair. In the setting of unilateral IRI (U-IRI), however, we have previously shown that macrophages persist beyond the time of repair and may promote fibrosis. METHODS Macrophage homing/survival signals were determined at 14 days after injury in mice subjected to U-IRI and in vitro using coculture of macrophages and tubular cells. Mice genetically engineered to lack Ccr2 and wild-type mice were treated ±CCR2 antagonist RS102895 and subjected to U-IRI to quantify macrophage accumulation, kidney fibrosis, and inflammation 14 and 30 days after the injury. RESULTS Failure to resolve tubular injury after U-IRI results in sustained expression of granulocyte-macrophage colony-stimulating factor by renal tubular cells, which directly stimulates expression of monocyte chemoattractant protein-1 (Mcp-1) by macrophages. Analysis of CD45+ immune cells isolated from wild-type kidneys 14 days after U-IRI reveals high-level expression of the MCP-1 receptor Ccr2. In mice lacking Ccr2 and wild-type mice treated with RS102895, the numbers of macrophages, dendritic cells, and T cell decreased following U-IRI, as did the expression of profibrotic growth factors and proimflammatory cytokines. This results in a reduction in extracellular matrix and kidney injury markers. CONCLUSIONS GM-CSF-induced MCP-1/CCR2 signaling plays an important role in the cross-talk between injured tubular cells and infiltrating immune cells and myofibroblasts, and promotes sustained inflammation and tubular injury with progressive interstitial fibrosis in the late stages of U-IRI.
Collapse
Affiliation(s)
- Leyuan Xu
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Diana Sharkey
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut
| | - Lloyd G Cantley
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| |
Collapse
|
23
|
Immunosuppressive Tumor Microenvironment Status and Histological Grading of Endometrial Carcinoma. CANCER MICROENVIRONMENT 2019; 12:169-179. [PMID: 31134527 DOI: 10.1007/s12307-019-00225-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/02/2019] [Indexed: 01/05/2023]
Abstract
The recent successes of new cancer immunotherapy approaches have led to investigate their relevance in the context of the Endometrial Carcinoma (EC). These therapies, that take the tumor-induced immunosuppressive microenvironment into account, target the tumor immune escape, in particular the inhibitory receptors involved in the regulation of the effector T cells' activity (immune checkpoints). The aim of this study was to identify, in ECs, differences in intergrades immune status that could contribute to the differences in tumor aggressiveness, and could also be used as theranostic tools. The immune status of tumors was assessed by quantitative real-time PCR. We analyzed the expression of specific genes associated to specific leukocytes subpopulations and the expression of reporting genes associated with the tumor escape/resistance. This study highlights significant differences in the EC intergrades immune status especially the tumor-infiltrating cell types and their activation status as well as in the molecular factors produced by the environment. The immune microenvironment of grade 1 ECs hints at a robust tumoricidal milieu while that of higher grades is more evocative of a tolerogenic milieu. This genes-based immunological monitoring of tumors that easily highlights significant intergrade differences relating to the density, composition and functional state of the leukocyte infiltrate, could give solid arguments for choosing the best therapeutic options, especially those targeting immune checkpoints. Moreover it could enable an easy adaptation of individual treatment approaches for each patient.
Collapse
|
24
|
Heymann F, von Trotha KT, Preisinger C, Lynen-Jansen P, Roeth AA, Geiger M, Geisler LJ, Frank AK, Conze J, Luedde T, Trautwein C, Binnebösel M, Neumann UP, Tacke F. Polypropylene mesh implantation for hernia repair causes myeloid cell-driven persistent inflammation. JCI Insight 2019; 4:123862. [PMID: 30674727 DOI: 10.1172/jci.insight.123862] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022] Open
Abstract
Polypropylene meshes that are commonly used for inguinal hernia repair may trigger granulomatous foreign body reactions. Here, we show that asymptomatic patients display mesh-associated inflammatory granulomas long after surgery, which are dominated by monocyte-derived macrophages expressing high levels of inflammatory activation markers. In mice, mesh implantation by the onlay technique induced rapid and strong myeloid cell accumulation, without substantial attenuation for up to 90 days. Myeloid cells segregated into distinct macrophage subsets with separate spatial distribution, activation profiles, and functional properties, showing a stable inflammatory phenotype in the tissue surrounding the biomaterial and a mixed, wound-healing phenotype in the surrounding stromal tissue. Protein mass spectrometry confirmed the inflammatory nature of the foreign body reaction, as characterized by cytokines, complement activation, and matrix-modulating factors. Moreover, immunoglobulin deposition increased over time around the implant, arguing for humoral immune responses in association with the cell-driven inflammation. Intravital multiphoton microscopy revealed a high motility and continuous recruitment of myeloid cells, which is partly dependent on the chemokine receptor CCR2. CCR2-dependent macrophages are particular drivers of fibroblast proliferation. Thus, our work functionally characterizes myeloid cell-dependent inflammation following mesh implantation, thereby providing insights into the dynamics and mechanisms of foreign body reactions to implanted biomaterials.
Collapse
Affiliation(s)
| | - Klaus-Thilo von Trotha
- Department of General, Visceral and Transplantation Surgery.,Department of Vascular Surgery, and
| | - Christian Preisinger
- Proteomics Core Facility Interdisciplinary Center for Clinical Research, University Hospital Aachen, Aachen, Germany
| | - Petra Lynen-Jansen
- Department of General, Visceral and Transplantation Surgery.,German Association for Gastroenterology, Berlin, Germany
| | - Anjali A Roeth
- Department of General, Visceral and Transplantation Surgery
| | | | | | | | - Joachim Conze
- Department of General, Visceral and Transplantation Surgery.,Hernienzentrum Dr. Conze, Munich, Germany
| | | | | | - Marcel Binnebösel
- Department of General, Visceral and Transplantation Surgery.,Department of Surgery, University Medical Center Maastricht, Maastricht, Netherlands
| | - Ulf P Neumann
- Department of General, Visceral and Transplantation Surgery.,Hernienzentrum Dr. Conze, Munich, Germany
| | | |
Collapse
|
25
|
Oleoylethanolamide treatment reduces neurobehavioral deficits and brain pathology in a mouse model of Gulf War Illness. Sci Rep 2018; 8:12921. [PMID: 30150699 PMCID: PMC6110778 DOI: 10.1038/s41598-018-31242-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 08/15/2018] [Indexed: 12/12/2022] Open
Abstract
There are nearly 250,000 Gulf War (GW) veterans who suffer from Gulf War Illness (GWI), a multi-symptom condition that remains untreatable. The main objective was to determine if targeting peroxisomal function could be of therapeutic value in GWI. We performed a pilot study that showed accumulation of very long chain fatty acids (VLCFA), which are metabolized in peroxisomes, in plasma from veterans with GWI. We then examined if targeting peroxisomal β-oxidation with oleoylethanolamide (OEA) restores these lipids to the normal levels and mitigates neuroinflammation and neurobehavioral deficits in a well-established mouse model of GWI. In GWI mice, treatment with OEA corresponded with cognitive benefits and reduced fatigue and disinhibition-like behavior in GWI mice. Biochemical and molecular analysis of the brain tissue showed reduced astroglia and microglia staining, decreased levels of chemokines and cytokines, and decreased NFκB phosphorylation. Treatment with OEA reduced accumulation of peroxisome specific VLCFA in the brains of GWI mice. These studies further support the translational value of targeting peroxisomes. We expect that OEA may be a potential therapy for treating neurobehavioral symptoms and the underlying lipid dysfunction and neuroinflammation associated with GWI. Oleoylethanolamide is available as a dietary supplement, making it appealing for human translational studies.
Collapse
|
26
|
Song N, Zhang T, Xu X, Lu Z, Yu X, Fang Y, Hu J, Jia P, Teng J, Ding X. miR-21 Protects Against Ischemia/Reperfusion-Induced Acute Kidney Injury by Preventing Epithelial Cell Apoptosis and Inhibiting Dendritic Cell Maturation. Front Physiol 2018; 9:790. [PMID: 30013485 PMCID: PMC6036242 DOI: 10.3389/fphys.2018.00790] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/06/2018] [Indexed: 02/02/2023] Open
Abstract
Renal tubular injury and innate immune responses induced by hypoxia contribute to acute kidney injury. Accumulating evidence suggests that miR-21 overexpression protects against kidney ischemia injury. Additionally, miR-21 emerges as a key inhibitor in dendritic cell maturation. Thus, we hypothesized that miR-21 protects the kidney from IR injury by suppressing epithelial cell damage and inflammatory reaction. In this study, we investigated effects of miR-21 and its signaling pathways (PTEN/AKT/mTOR/HIF, PDCD4/NFκ-B) on kidney ischemia/reperfusion (IR) injury in vitro and in vivo. The results revealed that IR increased miR-21, HIF1α, and 2α expression in vivo and in vitro. MiR-21 interacted with HIF1α and 2α through the PTEN/AKT/mTOR pathway. Moreover, inhibition of miR-21 activated PDCD4/NFκ-B pathways, which are critical for dendritic cell maturation. Renal IR triggers local inflammation by inducing the dendritic cell maturation and promoting the secretion of IL-12, IL-6, and TNF-α cytokines. Knockdown of miR-21 intensified the effect of IR on tubular epithelial cell apoptosis and dendritic cell maturation. Our results suggested that IR-inducible miR-21 protects epithelial cells from IR injury via a feedback interaction with HIF (PTEN/AKT/mTOR/HIF/miR-21) and by inhibiting maturation of DCs through the PDCD4/NF-κB pathway. These findings highlight new therapeutic opportunities in AKI.
Collapse
Affiliation(s)
- Nana Song
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Ting Zhang
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - XiaLian Xu
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Zhihui Lu
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xiaofang Yu
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Yi Fang
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Jiachang Hu
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Ping Jia
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Jie Teng
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| | - Xiaoqiang Ding
- Division of Nephrology, Zhongshan Hospital,Fudan University, Shanghai, China.,Shanghai Medical Center of Kidney, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China.,Hemodialysis Quality Control Center of Shanghai, Shanghai, China
| |
Collapse
|
27
|
Abolhalaj M, Askmyr D, Sakellariou CA, Lundberg K, Greiff L, Lindstedt M. Profiling dendritic cell subsets in head and neck squamous cell tonsillar cancer and benign tonsils. Sci Rep 2018; 8:8030. [PMID: 29795118 PMCID: PMC5966442 DOI: 10.1038/s41598-018-26193-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 04/19/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) have a key role in orchestrating immune responses and are considered important targets for immunotherapy against cancer. In order to develop effective cancer vaccines, detailed knowledge of the micromilieu in cancer lesions is warranted. In this study, flow cytometry and human transcriptome arrays were used to characterize subsets of DCs in head and neck squamous cell tonsillar cancer and compare them to their counterparts in benign tonsils to evaluate subset-selective biomarkers associated with tonsillar cancer. We describe, for the first time, four subsets of DCs in tonsillar cancer: CD123+ plasmacytoid DCs (pDC), CD1c+, CD141+, and CD1c-CD141- myeloid DCs (mDC). An increased frequency of DCs and an elevated mDC/pDC ratio were shown in malignant compared to benign tonsillar tissue. The microarray data demonstrates characteristics specific for tonsil cancer DC subsets, including expression of immunosuppressive molecules and lower expression levels of genes involved in development of effector immune responses in DCs in malignant tonsillar tissue, compared to their counterparts in benign tonsillar tissue. Finally, we present target candidates selectively expressed by different DC subsets in malignant tonsils and confirm expression of CD206/MRC1 and CD207/Langerin on CD1c+ DCs at protein level. This study descibes DC characteristics in the context of head and neck cancer and add valuable steps towards future DC-based therapies against tonsillar cancer.
Collapse
Affiliation(s)
- Milad Abolhalaj
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - David Askmyr
- Department of ORL, Head & Neck Surgery, Skåne University Hospital, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | | | | | - Lennart Greiff
- Department of ORL, Head & Neck Surgery, Skåne University Hospital, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Malin Lindstedt
- Department of Immunotechnology, Lund University, Lund, Sweden.
| |
Collapse
|
28
|
Novel Models to Study Stromal Cell-Leukocyte Interactions in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:131-146. [PMID: 30155626 DOI: 10.1007/978-3-319-78127-3_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
To study human immunology in general and stromal immunology in particular, it is highly motivated to move from monolayers to 3D cultures, such as organotypic models, that better mimic the function of living tissue. These models can potentially contain most if not all cell types present in tissues, in combination with different extracellular matrix components that can critically affect cell phenotype. Besides their well-established use in studies of tissue-specific cells, such as epithelial cells, endothelial cells and stromal fibroblasts in combination with extracellular components, these models have also been shown to be valuable to study how tissue participates in the regulation of leukocyte differentiation and function. Organotypic models with leukocytes represent novel powerful tools to study human stromal immunology and mechanisms involved in the regulation of leukocyte functions and inflammatory processes in human health and disease. In particular, these models are robust, long-lived and reproducible and allow monitoring of disease progression in real time, as well as the mixing of cellular constituents from healthy and pathological tissues. These models are also easy to manipulate, either genetically or by adding external stimulants, such as cytokines and pathogens, to mimic pathological conditions. It is thus not surprising that these models are proposed to be useful in toxicology screening assays, evaluating therapeutic efficacy of drugs and antibiotics, as well as in personalized medicine. Within this chapter, the most recent developments in creating organotypic models for the purpose of study of human leukocyte and stromal cell interactions, in health and disease, will be discussed, in particular focusing on live imaging. Special emphasis will be given on an organotypic model resembling human lung and its usefulness in studying the fine control of physiological and pathological processes in human health and disease. Using these models in studies on human stromal cell and leukocyte interactions will likely help identifying novel disease traits and may point out new potential targets to monitor and treat human diseases.
Collapse
|
29
|
Smirnov A, Pohlmann S, Nehring M, Ali S, Mann-Nüttel R, Scheu S, Antoni AC, Hansen W, Büettner M, Gardiasch MJ, Westendorf AM, Wirsdörfer F, Pastille E, Dudda M, Flohé SB. Sphingosine 1-Phosphate- and C-C Chemokine Receptor 2-Dependent Activation of CD4 + Plasmacytoid Dendritic Cells in the Bone Marrow Contributes to Signs of Sepsis-Induced Immunosuppression. Front Immunol 2017; 8:1622. [PMID: 29218051 PMCID: PMC5703700 DOI: 10.3389/fimmu.2017.01622] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 11/08/2017] [Indexed: 12/24/2022] Open
Abstract
Sepsis is the dysregulated response of the host to systemic, mostly bacterial infection, and is associated with an enhanced susceptibility to life-threatening opportunistic infections. During polymicrobial sepsis, dendritic cells (DCs) secrete enhanced levels of interleukin (IL) 10 due to an altered differentiation in the bone marrow and contribute to the development of immunosuppression. We investigated the origin of the altered DC differentiation using murine cecal ligation and puncture (CLP), a model for human polymicrobial sepsis. Bone marrow cells (BMC) were isolated after sham or CLP operation, the cellular composition was analyzed, and bone marrow-derived DCs (BMDCs) were generated in vitro. From 24 h on after CLP, BMC gave rise to BMDC that released enhanced levels of IL-10. In parallel, a population of CD11chiMHCII+CD4+ DCs expanded in the bone marrow in a MyD88-dependent manner. Prior depletion of the CD11chiMHCII+CD4+ DCs from BMC in vitro reversed the increased IL-10 secretion of subsequently differentiating BMDC. The expansion of the CD11chiMHCII+CD4+ DC population in the bone marrow after CLP required the function of sphingosine 1-phosphate receptors and C-C chemokine receptor (CCR) 2, the receptor for C-C chemokine ligand (CCL) 2, but was not associated with monocyte mobilization. CD11chiMHCII+CD4+ DCs were identified as plasmacytoid DCs (pDCs) that had acquired an activated phenotype according to their increased expression of MHC class II and CD86. A redistribution of CD4+ pDCs from MHC class II− to MHC class II+ cells concomitant with enhanced expression of CD11c finally led to the rise in the number of CD11chiMHCII+CD4+ DCs. Enhanced levels of CCL2 were found in the bone marrow of septic mice and the inhibition of CCR2 dampened the expression of CD86 on CD4+ pDCs after CLP in vitro. Depletion of pDCs reversed the bias of splenic DCs toward increased IL-10 synthesis after CLP in vivo. Thus, during polymicrobial sepsis, CD4+ pDCs are activated in the bone marrow and induce functional reprogramming of differentiating BMDC toward an immunosuppressive phenotype.
Collapse
Affiliation(s)
- Anna Smirnov
- Department of Orthopedics and Trauma Surgery, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stephanie Pohlmann
- Department of Orthopedics and Trauma Surgery, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Melanie Nehring
- Department of Orthopedics and Trauma Surgery, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Shafaqat Ali
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany.,Cells in Motion, Cluster of Excellence, University of Münster, Münster, Germany
| | - Ritu Mann-Nüttel
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| | - Anne-Charlotte Antoni
- Department of Orthopedics and Trauma Surgery, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Manuela Büettner
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Miriam J Gardiasch
- Department of Orthopedics and Trauma Surgery, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Florian Wirsdörfer
- Medical Faculty, Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| | - Eva Pastille
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Marcel Dudda
- Department of Orthopedics and Trauma Surgery, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stefanie B Flohé
- Department of Orthopedics and Trauma Surgery, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
30
|
Nakagaki BN, Freitas-Lopes MA, Carvalho É, Carvalho-Gontijo R, Castro-Oliveira HM, Rezende RM, Cara DC, Santos MM, Lopes RP, David BA, Menezes GB. Generation of a triple-fluorescent mouse strain allows a dynamic and spatial visualization of different liver phagocytes in vivo. AN ACAD BRAS CIENC 2017; 91:e20170317. [PMID: 29044327 DOI: 10.1590/0001-3765201720170317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/28/2017] [Indexed: 12/31/2022] Open
Abstract
Resident and circulating immune cells have been extensively studied due to their almost ubiquitous role in cell biology. Despite their classification under the "immune cell department", it is becoming increasingly clear that these cells are involved in many different non-immune related phenomena, including fetus development, vascular formation, memory, social behavior and many other phenotypes. There is a huge potential in combining high-throughput assays - including flow cytometry and gene analysis - with in vivo imaging. This can improve our knowledge in both basic and clinical cell biology, and accessing the expression of markers that are relevant in the context of both homeostasis and disease conditions might be instrumental. Here we describe how we generated a novel mouse strain that spontaneously express three different fluorescence markers under control of well-studied receptors (CX3CR1, CCR2 and CD11c) that are involved in a plethora of stages of cell ontogenesis, maturation, migration and behavior. Also, we assess the percentage of the expression and co-expression of each marker under homeostasis conditions, and how these cells behave when a local inflammation is induced in the liver applying a cutting-edge technology to image cells by confocal intravital microscopy.
Collapse
Affiliation(s)
- Brenda N Nakagaki
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Maria A Freitas-Lopes
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Érika Carvalho
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Raquel Carvalho-Gontijo
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Hortência M Castro-Oliveira
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Rafael M Rezende
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, 02115, Boston, MA, United States of America
| | - Denise C Cara
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Mônica M Santos
- Departamento de Biologia Animal, Universidade Federal de Viçosa, Av. Peter Henry Rolfs, s/n, Campus Universitário, 36570-900 Viçosa, MG, Brazil
| | - Rodrigo Pestana Lopes
- BD Biosciences, Rua Alexandre Dumas, 1976, Chácara Santo Antônio, 04717-040 São Paulo, SP, Brazil
| | - Bruna A David
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil.,Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo/USP, Av. Bandeirantes, 3900, Monte Alegre, 14049-900 Ribeirão Preto, SP, Brazil
| | - Gustavo B Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| |
Collapse
|
31
|
Feinstein J, Ramkhelawon B. Netrins & Semaphorins: Novel regulators of the immune response. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3183-3189. [PMID: 28918114 DOI: 10.1016/j.bbadis.2017.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 12/26/2022]
Abstract
Netrins and semaphorins, members of the neuronal guidance cue family, exhibit a rich biology with significant roles that extend beyond chemotactic guidance of the axons to build the neuronal patterns of the body. Screening of adult tissues and specific cellular subsets have illuminated that these proteins are also abundantly expressed under both steady state and pathological scenarios. This observation suggests that, in addition to their role in the development of the axonal tree, these proteins possess additional novel functions in adult physiopathology. Notably, a series of striking evidence has emerged in the literature describing their roles as potent regulators of both innate and adaptive immunity, providing extra dimension to our knowledge of neuronal guidance cues. In this review, we summarize the key complex roles of netrins and semaphorins outside the central nervous system (CNS) with focus on their immunomodulatory functions that impact pathophysiological conditions.
Collapse
Affiliation(s)
- Jordyn Feinstein
- Division of Vascular Surgery, Department of Surgery, New York University School of Medicine, 530 First Avenue, New York, NY 10016, USA; Department of Cell Biology, New York University School of Medicine, 530 First Avenue, New York, NY 10016, USA
| | - Bhama Ramkhelawon
- Division of Vascular Surgery, Department of Surgery, New York University School of Medicine, 530 First Avenue, New York, NY 10016, USA; Department of Cell Biology, New York University School of Medicine, 530 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
32
|
Nguyen MA, Karunakaran D, Geoffrion M, Cheng HS, Tandoc K, Perisic Matic L, Hedin U, Maegdefessel L, Fish JE, Rayner KJ. Extracellular Vesicles Secreted by Atherogenic Macrophages Transfer MicroRNA to Inhibit Cell Migration. Arterioscler Thromb Vasc Biol 2017; 38:49-63. [PMID: 28882869 DOI: 10.1161/atvbaha.117.309795] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 08/18/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE During inflammation, macrophages secrete vesicles carrying RNA, protein, and lipids as a form of extracellular communication. In the vessel wall, extracellular vesicles (EVs) have been shown to be transferred between vascular cells during atherosclerosis; however, the role of macrophage-derived EVs in atherogenesis is not known. Here, we hypothesize that atherogenic macrophages secrete microRNAs (miRNAs) in EVs to mediate cell-cell communication and promote proinflammatory and proatherogenic phenotypes in recipient cells. APPROACH AND RESULTS We isolated EVs from mouse and human macrophages treated with an atherogenic stimulus (oxidized low-density lipoprotein) and characterized the EV miRNA expression profile. We confirmed the enrichment of miR-146a, miR-128, miR-185, miR-365, and miR-503 in atherogenic EVs compared with controls and demonstrate that these EVs are taken up and transfer exogenous miRNA to naive recipient macrophages. Bioinformatic pathway analysis suggests that atherogenic EV miRNAs are predicted to target genes involved in cell migration and adhesion pathways, and indeed delivery of EVs to naive macrophages reduced macrophage migration both in vitro and in vivo. Inhibition of miR-146a, the most enriched miRNA in atherogenic EVs, reduced the inhibitory effect of EVs on macrophage migratory capacity. EV-mediated delivery of miR-146a repressed the expression of target genes IGF2BP1 (insulin-like growth factor 2 mRNA-binding protein 1) and HuR (human antigen R or ELAV-like RNA-binding protein 1) in recipient cells, and knockdown of IGF2BP1 and HuR using short interfering RNA greatly reduced macrophage migration, highlighting the importance of these EV-miRNA targets in regulating macrophage motility. CONCLUSIONS EV-derived miRNAs from atherogenic macrophages, in particular miR-146a, may accelerate the development of atherosclerosis by decreasing cell migration and promoting macrophage entrapment in the vessel wall.
Collapse
Affiliation(s)
- My-Anh Nguyen
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Denuja Karunakaran
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Michèle Geoffrion
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Henry S Cheng
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Kristofferson Tandoc
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Ljubica Perisic Matic
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Ulf Hedin
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Lars Maegdefessel
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Jason E Fish
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.)
| | - Katey J Rayner
- From the University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., D.K., M.G., K.T., K.J.R.); Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.); Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C., J.E.F.); Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden (L.P.M., U.H.); and Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.).
| |
Collapse
|
33
|
Gurses KM, Ozmen F, Kocyigit D, Yersal N, Bilgic E, Kaya E, Kopru CZ, Soyal T, Doganci S, Tokgozoglu L, Korkusuz P. Netrin-1 is associated with macrophage infiltration and polarization in human epicardial adipose tissue in coronary artery disease. J Cardiol 2017; 69:851-858. [DOI: 10.1016/j.jjcc.2016.08.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022]
|
34
|
Pahuja NK, Shetty R, Deshmukh R, Sharma A, Nuijts RMMA, Jhanji V, Sethu S, Ghosh A. In vivo confocal microscopy and tear cytokine analysis in post-LASIK ectasia. Br J Ophthalmol 2017; 101:1604-1610. [PMID: 28450380 DOI: 10.1136/bjophthalmol-2016-309142] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 01/23/2017] [Accepted: 03/21/2017] [Indexed: 11/03/2022]
Abstract
AIM Corneal keratectasia is one of the complications associated with laser in situ keratomileusis (LASIK) that results in vision impairment. The pathogenesis of post-LASIK ectasia (PLE) remains underexplored. We report the tear cytokine profile and in vivo confocal microscopy (IVCM) findings in eyes with PLE. METHODS This retrospective study included age-matched 7 (14 eyes) post-LASIK controls (PLCs) and 6 (12 eyes) PLE subjects. Corneal topography was used to categorise the subjects into PLC and PLE groups. Ocular Surface Disease Index (OSDI) scores obtained were based on standard questionnaire and IVCM images were used to determine corneal dendritic cells density (DCD) and sub-basal nerve plexus morphology. Inflammatory cytokines/chemokines in the tears were quantified using flow cytometry based cytometric bead array. RESULTS Pentacam-based scores, OSDI scores and corneal DCD were significantly (p<0.05) higher in patients with PLE compared with PLC. Discomfort-related subscale of OSDI score exhibited a positive correlation with total corneal DCD in the PLE cohort. The fold difference of chemokine (C-C motif) ligand/monocyte chemotactic protein-1 (CCL2/MCP1) (3.4±0.6) was found to be significantly (p<0.05) higher in the PLE cohorts and a positive correlation between CCL2/MCP1 levels and total corneal DCD was also observed in the PLE cohort. CONCLUSION The current study found a significant difference in the tear film cytokine profile between normal and PLE eyes. Presence of increased corneal dendritic cells and altered tear cytokines suggests an ongoing inflammatory response in PLE.
Collapse
Affiliation(s)
| | - Rohit Shetty
- Cornea and Refractive Services, Narayana Nethralaya, Bangalore, India
| | - Rashmi Deshmukh
- Cornea and Refractive Services, Narayana Nethralaya, Bangalore, India
| | - Anupam Sharma
- Narayana Nethralaya Foundation, GROW Research Laboratory, Bangalore, India
| | - Rudy M M A Nuijts
- Cornea Clinic, Department of Ophthalmology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Vishal Jhanji
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China.,UPMC Eye Center, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Swaminathan Sethu
- Narayana Nethralaya Foundation, GROW Research Laboratory, Bangalore, India
| | - Arkasubhra Ghosh
- Narayana Nethralaya Foundation, GROW Research Laboratory, Bangalore, India.,Singapore Eye Research Institute, Singapore
| |
Collapse
|
35
|
Baharom F, Thomas S, Rankin G, Lepzien R, Pourazar J, Behndig AF, Ahlm C, Blomberg A, Smed-Sörensen A. Dendritic Cells and Monocytes with Distinct Inflammatory Responses Reside in Lung Mucosa of Healthy Humans. THE JOURNAL OF IMMUNOLOGY 2016; 196:4498-509. [PMID: 27183618 DOI: 10.4049/jimmunol.1600071] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/04/2016] [Indexed: 12/24/2022]
Abstract
Every breath we take contains potentially harmful pathogens or allergens. Dendritic cells (DCs), monocytes, and macrophages are essential in maintaining a delicate balance of initiating immunity without causing collateral damage to the lungs because of an exaggerated inflammatory response. To document the diversity of lung mononuclear phagocytes at steady-state, we performed bronchoscopies on 20 healthy subjects, sampling the proximal and distal airways (bronchial wash and bronchoalveolar lavage, respectively), as well as mucosal tissue (endobronchial biopsies). In addition to a substantial population of alveolar macrophages, we identified subpopulations of monocytes, myeloid DCs (MDCs), and plasmacytoid DCs in the lung mucosa. Intermediate monocytes and MDCs were highly frequent in the airways compared with peripheral blood. Strikingly, the density of mononuclear phagocytes increased upon descending the airways. Monocytes from blood and airways produced 10-fold more proinflammatory cytokines than MDCs upon ex vivo stimulation. However, airway monocytes were less inflammatory than blood monocytes, suggesting a more tolerant nature. The findings of this study establish how to identify human lung mononuclear phagocytes and how they function in normal conditions, so that dysregulations in patients with respiratory diseases can be detected to elucidate their contribution to immunity or pathogenesis.
Collapse
Affiliation(s)
- Faezzah Baharom
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Saskia Thomas
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Gregory Rankin
- Division of Medicine, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden; and
| | - Rico Lepzien
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Jamshid Pourazar
- Division of Medicine, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden; and
| | - Annelie F Behndig
- Division of Medicine, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden; and
| | - Clas Ahlm
- Department of Clinical Microbiology, Infectious Diseases, Umeå University, 901 85 Umeå, Sweden
| | - Anders Blomberg
- Division of Medicine, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden; and
| | - Anna Smed-Sörensen
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden;
| |
Collapse
|
36
|
Costa DL, Lima-Júnior DS, Nascimento MS, Sacramento LA, Almeida RP, Carregaro V, Silva JS. CCR2 signaling contributes to the differentiation of protective inflammatory dendritic cells in Leishmania braziliensis infection. J Leukoc Biol 2016; 100:423-32. [PMID: 26884611 DOI: 10.1189/jlb.4a0715-288r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 01/26/2016] [Indexed: 12/11/2022] Open
Abstract
In vertebrate hosts, Leishmania braziliensis parasites infect mainly mononuclear phagocytic system cells, which when activated by T helper cell type 1 cytokines produce nitric oxide and kill the pathogens. Chemokine (C-C motif) receptor 2 is a chemokine receptor that binds primarily chemokine (C-C motif) ligand 2 and has an important role in the recruitment of monocytic phagocytes. Although it has been reported that Leishmania braziliensis infection induces CCR2 expression in the lesions, the role of CCR2 during Leishmania braziliensis infection remains unknown. Here, we showed that CCR2 has a role in mediating protection against Leishmania braziliensis infection in mice. The absence of CCR2 resulted in increased susceptibility to infection and was associated with low amounts of Ly6C(+) inflammatory dendritic cells in the lesions, which we found to be the major sources of tumor necrosis factor production and induced nitric oxide synthase expression in C57BL/6 mice lesions. Consequently, CCR2(-/-) mice showed decreased tumor necrosis factor production and induced nitric oxide synthase expression, resulting in impaired parasite elimination. We also demonstrated that CCR2 has a role in directly mediating the differentiation of monocytes into inflammatory dendritic cells at the infection sites, contributing to the accumulation of inflammatory dendritic cells in Leishmania braziliensis lesions and subsequent control of parasite replication. Therefore, these data provide new information on the role of chemokines during the immune response to infections and identify a potential target for therapeutic interventions in cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Diego L Costa
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| | - Djalma S Lima-Júnior
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| | - Manuela S Nascimento
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| | - Laís A Sacramento
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| | - Roque P Almeida
- Department of Internal Medicine and Pathology, Center for Biological and Health Sciences, Federal University of Sergipe, Aracajú, SE, Brazil
| | - Vanessa Carregaro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| | - João S Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| |
Collapse
|
37
|
Zhao L, Shao Q, Zhang Y, Zhang L, He Y, Wang L, Kong B, Qu X. Human monocytes undergo functional re-programming during differentiation to dendritic cell mediated by human extravillous trophoblasts. Sci Rep 2016; 6:20409. [PMID: 26857012 PMCID: PMC4746586 DOI: 10.1038/srep20409] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/06/2016] [Indexed: 01/02/2023] Open
Abstract
Maternal immune adaptation is required for a successful pregnancy to avoid rejection of the fetal–placental unit. Dendritic cells within the decidual microenvironment lock in a tolerogenic profile. However, how these tolerogenic DCs are induced and the underlying mechanisms are largely unknown. In this study, we show that human extravillous trophoblasts redirect the monocyte-to-DC transition and induce regulatory dendritic cells. DCs differentiated from blood monocytes in the presence of human extravillous trophoblast cell line HTR-8/SVneo displayed a DC-SIGN+CD14+CD1a− phenotype, similar with decidual DCs. HTR8-conditioned DCs were unable to develop a fully mature phenotype in response to LPS, and altered the cytokine secretory profile significantly. Functionally, conditioned DCs poorly induced the proliferation and activation of allogeneic T cells, whereas promoted CD4+CD25+Foxp3+ Treg cells generation. Furthermore, the supernatant from DC and HTR-8/SVneo coculture system contained significant high amount of M-CSF and MCP-1. Using neutralizing antibodies, we discussed the role of M-CSF and MCP-1 during monocyte-to-DCs differentiation mediated by extravillous trophoblasts. Our data indicate that human extravillous trophoblasts play an important role in modulating the monocyte-to-DC differentiation through M-CSF and MCP-1, which facilitate the establishment of a tolerogenic microenvironment at the maternal–fetal interface.
Collapse
Affiliation(s)
- Lei Zhao
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Qianqian Shao
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Yun Zhang
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Lin Zhang
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Ying He
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Lijie Wang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Xun Qu
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| |
Collapse
|
38
|
Didovic S, Opitz FV, Holzmann B, Förster I, Weighardt H. Requirement of MyD88 signaling in keratinocytes for Langerhans cell migration and initiation of atopic dermatitis-like symptoms in mice. Eur J Immunol 2016; 46:981-92. [DOI: 10.1002/eji.201545710] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 11/20/2015] [Accepted: 12/17/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Sonja Didovic
- Immunology and Environment; Life and Medical Sciences (LIMES) Institute; University of Bonn; Bonn Germany
- IUF Leibniz Research Institute for Environmental Medicine; Düsseldorf Germany
| | - Friederike V. Opitz
- Immunology and Environment; Life and Medical Sciences (LIMES) Institute; University of Bonn; Bonn Germany
- IUF Leibniz Research Institute for Environmental Medicine; Düsseldorf Germany
| | - Bernhard Holzmann
- Department of Surgery; Technische Universität München; Munich Germany
| | - Irmgard Förster
- Immunology and Environment; Life and Medical Sciences (LIMES) Institute; University of Bonn; Bonn Germany
| | - Heike Weighardt
- Immunology and Environment; Life and Medical Sciences (LIMES) Institute; University of Bonn; Bonn Germany
- IUF Leibniz Research Institute for Environmental Medicine; Düsseldorf Germany
| |
Collapse
|
39
|
Kaynar AM, Nowalk MP, Lin CJ, Moehling KK, Susick M, Bakalov V, Pitt BR, Bain DJ, Ross TM, Saul SG, Raymund M, Zimmerman RK. Are plasma mineral levels related to antibody response to influenza vaccination in older adults? Hum Vaccin Immunother 2016; 12:1003-8. [PMID: 26751915 DOI: 10.1080/21645515.2015.1113358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
INTRODUCTION An effective immune response to vaccination may be related to nutritional status. This study examined the association of plasma mineral levels with hemagglutination inhibition (HI) titers produced in response to influenza vaccine in older adults. METHODS Prior to (Day 0) and 21 (range = 19-28) days after receiving the 2013-14 influenza vaccine, 109 adults ages 51-81 years, provided blood samples. Serum samples were tested for HI activity against the A/H1N1 and A/H3N2 2013-2014 vaccine virus strains. Plasma minerals were collected in zinc-free tubes and assayed by inductively coupled plasma mass spectrometry. HI titers were reported as seroprotection (≥1:40) and seroconversion (≥ 4-fold rise from Day 0 (minimum HI = 1:10) to Day 21). Both HI titers and mineral values were skewed and thus log2 transformed. Magnesium (Mg), phosphorus (P), zinc (Zn), copper (Cu), iron (Fe), potassium (K) and the Cu to Zn ratio were tested. Logistic regression analyses were used to determine the associations between mineral levels and seroconversion and seroprotection of HI titers for each influenza A strain. RESULTS Participants were 61% white, 28% male, 39% diabetic, and 81% overweight/obese with a mean age of 62.6 y. In logistic regression, Day 21 A/H1N1 seroprotection was associated with P and Zn at Day 21(P < 0.05). Seroconversion of A/H1N1 was associated with Day 21 Cu, P, and Mg (P < 0.03). Day 21 A/H3N2 seroprotection and seroconversion were associated with Day 21 P (P < 0.05). CONCLUSIONS Phosphorus was associated with seroprotection and seroconversion to influenza A after vaccination; these associations warrant additional studies with larger, more diverse population groups.
Collapse
Affiliation(s)
- Ata Murat Kaynar
- a Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) Laboratory, Department of Critical Care Medicine , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,b Department of Environmental and Occupational Health , University of Pittsburgh Graduate School of Public Health , Pittsburgh , PA , USA
| | - Mary Patricia Nowalk
- c Department of Family Medicine , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Chyongchiou Jeng Lin
- c Department of Family Medicine , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Krissy K Moehling
- c Department of Family Medicine , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Michael Susick
- c Department of Family Medicine , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Veli Bakalov
- d Department of Critical Care Medicine , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Bruce R Pitt
- d Department of Critical Care Medicine , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Daniel J Bain
- e Department of Geology and Environmental Science , University of Pittsburgh , Pittsburgh , PA , USA
| | - Ted M Ross
- f Vaccine and Gene Therapy Institute of Florida , Port St. Lucie , FL , USA
| | - Sean G Saul
- c Department of Family Medicine , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Mahlon Raymund
- c Department of Family Medicine , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Richard K Zimmerman
- c Department of Family Medicine , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| |
Collapse
|
40
|
Nakajima A, Oda S, Yokoi T. Allopurinol induces innate immune responses through mitogen-activated protein kinase signaling pathways in HL-60 cells. J Appl Toxicol 2015; 36:1120-8. [PMID: 26641773 DOI: 10.1002/jat.3272] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/21/2015] [Accepted: 10/29/2015] [Indexed: 11/06/2022]
Abstract
Allopurinol, an inhibitor of xanthine oxidase, is a frequent cause of severe cutaneous adverse reactions (SCARs) in humans, including drug rash with eosinophilia and systemic symptoms, Stevens-Johnson syndrome and toxic epidermal necrolysis. Although SCARs have been suspected to be immune-mediated, the mechanisms of allopurinol-induced SCARs remain unclear. In this study, we examined whether allopurinol has the ability to induce innate immune responses in vitro using human dendritic cell (DC)-like cell lines, including HL-60, THP-1 and K562, and a human keratinocyte cell line, HaCaT. In this study, we demonstrate that treatment of HL-60 cells with allopurinol significantly increased the mRNA expression levels of interleukin-8, monocyte chemotactic protein-1 and tumor necrosis factor α in a time- and concentration-dependent manner. Furthermore, allopurinol induced the phosphorylation of mitogen-activated protein kinases (MAPK), such as c-Jun N-terminal kinase and extracellular signal-regulated kinase, which regulate cytokine production in DC. In addition, allopurinol-induced increases in cytokine expression were inhibited by co-treatment with the MAPK inhibitors. Collectively, these results suggest that allopurinol has the ability to induce innate immune responses in a DC-like cell line through activation of the MAPK signaling pathways. These results indicate that innate immune responses induced by allopurinol might be involved in the development of allopurinol-induced SCARs. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Akira Nakajima
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Shingo Oda
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tsuyoshi Yokoi
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
41
|
Papadopoulos A, Gagnaire A, Degos C, de Chastellier C, Gorvel JP. Brucella discriminates between mouse dendritic cell subsets upon in vitro infection. Virulence 2015; 7:33-44. [PMID: 26606688 PMCID: PMC4871654 DOI: 10.1080/21505594.2015.1108516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Brucella is a Gram-negative bacterium responsible for brucellosis, a worldwide re-emerging zoonosis. Brucella has been shown to infect and replicate within Granulocyte macrophage colony-stimulating factor (GMCSF) in vitro grown bone marrow-derived dendritic cells (BMDC). In this cell model, Brucella can efficiently control BMDC maturation. However, it has been shown that Brucella infection in vivo induces spleen dendritic cells (DC) migration and maturation. As DCs form a complex network composed by several subpopulations, differences observed may be due to different interactions between Brucella and DC subsets. Here, we compare Brucella interaction with several in vitro BMDC models. The present study shows that Brucella is capable of replicating in all the BMDC models tested with a high infection rate at early time points in GMCSF-IL15 DCs and Flt3l DCs. GMCSF-IL15 DCs and Flt3l DCs are more activated than the other studied DC models and consequently intracellular bacteria are not efficiently targeted to the ER replicative niche. Interestingly, GMCSF-DC and GMCSF-Flt3l DC response to infection is comparable. However, the key difference between these 2 models concerns IL10 secretion by GMCSF DCs observed at 48 h post-infection. IL10 secretion can explain the weak secretion of IL12p70 and TNFα in the GMCSF-DC model and the low level of maturation observed when compared to GMCSF-IL15 DCs and Flt3l DCs. These models provide good tools to understand how Brucella induce DC maturation in vivo and may lead to new therapeutic design using DCs as cellular vaccines capable of enhancing immune response against pathogens.
Collapse
Affiliation(s)
- Alexia Papadopoulos
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Aurélie Gagnaire
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Clara Degos
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Chantal de Chastellier
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| | - Jean-Pierre Gorvel
- a Centre d'Immunologie de Marseille-Luminy; Aix Marseille Université; CNRS UMR7280; INSERM U1104 ; Marseille , France
| |
Collapse
|
42
|
Zhang JX, Li BL, Lin ZQ, Zhang N, Peng X, Gong ZH, Long LC, Zhou X, Xiang DC. Decrease in circulating myeloid dendritic cell precursors in patients with intracranial large artery atherosclerosis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:11495-11502. [PMID: 26617881 PMCID: PMC4637697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 07/23/2015] [Indexed: 06/05/2023]
Abstract
Intracranial large artery atherosclerosis (ILAA) is a major cause of ischemic cerebrovascular disease. The aim of this study was to investigate whether the levels of circulating dendritic cell precursors (DCP) could reflect the severity of intracranial large artery atherosclerosis (ILAA). For this purpose, a series of angiography were taken to determine the severity and extent of coronary artery and intracranial large artery stenosis, and flow cytometry were taken to determine the levels of circulating mDC precursors and pDC precursors in patients with severe intracranial large artery atherosclerosis (ILAA) (n = 101) and mild intracranial large artery atherosclerosis (ILAA) (n = 123) according to the angiography. Circulating mDC precursors were lower in patients with severe intracranial large artery atherosclerosis (ILAA) than in mild intracranial large artery atherosclerosis (ILAA) (P < 0.05), but circulating pDC precursors were not significant differences (P > 0.05). According to these data, circulating mDC precursors could predict the severity of ILAA, which also could be able to reflect the severity of ILAA.
Collapse
Affiliation(s)
- Jin-Xia Zhang
- Department of Cardiovascular, Guangzhou General Hospital of Guangzhou Military CommandGuangzhou 510010, China
| | - Bing-Ling Li
- Department of Pharmacy, Guangzhou General Hospital of Guangzhou Military CommandGuangzhou 510010, China
| | - Zhong-Qiu Lin
- Department of Elderly Cardiovascular, Guangzhou General Hospital of Guangzhou Military CommandGuangzhou 510010, China
| | - Ni Zhang
- Department of Cardiovascular, Guangzhou General Hospital of Guangzhou Military CommandGuangzhou 510010, China
| | - Xiong Peng
- Department of Cardiovascular, Guangzhou General Hospital of Guangzhou Military CommandGuangzhou 510010, China
| | - Zhi-Hua Gong
- Department of Cardiovascular, Guangzhou General Hospital of Guangzhou Military CommandGuangzhou 510010, China
| | - Liu-Cheng Long
- Department of Cardiovascular, Guangzhou General Hospital of Guangzhou Military CommandGuangzhou 510010, China
| | - Xuan Zhou
- Department of Cardiovascular, Guangzhou General Hospital of Guangzhou Military CommandGuangzhou 510010, China
| | - Ding-Cheng Xiang
- Department of Cardiovascular, Guangzhou General Hospital of Guangzhou Military CommandGuangzhou 510010, China
| |
Collapse
|
43
|
Kimura Y, Fujimura C, Ito Y, Takahashi T, Nakajima Y, Ohmiya Y, Aiba S. Optimization of the IL-8 Luc assay as an in vitro test for skin sensitization. Toxicol In Vitro 2015; 29:1816-30. [PMID: 26187477 DOI: 10.1016/j.tiv.2015.07.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 06/25/2015] [Accepted: 07/06/2015] [Indexed: 01/22/2023]
Abstract
We previously reported a dataset of the IL-8 Luc assay covering reference chemicals published by ECVAM, in which the effects of chemicals on IL-8 promoter activity were evaluated by an IL-8 reporter cell line, THP-G8 cells. To clarify its performance, we created another dataset of 88 sensitizers and 34 non-sensitizers. Simultaneously, to improve its performance, we changed the incubation time from 5 h to 16 h, deleted the criterion regarding the effects of N-acetylcysteine, and set an exclusion criterion for detergents. These modifications significantly improved its performance. In addition, we examined the following three criteria to judge chemicals as sensitizers: Criterion 1: Fold induction of SLO luciferase activity (FlnSLO-LA)⩾1.4, Criterion 2: the lower limit of the 95% confidence interval of FInSLO-LA⩾1.0, Criterion 3: the intersection of criteria 1 and 2. Among them, Criterion 1 produced the best performance, demonstrating that the accuracy, sensitivity and specificity were 81%, 79%, and 90%, respectively. In addition, we found that the IL-8 Luc assay solubilizing chemicals with X-VIVO substantially improved its performance. Finally, the IL-8 Luc assay combined with DPRA and DEREK could improve substantially its performance. These data suggest that the IL-8 Luc assay is a promising test method to screen skin sensitizers.
Collapse
Affiliation(s)
- Yutaka Kimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa 761-0395, Japan; Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Chizu Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa 761-0395, Japan; Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Yumiko Ito
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa 761-0395, Japan; Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Toshiya Takahashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa 761-0395, Japan; Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Yoshihiro Nakajima
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa 761-0395, Japan; Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Yoshihiro Ohmiya
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa 761-0395, Japan; Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan; Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa 761-0395, Japan; Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan.
| |
Collapse
|
44
|
Cui G, Chen J, He J, Lu C, Wei Y, Wang L, Xu X, Li L, Uede T, Diao H. Osteopontin promotes dendritic cell maturation and function in response to HBV antigens. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3003-16. [PMID: 26109844 PMCID: PMC4472071 DOI: 10.2147/dddt.s81656] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Purpose Dendritic cells (DCs) play critical roles in promoting innate and adaptive immunity in microbial infection. Functional impairment of DCs may mediate the suppression of viral-specific T-cell immune response in chronic hepatitis B (CHB) patients. Osteopontin (OPN) is involved in several liver diseases and infectious diseases. However, whether OPN affects DC function in hepatitis B virus (HBV) infection is unknown. Methods Twenty CHB patients and 20 healthy volunteers were recruited. OPN secreted by DCs was compared. Peripheral blood mononuclear cells cultured with OPN antibody were examined to study the costimulatory molecular expression and interleukin (IL)-12 production of DCs after HBV antigenic stimulation. OPN-deficient mice were used to investigate the influence of OPN on DC maturation and function after HBV antigenic stimulation in vitro and in vivo. Exogenous OPN was administrated to further verify the functioning of DCs from CHB patients upon HBV antigenic stimulation. Results We found that OPN production of DCs from CHB patients was significantly lower than those from healthy volunteers. The absence of OPN impaired IL-12 production and costimulatory molecular expression of DCs upon stimulation with HBV antigens. Defective DC function led to reduced activation of Th1 response to HBV antigens. In addition, OPN deficiency in DCs reduced the HBV antigen-induced inflammatory response in the liver of mice. Importantly, OPN administration significantly promoted the maturation of DCs from CHB patients in vitro. Conclusion These findings suggested that OPN could improve the maturation and functioning of DCs in the immune response to HBV antigens, which might be useful to further improve the effect of DC vaccine.
Collapse
Affiliation(s)
- Guangying Cui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China ; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, People's Republic of China
| | - Jianing Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China ; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, People's Republic of China
| | - Jianqin He
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China ; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, People's Republic of China
| | - Chong Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China ; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, People's Republic of China
| | - Yingfeng Wei
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China ; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, People's Republic of China
| | - Lin Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China ; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, People's Republic of China
| | - Xuejun Xu
- Department of Oral Orthodontics, Affiliated Stomatology Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China ; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, People's Republic of China
| | - Toshimitsu Uede
- Molecular Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China ; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
45
|
Banin-Hirata BK, Losi-Guembarovski R, Oda JMM, de Oliveira CEC, Campos CZ, Mazzuco TL, Borelli SD, Ceribelli JR, Watanabe MAE. CCR2-V64I genetic polymorphism: a possible involvement in HER2+ breast cancer. Clin Exp Med 2015; 16:139-45. [DOI: 10.1007/s10238-015-0342-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/16/2015] [Indexed: 12/14/2022]
|
46
|
SATOH-NAKAMURA T, KUROSE N, KAWANAMI T, NAKAMURA T, IWAO-KAWANAMI H, NAKAJIMA A, MIKI M, SAKAI T, FUJITA Y, TANAKA M, FUKUSHIMA T, MASAKI Y. CD14+ follicular dendritic cells in lymphoid follicles may play a role in the pathogenesis of IgG4-related disease . Biomed Res 2015; 36:143-53. [DOI: 10.2220/biomedres.36.143] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
| | - Nozomu KUROSE
- Department of Pathology and Laboratory Medicine, Kanazawa Medical University
| | - Takafumi KAWANAMI
- Department of Hematology and Immunology, Kanazawa Medical University
| | - Takuji NAKAMURA
- Department of Hematology and Immunology, Kanazawa Medical University
| | | | - Akio NAKAJIMA
- Department of Hematology and Immunology, Kanazawa Medical University
| | - Miyuki MIKI
- Department of Hematology and Immunology, Kanazawa Medical University
| | - Tomoyuki SAKAI
- Department of Hematology and Immunology, Kanazawa Medical University
| | - Yoshimasa FUJITA
- Department of Hematology and Immunology, Kanazawa Medical University
| | - Masao TANAKA
- Department of Hematology and Immunology, Kanazawa Medical University
| | | | - Yasufumi MASAKI
- Department of Hematology and Immunology, Kanazawa Medical University
| |
Collapse
|
47
|
Heuss ND, Pierson MJ, Montaniel KRC, McPherson SW, Lehmann U, Hussong SA, Ferrington DA, Low WC, Gregerson DS. Retinal dendritic cell recruitment, but not function, was inhibited in MyD88 and TRIF deficient mice. J Neuroinflammation 2014; 11:143. [PMID: 25116321 PMCID: PMC4149240 DOI: 10.1186/s12974-014-0143-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 07/29/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Immune system cells are known to affect loss of neurons due to injury or disease. Recruitment of immune cells following retinal/CNS injury has been shown to affect the health and survival of neurons in several models. We detected close, physical contact between dendritic cells and retinal ganglion cells following an optic nerve crush, and sought to understand the underlying mechanisms. METHODS CD11c-DTR/GFP mice producing a chimeric protein of diphtheria toxin receptor (DTR) and GFP from a transgenic CD11c promoter were used in conjunction with mice deficient in MyD88 and/or TRIF. Retinal ganglion cell injury was induced by an optic nerve crush, and the resulting interactions of the GFPhi cells and retinal ganglion cells were examined. RESULTS Recruitment of GFPhi dendritic cells to the retina was significantly compromised in MyD88 and TRIF knockout mice. GFPhi dendritic cells played a significant role in clearing fluorescent-labeled retinal ganglion cells post-injury in the CD11c-DTR/GFP mice. In the TRIF and MyD88 deficient mice, the resting level of GFPhi dendritic cells was lower, and their influx was reduced following the optic nerve crush injury. The reduction in GFPhi dendritic cell numbers led to their replacement in the uptake of fluorescent-labeled debris by GFPlo microglia/macrophages. Depletion of GFPhi dendritic cells by treatment with diphtheria toxin also led to their displacement by GFPlo microglia/macrophages, which then assumed close contact with the injured neurons. CONCLUSIONS The contribution of recruited cells to the injury response was substantial, and regulated by MyD88 and TRIF. However, the presence of these adaptor proteins was not required for interaction with neurons, or the phagocytosis of debris. The data suggested a two-niche model in which resident microglia were maintained at a constant level post-optic nerve crush, while the injury-stimulated recruitment of dendritic cells and macrophages led to their transient appearance in numbers equivalent to or greater than the resident microglia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dale S Gregerson
- Department of Ophthalmology & Visual Neurosciences, University of Minnesota, Lions Research Bldg, Rm 314, 2001 6th St SE, Minneapolis 55455, MN, USA.
| |
Collapse
|
48
|
Zweemer AJ, Bunnik J, Veenhuizen M, Miraglia F, Lenselink EB, Vilums M, de Vries H, Gibert A, Thiele S, Rosenkilde MM, IJzerman AP, Heitman LH. Discovery and Mapping of an Intracellular Antagonist Binding Site at the Chemokine Receptor CCR2. Mol Pharmacol 2014; 86:358-68. [DOI: 10.1124/mol.114.093328] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
49
|
Paccosi S, Musilli C, Caporale R, Gelli AMG, Guasti D, Clemente AM, Torcia MG, Filippelli A, Romagnoli P, Parenti A. Stimulatory interactions between human coronary smooth muscle cells and dendritic cells. PLoS One 2014; 9:e99652. [PMID: 24932497 PMCID: PMC4059651 DOI: 10.1371/journal.pone.0099652] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/16/2014] [Indexed: 01/26/2023] Open
Abstract
Despite inflammatory and immune mechanisms participating to atherogenesis and dendritic cells (DCs) driving immune and non-immune tissue injury response, the interactions between DCs and vascular smooth muscle cells (VSMCs) possibly relevant to vascular pathology including atherogenesis are still unclear. To address this issue, immature DCs (iDCs) generated from CD14+ cells isolated from healthy donors were matured either with cytokines (mDCs), or co-cultured (ccDCs) with human coronary artery VSMCs (CASMCs) using transwell chambers. Co-culture induced DC immunophenotypical and functional maturation similar to cytokines, as demonstrated by flow cytometry and mixed lymphocyte reaction. In turn, factors from mDCs and ccDCs induced CASMC migration. MCP-1 and TNFα, secreted from DCs, and IL-6 and MCP-1, secreted from CASMCs, were primarily involved. mDCs adhesion to CASMCs was enhanced by CASMC pre-treatment with IFNγ and TNFα ICAM-1 and VCAM-1 were involved, since the expression of specific mRNAs for these molecules increased and adhesion was inhibited by neutralizing antibodies to the counter-receptors CD11c and CD18. Adhesion was also inhibited by CASMC pre-treatment with the HMG-CoA-reductase inhibitor atorvastatin and the PPARγ agonist rosiglitazone, which suggests a further mechanism for the anti-inflammatory action of these drugs. Adhesion of DCs to VSMCs was shown also in vivo in rat carotid 7 to 21 days after crush and incision injury. The findings indicate that DCs and VSMCs can interact with reciprocal stimulation, possibly leading to perpetuate inflammation and vascular wall remodelling, and that the interaction is enhanced by a cytokine-rich inflammatory environment and down-regulated by HMGCoA-reductase inhibitors and PPARγ agonists.
Collapse
Affiliation(s)
- Sara Paccosi
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence, Italy
| | - Claudia Musilli
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence, Italy
| | - Roberto Caporale
- Central Laboratory, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | | | - Daniele Guasti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Ann Maria Clemente
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Maria Gabriella Torcia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Amelia Filippelli
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - Paolo Romagnoli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Astrid Parenti
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence, Italy
- * E-mail:
| |
Collapse
|
50
|
Deficiency of lymph node-resident dendritic cells (DCs) and dysregulation of DC chemoattractants in a malnourished mouse model of Leishmania donovani infection. Infect Immun 2014; 82:3098-112. [PMID: 24818662 DOI: 10.1128/iai.01778-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Malnutrition is thought to contribute to more than one-third of all childhood deaths via increased susceptibility to infection. Malnutrition is a significant risk factor for the development of visceral leishmaniasis, which results from skin inoculation of the intracellular protozoan Leishmania donovani. We previously established a murine model of childhood malnutrition and found that malnutrition decreased the lymph node barrier function and increased the early dissemination of L. donovani. In the present study, we found reduced numbers of resident dendritic cells (conventional and monocyte derived) but not migratory dermal dendritic cells in the skin-draining lymph nodes of L. donovani-infected malnourished mice. Expression of chemokines and their receptors involved in trafficking of dendritic cells and their progenitors to the lymph nodes was dysregulated. C-C chemokine receptor type 2 (CCR2) and its ligands (CCL2 and CCL7) were reduced in the lymph nodes of infected malnourished mice, as were CCR2-bearing monocytes/macrophages and monocyte-derived dendritic cells. However, CCR7 and its ligands (CCL19 and CCL21) were increased in the lymph node and CCR7 was increased in lymph node macrophages and dendritic cells. CCR2-deficient mice recapitulated the profound reduction in the number of resident (but not migratory dermal) dendritic cells in the lymph node but showed no alteration in the expression of CCL19 and CCL21. Collectively, these results suggest that the malnutrition-related reduction in the lymph node barrier to dissemination of L. donovani is related to insufficient numbers of lymph node-resident but not migratory dermal dendritic cells. This is likely driven by the altered activity of the CCR2 and CCR7 chemoattractant pathways.
Collapse
|