1
|
Planckaert G, Burssens A, Stappers F, Coudenys J, Demolder S, Kaya I, Van der Linden M, Gonçalves A, Lemeire K, Pavie B, Van Ovost E, Burssens P, Vanhaecke A, Van Dorpe J, Pringels L, Wezenbeek E, Snedeker J, De Bock K, Bonar F, Cook J, Victor J, Elewaut D, Gracey E. AI-driven histologic analysis of human Achilles tendinopathy provides a roadmap to unravel pathogenesis. Ann Rheum Dis 2025; 84:866-876. [PMID: 39922782 DOI: 10.1016/j.ard.2025.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 02/10/2025]
Abstract
OBJECTIVES Achilles tendinopathy is a common source of pain and dysfunction, yet its pathogenesis remains poorly understood. Research on human tendons is hampered by lack of standardisation in tissue sample validation, making interpretation of results challenging. We sought to develop an automated and operator-independent approach to histologically score human tendons. METHODS We assembled a cohort of 15 tendinopathic and 10 normal control Achilles tendon samples. We stained longitudinal sections with haematoxylin and eosin and Alcian blue and developed a low temperature epitope-retrieval protocol for immunostaining of blood vessels. Histologic sections were scored by pathologists using the current gold standard Bonar score. Whole sections were then analysed with open-source software (QuPath). Histologic features were automatically quantified across the entire section and summarised in the BonAIr score. Tissue from the same patients was subsequently analysed by quantitative polymerase chain reaction and flow cytometry to validate elements of the BonAIr score. RESULTS We observed increased cell roundness, collagen disarrangement, ground substance, and vascularity in tendinopathy using both the Bonar and BonAIr scores. Increased cellularity was only detected by the BonAIr score. Cellular and transcriptomic analyses corroborated tendinopathic shifts in all elements of the BonAIr score and further identified elevated THY1/CD90 expression in tendinopathy. CD90+ cells were found to localise to areas of low collagen alignment. These results align with the concept of stromal cell dysregulation in tendinopathy. CONCLUSIONS Automated analysis of whole tendon sections refines conventional histopathologic scoring and predicts cellular and molecular changes found in tendinopathy. The BonAIr score should be further developed for standardised assessment of tendons samples across other anatomical locations and different research centres.
Collapse
Affiliation(s)
- Guillaume Planckaert
- Unit for Molecular Immunology and Inflammation, Center for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium; Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Department of Orthopaedics and Traumatology, Ghent University Hospital, Ghent, Belgium
| | - Arne Burssens
- Unit for Molecular Immunology and Inflammation, Center for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium; Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Department of Orthopaedics and Traumatology, Ghent University Hospital, Ghent, Belgium
| | - Flore Stappers
- Unit for Molecular Immunology and Inflammation, Center for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium; Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Julie Coudenys
- Unit for Molecular Immunology and Inflammation, Center for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium; Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Sofía Demolder
- Unit for Molecular Immunology and Inflammation, Center for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium; Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Irem Kaya
- Department of Pathology, Ghent University Hospital, Ghent, Belgium; Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Malaïka Van der Linden
- Department of Pathology, Ghent University Hospital, Ghent, Belgium; Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Amanda Gonçalves
- Bioimaging Core, Center for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
| | - Kelly Lemeire
- Bioimaging Core, Center for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
| | - Benjamin Pavie
- Bioimaging Core, Center for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
| | - Edwin Van Ovost
- Department of Orthopaedics and Traumatology, General Hospital (AZ) Sint-Lucas, Ghent, Belgium
| | - Peter Burssens
- Department of Orthopaedics and Traumatology, General Hospital (AZ) Jan-Palfijn, Ghent, Belgium
| | - Amber Vanhaecke
- Department of Pathology, Ghent University Hospital, Ghent, Belgium; Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, Ghent, Belgium; Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Lauren Pringels
- Department of Rehabilitation Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Evi Wezenbeek
- Movement Antwerp (MOVANT) Research Group, Department of Rehabilitation Sciences and Physiotherapy (REVAKI), University of Antwerp, Wilrijk, Belgium
| | - Jess Snedeker
- Department of Health Sciences and Technology, Federal Institute of Technology (ETH) Zürich, Zürich, Switzerland
| | - Katrien De Bock
- Department of Health Sciences and Technology, Federal Institute of Technology (ETH) Zürich, Zürich, Switzerland
| | - Fiona Bonar
- Douglass Hanly Moir Pathology, Macquarie Park, Sydney, NSW, Australia
| | - Jill Cook
- Sport and Exercise Medicine Research Centre, La Trobe University, Melbourne, Victoria, Australia
| | - Jan Victor
- Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Department of Orthopaedics and Traumatology, Ghent University Hospital, Ghent, Belgium
| | - Dirk Elewaut
- Unit for Molecular Immunology and Inflammation, Center for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium; Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Department of Rheumatology, Ghent University Hospital, Ghent, Belgium.
| | - Eric Gracey
- Unit for Molecular Immunology and Inflammation, Center for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium; Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
2
|
Eissa MM, Allam SRA, Ismail CA, Ghazala RA, El Skhawy N, Zaki IIA, Ibrahim EIES. Unveiling the anti-neoplastic potential of Schistosoma mansoni-derived antigen against breast cancer: a pre-clinical study. Eur J Med Res 2025; 30:304. [PMID: 40247360 PMCID: PMC12007238 DOI: 10.1186/s40001-025-02531-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Cancer is a global health concern, with millions of new cases and deaths annually. Recently, immunotherapy has strengthened cancer treatment by harnessing the body's immune system to fight cancer. The search for advanced cancer immunotherapies has expanded to explore pathogens like parasites for their potential anti-neoplastic effects. While some parasites have shown promising results, the role of Schistosoma mansoni in breast cancer remains unexplored. METHODS This pre-clinical study investigated the anti-neoplastic potential of autoclaved Schistosoma mansoni antigen against breast cancer. In vitro, autoclaved Schistosoma mansoni antigen was evaluated on the MCF-7 human breast cancer cell line, while in vivo experiments used a chemically induced breast cancer rat model to evaluate tumour growth, liver enzyme levels, and immune response. Histopathological and immunohistochemical analyses assessed changes in tumour tissue, cell proliferation (Ki-67), angiogenesis (CD31), immune cell infiltration (CD8+ T cells), regulatory T cells (FoxP3+), and programmed death ligand 1 (PD-L1) expression. RESULTS In vitro, autoclaved Schistosoma mansoni antigen significantly reduced MCF-7 cell viability in a dose- and time-dependent manner. In vivo, autoclaved Schistosoma mansoni antigen treatment significantly reduced tumour weight and volume, improved liver enzyme levels, increased tumour necrosis, and decreased fibrosis. Immunohistochemical analysis revealed decreased Ki-67 and CD31 expression, indicating reduced cell proliferation and angiogenesis, respectively. Autoclaved Schistosoma mansoni antigen also enhanced immune responses by increasing CD8+ T cells infiltration and decreasing FoxP3+ expression, resulting in a higher CD8+ T cells/FoxP3+ ratio within the tumour microenvironment. Notably, PD-L1 expression was also downregulated, suggesting potential immune checkpoint inhibition. CONCLUSIONS Autoclaved Schistosoma mansoni antigen demonstrated potent anti-neoplastic activity, significantly reducing tumour growth and modulating the immune response within the tumour microenvironment. These results highlight autoclaved Schistosoma mansoni antigen's potential as a novel immunotherapy for breast cancer.
Collapse
Affiliation(s)
- Maha Mohamed Eissa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Al-Moassat Medical Campus, Alexandria, Egypt.
| | - Sonia Rifaat Ahmed Allam
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Al-Moassat Medical Campus, Alexandria, Egypt
| | - Cherine Adel Ismail
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Rasha Abdelmawla Ghazala
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nahla El Skhawy
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Al-Moassat Medical Campus, Alexandria, Egypt
| | | | - Eman Ibrahim El-Said Ibrahim
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Al-Moassat Medical Campus, Alexandria, Egypt
| |
Collapse
|
3
|
Jiang H, Guo Y, Tan X, Jiang Y, Pang N, Niu C, Liu L, Zhou Z, Liu L, Li H. Oxidized cellulose microneedle patch combined with vascular embolization and local delivery of timolol maleate for hemangiomas. Colloids Surf B Biointerfaces 2024; 244:114174. [PMID: 39197327 DOI: 10.1016/j.colsurfb.2024.114174] [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: 06/20/2024] [Revised: 08/09/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Hemangiomas are superficial tumors characterized by dense vascular structures that often affect the patient's aesthetic appearance due to the obvious red appearance on the skin. Current treatments, especially timolol maleate in the form of eye drops and hydrogels, suffer from low transdermal drug delivery rates, resulting in prolonged treatment time. To address this challenge, our study introduced a soluble microneedle patch with dextran as the main material to form microcatheters for sustained delivery of timolol maleate. In addition, we proposed a vascular embolization strategy to disrupt the blood supply in hemangiomas. Oxidized cellulose (C-cellulose) was selected for its excellent hemostatic properties. We incorporated C-cellulose into dextran microneedles to facilitate thrombosis in the vascular-rich areas of hemangiomas. The innovative microneedle patch we developed can penetrate the skin to a depth of 430 μm and dissolve rapidly within 3 minutes, ensuring direct drug delivery to the subcutaneous layer. Notably, the treated skin area regained its original appearance within two hours after treatment. In addition to excellent skin permeability and rapid dissolution, these patches significantly promoted apoptosis and inhibited cell migration in mouse hemangioendothelioma EOMA cells. Our results demonstrate that this approach not only achieves significant tumor inhibition in a mouse hemangioma model, but also represents a more effective, convenient, and non-invasive treatment option. Therefore, dextran/C-cellulose/timolol maleate microneedle patch (MNs/Timolol) has broad clinical application prospects in the treatment of hemangiomas, minimizing the risk of additional damage and improving treatment efficacy.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Interventional Hemangioma, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510600, China
| | - Yiqun Guo
- Department of Interventional Hemangioma, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510600, China
| | - Xiaoyun Tan
- Department of Interventional Hemangioma, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510600, China
| | - Yizhou Jiang
- Department of Interventional Hemangioma, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510600, China
| | - Ningdong Pang
- Department of Interventional Hemangioma, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510600, China
| | - Chuanqiang Niu
- Department of Interventional Hemangioma, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510600, China
| | - Lang Liu
- Department of Interventional Hemangioma, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510600, China
| | - Zijun Zhou
- Department of Interventional Hemangioma, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510600, China
| | - Lu Liu
- Department of Interventional Hemangioma, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510600, China
| | - Haibo Li
- Department of Interventional Hemangioma, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510600, China.
| |
Collapse
|
4
|
Amarilla-Irusta A, Zenarruzabeitia O, Sevilla A, Sandá V, Lopez-Pardo A, Astarloa-Pando G, Pérez-Garay R, Pérez-Fernández S, Meijide S, Imaz-Ayo N, Arana-Arri E, Amo L, Borrego F. CD151 identifies an NK cell subset that is enriched in COVID-19 patients and correlates with disease severity. J Infect 2024; 89:106304. [PMID: 39374860 DOI: 10.1016/j.jinf.2024.106304] [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: 04/22/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/09/2024]
Abstract
Severe coronavirus disease 2019 (COVID-19) often leads to acute respiratory distress syndrome and multi-organ dysfunction, driven by a dysregulated immune response, including a cytokine storm with elevated proinflammatory cytokine levels. Natural killer (NK) cells are part of the innate immune system with a fundamental role in the defense against viral infections. However, during COVID-19 acute infection, they exhibit an altered phenotype and impaired functionality contributing to the immunopathogenesis of the disease. In this work, we have studied a cohort of patients with COVID-19 (ranging from mild to severe) by analyzing IL-15, TGF-β, PlGF and GDF-15 plasma levels and performing multiparametric flow cytometry studies. Our results revealed that severe COVID-19 patients exhibited high levels of IL-15, PlGF and GDF-15, along with an enrichment of an NK cell subset expressing the CD151 tetraspanin, which correlated with IL-15 plasma levels and disease severity. In patients, these CD151+ NK cells displayed a more activated phenotype characterized by an increased expression of HLA-DR, CD38 and granzyme B, a distinct receptor repertoire, with lower levels of CD160 and CD31 and higher levels of CD55 and, remarkably, a higher expression of tissue-resident markers CD103 and the NK cell decidual marker CD9. Last of all, in individuals with severe disease, we identified an expansion of a CD151brightCD9+ NK cell subset, suggesting that these cells play a specific role in COVID-19. Altogether, our findings suggest that CD151+ NK cells may have a relevant role in COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Arrate Sevilla
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Víctor Sandá
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Ainara Lopez-Pardo
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | | | - Raquel Pérez-Garay
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Clinical Analysis Service, Cruces University Hospital, OSI Ezkerraldea-Enkarterri-Cruces, Barakaldo, Spain
| | - Silvia Pérez-Fernández
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Susana Meijide
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Natale Imaz-Ayo
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Eunate Arana-Arri
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Laura Amo
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Francisco Borrego
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
5
|
Yu J, Cai W, Zhou T, Men B, Chen S, Tu D, Guo W, Wang J, Zhao F, Wang Y. CEACAM1 increased the lymphangiogenesis through miR-423-5p and NF- kB in Non-Small Cell Lung Cancer. Biochem Biophys Rep 2024; 40:101833. [PMID: 39398537 PMCID: PMC11470192 DOI: 10.1016/j.bbrep.2024.101833] [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: 07/01/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024] Open
Abstract
Background Lung cancer causes significant mortality, with invasion and metastasis being the main features that cause most cancer deaths. Lymph node metastasis is the primary metastatic route in non-small cell carcinoma (NSCLC) and influences the staging and prognosis of NSCLC. Cumulative studies have reported that Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is involved in the progression of various cancers. However, few studies have discussed the function of CEACAM1 in lymphangiogenesis in NSCLC. Here, we examined how CEACAM1 influences lymphangiogenesis in NSCLC. Methods A total of 30 primary squamous cell carcinoma (LUSC) patients diagnosed with LN metastasis were prospectively selected. LUSC tumor tissues, para-cancerous tissues, and positive lymph node tissues were harvested. The expression and subcellular location of CEACAM1, CD31, and LVYE1 in clinical samples were detected by immunohistochemistry. Next, the CEACAM1 and hsa-miR-423-5p expressions were detected by qPCR. The protein expression of lymphangiogenesis-associated proteins and critical cytokines of the NF-κB pathway in HDLECs was detected by Western blot. A tube formation assay was performed to detect the lymphangiogenesis in different groups. The interaction between CEACAM1 and hsa-miR-423-5p was verified using a dual luciferase assay. Results CEACAM1 was found to be a potential gene associated with lung cancer prognosis. It was positively correlated with angiogenesis and lymphangiogenesis. Then, we detected the function of CEACAM1 in lymphangiogenesis and found that CEACAM1 promoted lymphangiogenesis. hsa-miR-423-5p overexpression inhibited lymphangiogenesis via targeting CEACAM1. Finally, we observed that CEACAM1 can activate the NF-κB pathway and, therefore, promote lymphangiogenesis. Conclusion We found that CEACAM1 enhanced lymphangiogenesis in NSCLC via NF-kB activation and was repressed by miR-423-5p. This suggests the value of CEACAM1 as a new therapeutic marker in NSCLC.
Collapse
Affiliation(s)
- Jie Yu
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Wenke Cai
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Tao Zhou
- Department of Respiration, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Bo Men
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Shunqiong Chen
- Department of Respiration, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Dong Tu
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Wei Guo
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Jicui Wang
- Department of Thoracocardiac Surgery, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Feipeng Zhao
- Department of Plastic and Maxillofacial Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Wang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
6
|
Evrard R, Manon J, Maistriaux L, Fievé L, Darius T, Cornu O, Lengelé B, Schubert T. Enhancing the biological integration of massive bone allografts: A porcine preclinical in vivo pilot-study. Bone 2024; 187:117213. [PMID: 39084545 DOI: 10.1016/j.bone.2024.117213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/02/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Critical bone loss can have several origins: infections, tumors or trauma. Therefore, massive bone allograft can be a solution for limb salvage. Such a biological reconstruction should have the ideal biomechanical qualities. However, their complication rate remains too high. Perfusion-decellularization of massive allografts could promote the vitality of these grafts, thereby improving their integration and bone remodeling. Three perfusion-decellularized massive bone allografts were compared to 3 fresh frozen massive bone allografts in a preclinical in vivo porcine study using an orthopedic surgery model. Three pigs each underwent a critical diaphyseal femoral defects followed by an allogeneic intercalary femoral graft on their both femurs (one decellularized and one conventional fresh frozen as "native") to reconstruct the defect. Clinical imaging was performed over 3 months of follow-up. The grafts were then explanted and examined by non-decalcified histology, fluoroscopic microscopy and immunohistochemistry. Bone consolidation was achieved in both groups at the same time. However, the volume of bone callus appeared to be greater in the decellularized group. Histology demonstrated a superior bone remodeling in the decellularized group, with a higher number of osteoclasts (p < 0.001) and larger areas of osteoid matrix and newly formed bone as compared to the "native" group. Immunohistochemistry showed a superior vitality and remodeling in both the cortical and medullary cavities for osteocalcin (p < 0.001), Ki67 (p < 0.001), CD3 (p < 0.001) and α-SMA (p < 0.001) as compared the "native" group. Three months after implantation, the decellularized grafts were proven to be biologically more active compared to native grafts. Fluoroscopic microscopy revealed more ossification fronts in the depth of the decellularized grafts (p = 0.021). This pilot study provides the first in vivo demonstration on the enhanced biological capacities of massive bone allograft decellularized by perfusion as compared to conventional massive bone allografts.
Collapse
Affiliation(s)
- Robin Evrard
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium.
| | - Julie Manon
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium; Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium
| | - Louis Maistriaux
- Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium
| | - Lies Fievé
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium
| | - Tom Darius
- Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Département de Chirurgie, Chirurgie abdominale et unité de transplantation, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium
| | - Olivier Cornu
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium
| | - Benoit Lengelé
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Service de Chirurgie Plastique, Reconstructrice et Esthétique, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium
| | - Thomas Schubert
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium
| |
Collapse
|
7
|
Smits HJG, Bennink E, Ruiter LN, Breimer GE, Willems SM, Dankbaar JW, Philippens MEP. Spatial correlation between in vivo imaging and immunohistochemical biomarkers: A methodological study. Transl Oncol 2024; 48:102051. [PMID: 39018773 DOI: 10.1016/j.tranon.2024.102051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/09/2024] [Accepted: 07/01/2024] [Indexed: 07/19/2024] Open
Abstract
In this study, we present a method that enables voxel-by-voxel comparison of in vivo imaging to immunohistochemistry (IHC) biomarkers. As a proof of concept, we investigated the spatial correlation between dynamic contrast enhanced (DCE-)CT parameters and IHC biomarkers Ki-67 (proliferation), HIF-1α (hypoxia), and CD45 (immune cells). 54 whole-mount tumor slices of 15 laryngeal and hypopharyngeal carcinomas were immunohistochemically stained and digitized. Heatmaps of biomarker positivity were created and registered to DCE-CT parameter maps. The adiabatic approximation to the tissue homogeneity model was used to fit the following DCE parameters: Ktrans (transfer constant), Ve (extravascular and extracellular space), and Vi (intravascular space). Both IHC and DCE maps were downsampled to 4 × 4 × 3 mm[3] voxels. The mean values per tumor were used to calculate the between-subject correlations between parameters. For the within-subject (spatial) correlation, values of all voxels within a tumor were compared using the repeated measures correlation (rrm). No between-subject correlations were found between IHC biomarkers and DCE parameters, whereas we found multiple significant within-subject correlations: Ve and Ki-67 (rrm = -0.17, P < .001), Ve and HIF-1α (rrm = -0.12, P < .001), Ktrans and CD45 (rrm = 0.13, P < .001), Vi and CD45 (rrm = 0.16, P < .001), and Vi and Ki-67 (rrm = 0.08, P = .003). The strongest correlation was found between IHC biomarkers Ki-67 and HIF-1α (rrm = 0.35, P < .001). This study shows the technical feasibility of determining the 3 dimensional spatial correlation between histopathological biomarker heatmaps and in vivo imaging. It also shows that between-subject correlations do not reflect within-subject correlations of parameters.
Collapse
Affiliation(s)
- Hilde J G Smits
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Edwin Bennink
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Lilian N Ruiter
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Gerben E Breimer
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Stefan M Willems
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan W Dankbaar
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | |
Collapse
|
8
|
Evrard R, Manon J, Rafferty C, Fieve L, Cornu O, Kirchgesner T, Lecouvet FE, Schubert T, Lengele B. Vascular study of decellularized porcine long bones: Characterization of a tissue engineering model. Bone 2024; 182:117073. [PMID: 38493932 DOI: 10.1016/j.bone.2024.117073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
INTRODUCTION Massive bone allografts enable the reconstruction of critical bone defects in numerous conditions (e.g. tumoral, infection or trauma). Unfortunately, their biological integration remains insufficient and the reconstruction may suffer from several postoperative complications. Perfusion-decellularization emerges as a tissue engineering potential solution to enhance osseointegration. Therefore, an intrinsic vascular study of this novel tissue engineering tool becomes essential to understand its efficacy and applicability. MATERIAL AND METHODS 32 porcine long bones (humeri and femurs) were used to assess the quality of their vascular network prior and after undergoing a perfusion-decellularization protocol. 12 paired bones were used to assess the vascular matrix prior (N = 6) and after our protocol (N = 6) by immunohistochemistry. Collagen IV, Von Willebrand factor and CD31 were targeted then quantified. The medullary macroscopic vascular network was evaluated with 12 bones: 6 were decellularized and the other 6 were, as control, not treated. All 12 underwent a contrast-agent injection through the nutrient artery prior an angio CT-scan acquisition. The images were processed and the length of medullary vessels filled with contrast agent were measured on angiographic cT images obtained in control and decellularized bones by 4 independent observers to evaluate the vascular network preservation. The microscopic cortical vascular network was evaluated on 8 bones: 4 control and 4 decellularized. After injection of gelatinous fluorochrome mixture (calcein green), non-decalcified fluoroscopic microscopy was performed in order to assess the perfusion quality of cortical vascular lacunae. RESULTS The continuity of the microscopic vascular network was assessed with Collagen IV immunohistochemistry (p-value = 0.805) while the decellularization quality was observed through CD31 and Von Willebrand factor immunohistochemistry (p-values <0.001). The macroscopic vascular network was severely impaired after perfusion-decellularization; nutrient arteries were still patent but the amount of medullary vascular channels measured was significantly higher in the control group compared to the decellularized group (p-value <0.001). On average, the observers show good agreement on these results, except in the decellularized group where more inter-observer discrepancies were observed. The microscopic vascular network was observed with green fluoroscopic signal in almost every canals and lacunae of the bone cortices, in three different bone locations (proximal metaphysis, diaphysis and distal metaphysis). CONCLUSION Despite the aggressiveness of the decellularization protocol on medullary vessels, total porcine long bones decellularized by perfusion retain an acellular cortical microvascular network. By injection through the intact nutrient arteries, this latter vascular network can still be used as a total bone infusion access for bone tissue engineering in order to enhance massive bone allografts prior implantation.
Collapse
Affiliation(s)
- R Evrard
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium.
| | - J Manon
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - C Rafferty
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, UCLouvain, Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium
| | - L Fieve
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, UCLouvain, Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium
| | - O Cornu
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - T Kirchgesner
- Département d'Imagerie Médicale, Institut de Recherche Expérimentale et Clinique (Pôle IMAG), Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - F E Lecouvet
- Département d'Imagerie Médicale, Institut de Recherche Expérimentale et Clinique (Pôle IMAG), Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - T Schubert
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain (UCLouvain), Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| | - B Lengele
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, UCLouvain, Avenue E. Mounier, 52-B1.52.04, 1200 Bruxelles, Belgium; Service de Chirurgie Plastique, Reconstructrice et Esthétique, Cliniques Universitaires Saint-Luc, UCLouvain, Avenue Hippocrate 10, 1200 Bruxelles, Belgium
| |
Collapse
|
9
|
Rana M, Choubey P, Nandi G, Jain S, Bajaj D, Sharma S, Basu-Modak S. Expression of angiogenic factors in the placenta of heme oxygenase-1 deficient mouse embryo. Reprod Biol 2023; 23:100822. [PMID: 37979494 DOI: 10.1016/j.repbio.2023.100822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/20/2023]
Abstract
Heme oxygenase 1 (Hmox1), the inducible form of heme degrading enzymes Hmoxs, is important for establishment and maintenance of pregnancy. A growing body of evidence suggests an association between Hmox1 and angiogenesis, including placental angiogenesis. In this study, we examined the expression of two angiogenic factors in the placentas of Hmox1 deficient mouse embryos, whose expression was found to be related to that of Hmox1. Relative protein levels and localization of Hmoxs and two angiogenic factors [Vegf and Prolactin along with their receptors, and Cd31/Pecam1] were compared in the placentas of Hmox1 wildtype and knockout mouse embryos using western blotting and immunohistochemistry along with histological analysis. The results revealed tissue disorganisation, reduced area of labyrinth and smaller nuclear size of trophoblast giant cell in the placentas of knockout embryos. The levels of Hmox2, prolactin, and Cd31/Pecam1 were found to be altered in knockout placentas, whereas Vegf and its receptors seem to be unaltered in our samples. Overall, our findings imply that Hmox2 is unlikely to compensate for Hmox1 deficiency in knockout placentas, and altered levels of prolactin and Cd31/Pecam1 hint towards impaired angiogenesis in these placentas. Further investigation would be needed to understand the molecular mechanism of defective angiogenesis in the placentas of Hmox1 knockout mouse embryos.
Collapse
Affiliation(s)
- Meenakshi Rana
- Department of Zoology, University of Delhi, India; Department of Zoology, Dyal Singh College, University of Delhi, India.
| | | | - Gouri Nandi
- Department of Zoology, University of Delhi, India; Department of Zoology, Deshbandhu College, University of Delhi, India
| | - Sidhant Jain
- Department of Zoology, University of Delhi, India; Institute for Globally Distributed Open Research and Education, India
| | - Divya Bajaj
- Department of Zoology, University of Delhi, India; Department of Zoology, Hindu College, University of Delhi, India
| | - Sonika Sharma
- Department of Zoology, University of Delhi, India; Department of Zoology, Daulat Ram College, University of Delhi, India
| | | |
Collapse
|
10
|
Andreata F, Clément M, Benson RA, Hadchouel J, Procopio E, Even G, Vorbe J, Benadda S, Ollivier V, Ho-Tin-Noe B, Le Borgne M, Maffia P, Nicoletti A, Caligiuri G. CD31 signaling promotes the detachment at the uropod of extravasating neutrophils allowing their migration to sites of inflammation. eLife 2023; 12:e84752. [PMID: 37549051 PMCID: PMC10431918 DOI: 10.7554/elife.84752] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 08/04/2023] [Indexed: 08/09/2023] Open
Abstract
Effective neutrophil migration to sites of inflammation is crucial for host immunity. A coordinated cascade of steps allows intravascular leukocytes to counteract the shear stress, transmigrate through the endothelial layer, and move toward the extravascular, static environment. Those events are tightly orchestrated by integrins, but, while the molecular mechanisms leading to their activation have been characterized, the regulatory pathways promoting their detachment remain elusive. In light of this, it has long been known that platelet-endothelial cell adhesion molecule (Pecam1, also known as CD31) deficiency blocks leukocyte transmigration at the level of the outer vessel wall, yet the associated cellular defects are controversial. In this study, we combined an unbiased proteomic study with in vitro and in vivo single-cell tracking in mice to study the dynamics and role of CD31 during neutrophil migration. We found that CD31 localizes to the uropod of migrating neutrophils along with closed β2-integrin and is required for essential neutrophil actin/integrin polarization. Accordingly, the uropod of Pecam1-/- neutrophils is unable to detach from the extracellular matrix, while antagonizing integrin binding to extracellular matrix components rescues this in vivo migratory defect. Conversely, we showed that sustaining CD31 co-signaling actively favors uropod detachment and effective migration of extravasated neutrophils to sites of inflammation in vivo. Altogether, our results suggest that CD31 acts as a molecular rheostat controlling integrin-mediated adhesion at the uropod of egressed neutrophils, thereby triggering their detachment from the outer vessel wall to reach the inflammatory sites.
Collapse
Affiliation(s)
- Francesco Andreata
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Marc Clément
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Robert A Benson
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Juliette Hadchouel
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center (PARCC)ParisFrance
| | - Emanuele Procopio
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Guillaume Even
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Julie Vorbe
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Samira Benadda
- Cell and Tissue Imaging Platform, INSERM, CNRS, ERL8252, Centre de Recherche sur l’Inflammation (CRI)ParisFrance
| | - Véronique Ollivier
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Benoit Ho-Tin-Noe
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Marie Le Borgne
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUnited Kingdom
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico IINaplesItaly
| | - Antonino Nicoletti
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Giuseppina Caligiuri
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
- Department of Cardiology and of Physiology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Paris Nord Val-de-Seine, Site BichatParisFrance
| |
Collapse
|
11
|
FaragAllah EM, Algharib SA, Goda NIA, El-Malkey NF, Mahboub HH, El-Fayoumi SH, Al-Hoshani N, Wahid RM, Aref M. Protective effect of Salvia hispanica (chia seeds) against obesity induced ovarian disorders in rat model. Tissue Cell 2023; 83:102156. [PMID: 37437332 DOI: 10.1016/j.tice.2023.102156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/19/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023]
Abstract
The main goal of the current report is to assess the protective impacts of chia seeds against obesity-induced ovarian dysfunctions with a trial to elucidate the mechanism of action. Forty rats were divided into 4 groups including lean untreated, lean consuming chia seeds, obese untreated, and rats consumed high-fat diet (HFD) mixed with ground chia seeds for 10 weeks. Anthropometric measures including visceral fat, peri-ovarian fat, ovarian weights, and duration of the estrous cycle were computed. Serum luteinizing (LH), follicular stimulating (FSH), progesterone, estradiol hormones, and tumor necrosis-α (TNF-α) were estimated. Ovarian histopathology and immunohistochemistry (CD31) were performed. Results showed that chia seeds clearly reduced obesity and induced alteration in anthropometric measures with a clear increase in LH and progesterone. Such seeds notably reversed histopathological alteration and reduced TNF-α, and CD31 induced by HFD. Conclusively, chia seeds have a potential protective role against obesity-induced ovarian dysfunction owing to their anti-inflammatory properties.
Collapse
Affiliation(s)
- Eman Mahmoud FaragAllah
- Physiology Department, Faculty of Medicine, Zagazig University, 44511 Zagazig, Sharkia, Egypt
| | - Samah Attia Algharib
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Moshtohor, 13736 Toukh, QG, Egypt; National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, China
| | - Nehal I A Goda
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkia, Egypt
| | - Nanees F El-Malkey
- Physiology Department, Faculty of Medicine, Zagazig University, 44511 Zagazig, Sharkia, Egypt
| | - Heba H Mahboub
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkia, Egypt.
| | - Shaimaa H El-Fayoumi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Heliopolis University, Egypt
| | - Nawal Al-Hoshani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Reham M Wahid
- Physiology Department, Faculty of Medicine, Zagazig University, 44511 Zagazig, Sharkia, Egypt
| | - Mohamed Aref
- Anatomy Department, Faculty of Veterinary Medicine, Zagazig University, 44511 Zagazig, Sharkia, Egypt
| |
Collapse
|
12
|
Zhang Z, Gan Q, Han J, Tao Q, Qiu WQ, Madri JA. CD31 as a probable responding and gate-keeping protein of the blood-brain barrier and the risk of Alzheimer's disease. J Cereb Blood Flow Metab 2023; 43:1027-1041. [PMID: 37051650 PMCID: PMC10291450 DOI: 10.1177/0271678x231170041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 03/17/2023] [Accepted: 03/24/2023] [Indexed: 04/14/2023]
Abstract
Several studies have shown that an abnormal vascular-immunity link could increase Alzheimer's disease (AD) risk; however, the mechanism is unclear. CD31, also named platelet endothelial cell adhesion molecule (PECAM), is a surface membrane protein of both endothelial and immune cells and plays important roles in the interaction between the vascular and immune systems. In this review, we focus on research regarding CD31 biological actions in the pathological process that may contribute to AD based on the following rationales. First, endothelial, leukocyte and soluble forms of CD31 play multi-roles in regulating transendothelial migration, increasing blood-brain barrier (BBB) permeability and resulting in neuroinflammation. Second, CD31 expressed by endothelial and immune cells dynamically modulates numbers of signaling pathways, including Src family kinases, selected G proteins, and β-catenin which in turn affect cell-matrix and cell-cell attachment, activation, permeability, survival, and ultimately neuronal cell injury. In endothelia and immune cells, these diverse CD31-mediated pathways act as a critical regulator in the immunity-endothelia-brain axis, thereby mediating AD pathogenesis in ApoE4 carriers, which is the major genetic risk factor for AD. This evidence suggests a novel mechanism and potential drug target for CD31 in the background of genetic vulnerabilities and peripheral inflammation for AD development and progression.
Collapse
Affiliation(s)
- Zhengrong Zhang
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Qini Gan
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Jingyan Han
- Whitaker Cardiovascular Research Institute, Boston University School of Medicine, Boston, MA, USA
| | - Qiushan Tao
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Psychiatry, Boston University School of Medicine, Boston, MA, USA
- The Alzheimer’s Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Joseph A Madri
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
13
|
Danila E, Aleksonienė R, Besusparis J, Gruslys V, Jurgauskienė L, Laurinavičienė A, Laurinavičius A, Mainelis A, Zablockis R, Zeleckienė I, Žurauskas E, Malickaitė R. Lymphocyte Subsets and Pulmonary Nodules to Predict the Progression of Sarcoidosis. Biomedicines 2023; 11:biomedicines11051437. [PMID: 37239108 DOI: 10.3390/biomedicines11051437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
The search for biological markers, which allow a relatively accurate assessment of the individual course of pulmonary sarcoidosis at the time of diagnosis, remains one of the research priorities in this field of pulmonary medicine. The aim of our study was to investigate possible prognostic factors for pulmonary sarcoidosis with a special focus on cellular immune inflammation markers. A 2-year follow-up of the study population after the initial prospective and simultaneous analysis of lymphocyte activation markers expression in the blood, as well as bronchoalveolar lavage fluid (BALF) and lung biopsy tissue of patients with newly diagnosed pulmonary sarcoidosis, was performed. We found that some blood and BAL fluid immunological markers and lung computed tomography (CT) patterns have been associated with a different course of sarcoidosis. We revealed five markers that had a significant negative association with the course of sarcoidosis (worsening pulmonary function tests and/or the chest CT changes)-blood CD4+CD31+ and CD4+CD44+ T lymphocytes, BALF CD8+CD31+ and CD8+CD103+ T lymphocytes and a number of lung nodules on chest CT at the time of the diagnosis. Cut-off values, sensitivity, specificity and odds ratio for predictors of sarcoidosis progression were calculated. These markers may be reasonable predictors of sarcoidosis progression.
Collapse
Affiliation(s)
- Edvardas Danila
- Clinic of Chest Diseases, Immunology and Allergology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Regina Aleksonienė
- Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Justinas Besusparis
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, 08406 Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Vygantas Gruslys
- Clinic of Chest Diseases, Immunology and Allergology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Laimutė Jurgauskienė
- Clinic of Cardiac and Vascular Diseases, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Center of Laboratory Medicine, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Aida Laurinavičienė
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, 08406 Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Arvydas Laurinavičius
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, 08406 Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Antanas Mainelis
- Faculty of Mathematics and Informatics, Vilnius University, 03225 Vilnius, Lithuania
| | - Rolandas Zablockis
- Clinic of Chest Diseases, Immunology and Allergology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Ingrida Zeleckienė
- Center of Radiology and Nuclear Medicine, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Edvardas Žurauskas
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, 08406 Vilnius, Lithuania
| | - Radvilė Malickaitė
- Clinic of Cardiac and Vascular Diseases, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Center of Laboratory Medicine, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| |
Collapse
|
14
|
Xiang J, Zhong W. The molecular mechanism of Gorham syndrome: an update. Front Immunol 2023; 14:1165091. [PMID: 37215116 PMCID: PMC10196207 DOI: 10.3389/fimmu.2023.1165091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Gorham syndrome, also known as "vanishing osteopathy" and "invasive hemangiomatosis," is a rare clinical syndrome whose etiology is unknown and can invade the whole-body skeleton. At present, more than 300 cases have been reported at home and abroad, usually manifesting as spontaneous chronic osteolysis with no periosteal reaction at the lysis site and occult onset, often with fractures, scoliosis, chylothorax, etc. When waiting for medical treatment, the condition is serious, and the prognosis is poor. At present, there is no effective treatment. The main pathological manifestations of Gorham syndrome are the non-neoplastic abnormal proliferation of lymphatic vessels or blood vessels and osteolysis caused by osteoclast proliferation or increased activity. At present, there is no unified conclusion regarding Gorham syndrome's pathogenesis. This paper starts with the two most studied osteolysis methods at present, osteoclast osteolysis and osteolysis caused by vascular and lymphatic proliferation and summarizes the corresponding most possible molecular mechanisms in recent years to provide more ideas for Gorham syndrome treatment.
Collapse
Affiliation(s)
- Juqin Xiang
- Chongqing Medical University, Chongqing, China
| | - Weiyang Zhong
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Małys MS, Köller MC, Papp K, Aigner C, Dioso D, Mucher P, Schachner H, Bonelli M, Haslacher H, Rees AJ, Kain R. Small extracellular vesicles are released ex vivo from platelets into serum and from residual blood cells into stored plasma. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e88. [PMID: 38938276 PMCID: PMC11080719 DOI: 10.1002/jex2.88] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 03/29/2023] [Accepted: 04/18/2023] [Indexed: 06/29/2024]
Abstract
Small extracellular vesicles (sEV) purified from blood have great potential clinically as biomarkers for systemic disease; however interpretation is complicated by release of sEV ex vivo after blood taking. To quantify the problem and devise ways to minimise it, we characterised sEV in paired serum, plasma and platelet poor plasma (PPP) samples from healthy donors. Immunoblotting showed twofold greater abundance of CD9 in sEV fractions from fresh serum than from fresh plasma or PPP. MACSPlex confirmed this, and showed that proteins expressed on platelet sEV, either exclusively (CD41b, CD42a and CD62P) or more widely (HLA-ABC, CD24, CD29 and CD31) were also twofold more abundant; by contrast non-platelet proteins (including CD81) were no different. Storage of plasma (but not serum) increased abundance of platelet and selected leukocyte sEV proteins to at least that of serum, and this could be recapitulated by activating cells in fresh plasma by Ca2+, an effect abrogated in PPP. This suggests that a substantial proportion of sEV in serum and stored plasma were generated ex vivo, which is not the case for fresh plasma or PPP. Thus we provide strategies to minimise ex vivo sEV generation and criteria for identifying those that were present in vivo.
Collapse
Affiliation(s)
| | | | - Kristin Papp
- Department of PathologyMedical University ViennaViennaAustria
| | - Christof Aigner
- Department of PathologyMedical University ViennaViennaAustria
- Department of Medicine III, Division of Nephrology and DialysisMedical University ViennaViennaAustria
| | - Daffodil Dioso
- Department of Internal Medicine III, Division of RheumatologyMedical University of ViennaViennaAustria
| | - Patrick Mucher
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Helga Schachner
- Department of PathologyMedical University ViennaViennaAustria
| | - Michael Bonelli
- Department of Internal Medicine III, Division of RheumatologyMedical University of ViennaViennaAustria
| | - Helmuth Haslacher
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Andrew J. Rees
- Department of PathologyMedical University ViennaViennaAustria
| | - Renate Kain
- Department of PathologyMedical University ViennaViennaAustria
| |
Collapse
|
16
|
Zagorulya M, Yim L, Morgan DM, Edwards A, Torres-Mejia E, Momin N, McCreery CV, Zamora IL, Horton BL, Fox JG, Wittrup KD, Love JC, Spranger S. Tissue-specific abundance of interferon-gamma drives regulatory T cells to restrain DC1-mediated priming of cytotoxic T cells against lung cancer. Immunity 2023; 56:386-405.e10. [PMID: 36736322 PMCID: PMC10880816 DOI: 10.1016/j.immuni.2023.01.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/27/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023]
Abstract
Local environmental factors influence CD8+ T cell priming in lymph nodes (LNs). Here, we sought to understand how factors unique to the tumor-draining mediastinal LN (mLN) impact CD8+ T cell responses toward lung cancer. Type 1 conventional dendritic cells (DC1s) showed a mLN-specific failure to induce robust cytotoxic T cells responses. Using regulatory T (Treg) cell depletion strategies, we found that Treg cells suppressed DC1s in a spatially coordinated manner within tissue-specific microniches within the mLN. Treg cell suppression required MHC II-dependent contact between DC1s and Treg cells. Elevated levels of IFN-γ drove differentiation Treg cells into Th1-like effector Treg cells in the mLN. In patients with cancer, Treg cell Th1 polarization, but not CD8+/Treg cell ratios, correlated with poor responses to checkpoint blockade immunotherapy. Thus, IFN-γ in the mLN skews Treg cells to be Th1-like effector Treg cells, driving their close interaction with DC1s and subsequent suppression of cytotoxic T cell responses.
Collapse
Affiliation(s)
- Maria Zagorulya
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Leon Yim
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Duncan M Morgan
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Austin Edwards
- Biological Imaging Development CoLab, UCSF, San Francisco, CA 94143, USA
| | - Elen Torres-Mejia
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Noor Momin
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - Chloe V McCreery
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - Izabella L Zamora
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139, USA
| | - Brendan L Horton
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - James G Fox
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA; Division of Comparative Medicine, MIT, Cambridge, MA 02139, USA
| | - K Dane Wittrup
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA; Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Biology, MIT, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
17
|
Personalized risk predictor for acute cellular rejection in lung transplant using soluble CD31. Sci Rep 2022; 12:17628. [PMID: 36271122 PMCID: PMC9587244 DOI: 10.1038/s41598-022-21070-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 09/22/2022] [Indexed: 01/13/2023] Open
Abstract
We evaluated the contribution of artificial intelligence in predicting the risk of acute cellular rejection (ACR) using early plasma levels of soluble CD31 (sCD31) in combination with recipient haematosis, which was measured by the ratio of arterial oxygen partial pressure to fractional oxygen inspired (PaO2/FiO2) and respiratory SOFA (Sequential Organ Failure Assessment) within 3 days of lung transplantation (LTx). CD31 is expressed on endothelial cells, leukocytes and platelets and acts as a "peace-maker" at the blood/vessel interface. Upon nonspecific activation, CD31 can be cleaved, released, and detected in the plasma (sCD31). The study included 40 lung transplant recipients, seven (17.5%) of whom experienced ACR. We modelled the plasma levels of sCD31 as a nonlinear dependent variable of the PaO2/FiO2 and respiratory SOFA over time using multivariate and multimodal models. A deep convolutional network classified the time series models of each individual associated with the risk of ACR to each individual in the cohort.
Collapse
|
18
|
Boss AL, Damani T, Wickman TJ, Chamley LW, James JL, Brooks AES. Full spectrum flow cytometry reveals mesenchymal heterogeneity in first trimester placentae and phenotypic convergence in culture, providing insight into the origins of placental mesenchymal stromal cells. eLife 2022; 11:76622. [PMID: 35920626 PMCID: PMC9371602 DOI: 10.7554/elife.76622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 08/01/2022] [Indexed: 12/05/2022] Open
Abstract
Single-cell technologies (RNA-sequencing, flow cytometry) are critical tools to reveal how cell heterogeneity impacts developmental pathways. The placenta is a fetal exchange organ, containing a heterogeneous mix of mesenchymal cells (fibroblasts, myofibroblasts, perivascular, and progenitor cells). Placental mesenchymal stromal cells (pMSC) are also routinely isolated, for therapeutic and research purposes. However, our understanding of the diverse phenotypes of placental mesenchymal lineages, and their relationships remain unclear. We designed a 23-colour flow cytometry panel to assess mesenchymal heterogeneity in first-trimester human placentae. Four distinct mesenchymal subsets were identified; CD73+CD90+ mesenchymal cells, CD146+CD271+ perivascular cells, podoplanin+CD36+ stromal cells, and CD26+CD90+ myofibroblasts. CD73+CD90+ and podoplanin + CD36+ cells expressed markers consistent with cultured pMSCs, and were explored further. Despite their distinct ex-vivo phenotype, in culture CD73+CD90+ cells and podoplanin+CD36+ cells underwent phenotypic convergence, losing CD271 or CD36 expression respectively, and homogenously exhibiting a basic MSC phenotype (CD73+CD90+CD31-CD144-CD45-). However, some markers (CD26, CD146) were not impacted, or differentially impacted by culture in different populations. Comparisons of cultured phenotypes to pMSCs further suggested cultured pMSCs originate from podoplanin+CD36+ cells. This highlights the importance of detailed cell phenotyping to optimise therapeutic capacity, and ensure use of relevant cells in functional assays.
Collapse
Affiliation(s)
- Anna Leabourn Boss
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Tanvi Damani
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Tayla J Wickman
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Larry W Chamley
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Jo L James
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Anna E S Brooks
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
19
|
Zarobkiewicz MK, Morawska I, Kowalska W, Halczuk P, Roliński J, Bojarska-Junak AA. PECAM-1 Is Down-Regulated in γδT Cells during Remission, but Up-Regulated in Relapse of Multiple Sclerosis. J Clin Med 2022; 11:3210. [PMID: 35683597 PMCID: PMC9181399 DOI: 10.3390/jcm11113210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction. PECAM-1 and NKRP1A are both involved in the vascular transmigration of T lymphocytes. Vascular transmigration is a crucial process in multiple sclerosis pathogenesis. Methods and aim. The current paper presents an analysis of PECAM-1 and NKRP1A expression on γδ T cells. Expression of PECAM-1 and NKRP1A on subsets of γδ T cells was performed with flow cytometry. Results. Based on the flow cytometry data, PECAM1 was slightly differentially modulated on γδ T cells-it was up-regulated during relapse, but down-regulated during remission. Moreover, a significant downregulation of CD3 expression was noted on γδ T cells from MS patients, most notably during relapse. Conclusions. This may be a sign of the overall activation of γδ T cells in the course of multiple sclerosis.
Collapse
Affiliation(s)
- Michał K. Zarobkiewicz
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (W.K.); (J.R.)
| | - Izabela Morawska
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (W.K.); (J.R.)
| | - Wioleta Kowalska
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (W.K.); (J.R.)
| | - Paweł Halczuk
- Department of Neurology, Medical University of Lublin, 20-090 Lublin, Poland;
- Department of Histology and Embryology with Experimental Cytology Unit, Medical University of Lublin, 20-080 Lublin, Poland
| | - Jacek Roliński
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (W.K.); (J.R.)
| | - Agnieszka A. Bojarska-Junak
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (W.K.); (J.R.)
| |
Collapse
|
20
|
The Regulatory-T-Cell Memory Phenotype: What We Know. Cells 2022; 11:cells11101687. [PMID: 35626725 PMCID: PMC9139615 DOI: 10.3390/cells11101687] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 01/25/2023] Open
Abstract
In immunology, the discovery of regulatory T (Treg) cells was a major breakthrough. Treg cells play a key role in pregnancy maintenance, in the prevention of autoimmune responses, and in the control of all immune responses, including responses to self cells, cancer, infection, and a transplant. It is currently unclear whether Treg cells are capable of long-term memory of an encounter with an antigen. Although the term “immunological memory” usually means an enhanced ability to protect the body from reinfection, the memory of the suppressive activity of Treg cells helps to avoid the state of generalized immunosuppression that may result from the second activation of the immune system. In this review, we would like to discuss the concept of regulatory memory and in which tissues memory Treg cells can perform their functions.
Collapse
|
21
|
Kretschmer T, Turnwald EM, Janoschek R, Wohlfarth M, Handwerk M, Dötsch J, Hucklenbruch-Rother E, Appel S. Treatment of high fat diet-induced obese pregnant mice with IL-6 receptor antibody does not ameliorate placental function and fetal growth restriction. Am J Reprod Immunol 2022; 88:e13564. [PMID: 35535415 DOI: 10.1111/aji.13564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 11/29/2022] Open
Abstract
PROBLEM Pregnancy complications and adverse birth outcomes are in part fueled by the rise in obesity and its associated co-morbidities in western societies. Fetal healthy development and placental function are disturbed by an obese, inflammatory environment associated with cytokines, such as interleukin-6, causing inadequate supply of nutrients to the fetus and perinatal programming with severe health consequences. METHOD OF STUDY Mice received high fat diet (HFD) before and during gestation to induce obesity. We performed an IL-6 receptor antibody (MR16-1) treatment in pregnant obese mice at embryonic days E0.5, E7.5 and E14.5 to investigate whether this could ameliorate HFD-induced and obesity-associated placental dysfunction, evaluated by stereology and western blot, and improve offspring outcome at E15.5 in obese dams. RESULTS We observed fewer fetuses below the 10th percentile and placental vascularization was less aggravated following MR16-1 treatment of obese dams, showing slight improvements in labyrinth zone (Lz) vascularization. However, placental dysfunction and fetal growth restriction were still apparent in MR16-1 dams compared to lean control dams. Molecular analysis showed significantly elevated IL-6 level in placentas of MR16-1 treated dams. CONCLUSION Treatment with MR16-1 blocks IL-6 signaling in the placenta, but has only limited effects on preventing HFD-associated placental dysfunction and offspring outcomes in mice, suggesting further mechanisms in the deterioration of placental vascularization and fetal nutrient supply as a consequence of maternal obesity.
Collapse
Affiliation(s)
- Tobias Kretschmer
- Department of Paediatrics and Adolescent Medicine, University Hospital of Cologne, Cologne, Germany.,Department Environmental Immunology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Eva-Maria Turnwald
- Department of Paediatrics and Adolescent Medicine, University Hospital of Cologne, Cologne, Germany
| | - Ruth Janoschek
- Department of Paediatrics and Adolescent Medicine, University Hospital of Cologne, Cologne, Germany
| | - Maria Wohlfarth
- Department of Paediatrics and Adolescent Medicine, University Hospital of Cologne, Cologne, Germany
| | - Marion Handwerk
- Department of Paediatrics and Adolescent Medicine, University Hospital of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Paediatrics and Adolescent Medicine, University Hospital of Cologne, Cologne, Germany
| | - Eva Hucklenbruch-Rother
- Department of Paediatrics and Adolescent Medicine, University Hospital of Cologne, Cologne, Germany
| | - Sarah Appel
- Department of Paediatrics and Adolescent Medicine, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
22
|
Hsu M, Laaker C, Madrid A, Herbath M, Choi YH, Sandor M, Fabry Z. Neuroinflammation creates an immune regulatory niche at the meningeal lymphatic vasculature near the cribriform plate. Nat Immunol 2022; 23:581-593. [PMID: 35347285 PMCID: PMC8989656 DOI: 10.1038/s41590-022-01158-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 02/12/2022] [Indexed: 12/29/2022]
Abstract
Meningeal lymphatics near the cribriform plate undergo lymphangiogenesis during neuroinflammation to drain excess fluid. Here, we hypothesized that lymphangiogenic vessels may acquire an altered phenotype to regulate immunity. Using single-cell RNA sequencing of meningeal lymphatics near the cribriform plate from healthy and experimental autoimmune encephalomyelitis in the C57BL/6 model, we report that neuroinflammation induces the upregulation of genes involved in antigen presentation such as major histocompatibility complex class II, adhesion molecules including vascular cell adhesion protein 1 and immunoregulatory molecules such as programmed cell death 1 ligand 1, where many of these changes are mediated by interferon-γ. The inflamed lymphatics retain CD11c+ cells and CD4 T cells where they capture and present antigen, creating an immunoregulatory niche that represents an underappreciated interface in the regulation of neuroinflammation. We also found discontinuity of the arachnoid membrane near the cribriform plate, which provides unrestricted access to the cerebrospinal fluid. These findings highlight a previously unknown function of local meningeal lymphatics in regulating immunity that has only previously been characterized in draining lymph nodes.
Collapse
Affiliation(s)
- Martin Hsu
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Collin Laaker
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Andy Madrid
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Melinda Herbath
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Yun Hwa Choi
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
23
|
Russell AE, Liao Z, Tkach M, Tarwater PM, Ostrowski M, Théry C, Witwer KW. Cigarette smoke-induced extracellular vesicles from dendritic cells alter T-cell activation and HIV replication. Toxicol Lett 2022; 360:33-43. [PMID: 35181468 PMCID: PMC9014967 DOI: 10.1016/j.toxlet.2022.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/26/2022] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
Abstract
Despite decreased rates of tobacco smoking in many areas, cigarette smoking remains a major contributor to many health problems. Cigarette smoking can reduce immune system functioning while concurrently increasing inflammation. Dendritic cells in the lung exposed to cigarette smoke become stimulated and go on to activate T-cells. Extracellular vesicles (EVs) are nano-sized particles released by cells. They participate in intercellular communication by transferring functional proteins and nucleic acids to recipient cells and have been implicated in immune responses. For example, they can display MHC-peptide complexes to activate T-cells. In the current study, we sought to understand the role of cigarette smoke extract (CSE) on dendritic cell-derived EVs and their capacity to activate and differentiate T-cells. Primary human dendritic cells (iDCs) were exposed to CSE and EVs were separated and characterized. We exposed autologous primary CD4 + T-cells to iDC-EVs and observed T helper cell populations skewing towards Th1 and Th17 phenotypes. As HIV + individuals are disproportionately likely to be current smokers, we also examined the effects of iDC-EVs on acutely infected T-cells as well as on a cell model of HIV latency (ACH-2). We found that in most cases, iDC-CSE EVs tended to reduce p24 release from the acutely infected primary T-cells, albeit with great variability. We did not observe large effects of iDC-EVs or direct CSE exposure on p24 release from the ACH-2 cell line. Together, these data suggest that iDC-CSE EVs have the capacity to modulate the immune responses, in part by pushing T-cells towards Th1 and Th17 phenotypes.
Collapse
Affiliation(s)
- Ashley E Russell
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biology, School of Science, Penn State Erie, The Behrend College, Erie, PA, United States.
| | - Zhaohao Liao
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mercedes Tkach
- INSERM U932, Institut Curie Centre de Recherche, PSL Research University, Paris, France
| | - Patrick M Tarwater
- Department of Epidemiology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Matias Ostrowski
- Instituto INBIRS, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Clotilde Théry
- INSERM U932, Institut Curie Centre de Recherche, PSL Research University, Paris, France
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease.
| |
Collapse
|
24
|
Liu G, Han L, Lu Y, Wang C, Ma L, Zhang P, Liu C, Lu X, Ma Z. Clinicopathological study of the polypoidal lesions of polypoidal choroidal vasculopathy. Graefes Arch Clin Exp Ophthalmol 2022; 260:2369-2377. [PMID: 35147748 DOI: 10.1007/s00417-021-05525-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 12/05/2021] [Accepted: 12/14/2021] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To investigate the pathogenic features of the polypoidal lesions from the specimens of polypoidal choroidal vasculopathy extracted from human subjects. METHODS Seven specimens of polypoidal lesions extracted from five eyes of six patients (mean age, 60.16 ± 10.41 years) of polypoidal choroidal vasculopathy were examined. The polypoidal lesions were obtained by surgical excision. Thereafter, a histopathological analysis of the specimens was performed. RESULTS The polypoidal lesions were oval nodules located underneath the retinal pigment epithelium. A pathological study of the lesions revealed that Bruch's membrane schisis was observed in all specimens and they were all located in the Bruch's membrane. The Bruch's membrane schisis and serosanguineous materials constituted the main structure of the lesions in five of the seven specimens, with small vessels being observed in two specimens. One specimen was composed of two polypoidal lesions of different characteristics, and one specimen had a neovessel membrane complex with several polypoidal lesions. Inflammatory cells and blood vessels were observed in the polypoidal lesion of the specimen with neovessel membrane complex. CONCLUSION Polypoidal lesions of polypoidal choroidal vasculopathy are abnormalities of the Bruch's membrane. The lesions are characterized by the Bruch's membrane schisis, which is filled with serosanguineous materials. The lesions are progressive and may contain inflammatory cells and blood vessels.
Collapse
Affiliation(s)
- Guangfeng Liu
- Department of Ophthalmology, Life Science Park of Zhong Guancun, Peking University International Hospital, Chang Ping District, Life Park Road No. 1 , Beijing, 102206, China
| | - Liang Han
- Department of Ophthalmology, Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Eye Centre, Peking University Third Hospital, Beijing, 100191, China
| | - Yao Lu
- Department of Ophthalmology, Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Eye Centre, Peking University Third Hospital, Beijing, 100191, China
| | - Changguan Wang
- Department of Ophthalmology, Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Eye Centre, Peking University Third Hospital, Beijing, 100191, China
| | - Lie Ma
- Department of Ophthalmology, Life Science Park of Zhong Guancun, Peking University International Hospital, Chang Ping District, Life Park Road No. 1 , Beijing, 102206, China
| | - Pei Zhang
- Department of Ophthalmology, Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Eye Centre, Peking University Third Hospital, Beijing, 100191, China
| | - Cong Liu
- Department of Ophthalmology, Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Eye Centre, Peking University Third Hospital, Beijing, 100191, China
| | - Xinrong Lu
- Department of Ophthalmology, Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Eye Centre, Peking University Third Hospital, Beijing, 100191, China
| | - Zhizhong Ma
- Department of Ophthalmology, Life Science Park of Zhong Guancun, Peking University International Hospital, Chang Ping District, Life Park Road No. 1 , Beijing, 102206, China. .,Department of Ophthalmology, Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Eye Centre, Peking University Third Hospital, Beijing, 100191, China. .,Department of Ophthalmology, Peking University Third Hospital, Haidian District, No. 49, North Huayuan Road, Beijing, 100191, China.
| |
Collapse
|
25
|
Lia G, Di Vito C, Bruno S, Tapparo M, Brunello L, Santoro A, Mariotti J, Bramanti S, Zaghi E, Calvi M, Comba L, Fascì M, Giaccone L, Camussi G, Boyle EM, Castagna L, Evangelista A, Mavilio D, Bruno B. Extracellular Vesicles as Biomarkers of Acute Graft-vs.-Host Disease After Haploidentical Stem Cell Transplantation and Post-Transplant Cyclophosphamide. Front Immunol 2022; 12:816231. [PMID: 35145514 PMCID: PMC8821147 DOI: 10.3389/fimmu.2021.816231] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Even with high-dose post-transplant cyclophosphamide (PT-Cy) which was initially introduced for graft-versus-host disease (GvHD) prevention in the setting of HLA-haploidentical transplantation, both acute and chronic GvHDs remain a major clinical challenge. Despite improvements in the understanding of the pathogenesis of both acute and chronic GvHDs, reliable biomarkers that predict their onset have yet to be identified. We recently studied the potential correlation between extracellular vesicles (EVs) and the onset of acute (a)GvHD in transplant recipients from related and unrelated donors. In the present study, we further investigated the role of the expression profile of membrane proteins and their microRNA (miRNA) cargo (miRNA100, miRNA155, and miRNA194) in predicting the onset of aGvHD in haploidentical transplant recipients with PT-Cy. Thirty-two consecutive patients were included. We evaluated the expression profile of EVs, by flow cytometry, and their miRNA cargo, by real-time PCR, at baseline, prior, and at different time points following transplant. Using logistic regression and Cox proportional hazard models, a significant association between expression profiles of antigens such as CD146, CD31, CD140a, CD120a, CD26, CD144, and CD30 on EVs, and their miRNA cargo with the onset of aGvHD was observed. Moreover, we also investigated a potential correlation between EV expression profile and cargo with plasma biomarkers (e.g., ST2, sTNFR1, and REG3a) that had been associated with aGVHD previously. This analysis showed that the combination of CD146, sTNFR1, and miR100 or miR194 strongly correlated with the onset of aGvHD (AUROC >0.975). A large prospective multicenter study is currently in progress to validate our findings.
Collapse
Affiliation(s)
- Giuseppe Lia
- Division of Hematology, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Clara Di Vito
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Stefania Bruno
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Marta Tapparo
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Lucia Brunello
- Division of Hematology, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Armando Santoro
- Bone Marrow Transplant Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Jacopo Mariotti
- Bone Marrow Transplant Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Stefania Bramanti
- Bone Marrow Transplant Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Elisa Zaghi
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Michela Calvi
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Lorenzo Comba
- Division of Hematology, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Martina Fascì
- Division of Hematology, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Luisa Giaccone
- Division of Hematology, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Eileen M. Boyle
- Division of Hematology and Medical Oncology, New York University Grossman School of Medicine, Perlmutter Cancer Center, New York University (NYU) Langone Health, New York, NY, United States
| | - Luca Castagna
- Bone Marrow Transplant Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Andrea Evangelista
- Clinical Epidemiology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Benedetto Bruno
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Division of Hematology and Medical Oncology, New York University Grossman School of Medicine, Perlmutter Cancer Center, New York University (NYU) Langone Health, New York, NY, United States
- *Correspondence: Benedetto Bruno,
| |
Collapse
|
26
|
Yang H, Yang Q, Wang Y, Zheng L. Inhibition of hypoxia-inducible factor-1 by Salidroside in an in vitro model of choroidal neovascularization. Cutan Ocul Toxicol 2021; 41:203-209. [PMID: 34428999 DOI: 10.1080/15569527.2021.1973023] [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: 11/03/2022]
Abstract
PURPOSE As a characteristic of age-related macular degeneration (AMD), choroidal neovascularization (CNV) causes severe vision loss. The current treatment has limited efficacy. This study was to investigate effects of Salidroside against CNV and explore its underlying mechanisms. METHODS RF/6A cells were treated with 200 mM cobalt chloride (CoCl2) for 6 hr to mimic hypoxic condition. Cells were then treated with Salidroside at 10, 30 and 100 µM for 24 hr. Cells treated with DMSO were used as negative control. The cell proliferation was assessed using 3-(4,5-dimethylthiazol)-2,5-diphenyltetrazolium-bromid assay. The tube formation was investigated on Matrigel. The cell migration was measured by a Transwell assay. RT-qPCR was used to detect the gene expression. Immuohistochemistry and western blot were used to detect the expression of proteins. RESULTS Salidroside significantly inhibited the cell migration and tube formation activity of RF/6A cells under hypoxia. Moreover, Salidroside reduced the expression levels of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1 (HIF-1) in RF/6A cells. CONCLUSIONS Our data suggested that Salidroside could be a potential novel therapeutic agent against CNV.
Collapse
Affiliation(s)
- Haitao Yang
- Department of Neurosurgery, The second Fuzhou Hospital Affiliated to Xiamen University, Fuzhou 350007, People's Republic of China
| | - Qingwu Yang
- Department of Neurosurgery, The second Fuzhou Hospital Affiliated to Xiamen University, Fuzhou 350007, People's Republic of China
| | - Yunfeng Wang
- Department of Neurosurgery, The second Fuzhou Hospital Affiliated to Xiamen University, Fuzhou 350007, People's Republic of China
| | - Linfei Zheng
- Department of Neurosurgery, The second Fuzhou Hospital Affiliated to Xiamen University, Fuzhou 350007, People's Republic of China
| |
Collapse
|
27
|
Alizadehgiashi M, Nemr CR, Chekini M, Pinto Ramos D, Mittal N, Ahmed SU, Khuu N, Kelley SO, Kumacheva E. Multifunctional 3D-Printed Wound Dressings. ACS NANO 2021; 15:12375-12387. [PMID: 34133121 DOI: 10.1021/acsnano.1c04499] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Personalized wound dressings provide enhanced healing for different wound types; however multicomponent wound dressings with discretely controllable delivery of different biologically active agents are yet to be developed. Here we report 3D-printed multicomponent biocomposite hydrogel wound dressings that have been selectively loaded with small molecules, metal nanoparticles, and proteins for independently controlled release at the wound site. Hydrogel wound dressings carrying antibacterial silver nanoparticles and vascular endothelial growth factor with predetermined release profiles were utilized to study the physiological response of the wound in a mouse model. Compared to controls, the application of dressings resulted in improvement in granulation tissue formation and differential levels of vascular density, dependent on the release profile of the growth factor. Our study demonstrates the versatility of the 3D-printed hydrogel dressings that can yield varied physiological responses in vivo and can further be adapted for personalized treatment of various wound types.
Collapse
Affiliation(s)
- Moien Alizadehgiashi
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
| | - Carine R Nemr
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
| | - Mahshid Chekini
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
| | - Daniel Pinto Ramos
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
| | - Nitesh Mittal
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
- Linné FLOW Centre, KTH Mechanics, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
- Wallenberg Wood Science Center, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
| | - Sharif U Ahmed
- Department of Pharmaceutical Sciences, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Nancy Khuu
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
| | - Shana O Kelley
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
- Department of Pharmaceutical Sciences, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
- The Institute of Biomaterials and Biomedical Engineering, University of Toronto, 4 Taddle Creek Road, Toronto, Ontario M5S 3G9, Canada
| | - Eugenia Kumacheva
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
- The Institute of Biomaterials and Biomedical Engineering, University of Toronto, 4 Taddle Creek Road, Toronto, Ontario M5S 3G9, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| |
Collapse
|
28
|
Proteomics and metabonomics analyses of Covid-19 complications in patients with pulmonary fibrosis. Sci Rep 2021; 11:14601. [PMID: 34272434 PMCID: PMC8285535 DOI: 10.1038/s41598-021-94256-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 06/29/2021] [Indexed: 12/23/2022] Open
Abstract
Pulmonary fibrosis is a devastating disease, and the pathogenesis of this disease is not completely clear. Here, the medical records of 85 Covid-19 cases were collected, among which fibrosis and progression of fibrosis were analyzed in detail. Next, data independent acquisition (DIA) quantification proteomics and untargeted metabolomics were used to screen disease-related signaling pathways through clustering and enrichment analysis of the differential expression of proteins and metabolites. The main imaging features were lesions located in the bilateral lower lobes and involvement in five lobes. The closed association pathways were FcγR-mediated phagocytosis, PPAR signaling, TRP-inflammatory pathways, and the urea cycle. Our results provide evidence for the detection of serum biomarkers and targeted therapy in patients with Covid-19.
Collapse
|
29
|
Lansky A, Chun H, Pietras C, Hussain Y. Refining drug-eluting stent technologies: from engineering to basic science. Eur Heart J 2021; 42:1770-1772. [PMID: 33624813 DOI: 10.1093/eurheartj/ehab091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Alexandra Lansky
- Yale School of Medicine, Department of Cardiology New Haven, CT, USA.,Yale Cardiovascular Research Group, New Haven, CT, USA
| | - Hyung Chun
- Yale School of Medicine, Department of Cardiology New Haven, CT, USA
| | - Cody Pietras
- Yale School of Medicine, Department of Cardiology New Haven, CT, USA
| | - Yasin Hussain
- Yale School of Medicine, Department of Cardiology New Haven, CT, USA
| |
Collapse
|
30
|
Gorelova A, Berman M, Al Ghouleh I. Endothelial-to-Mesenchymal Transition in Pulmonary Arterial Hypertension. Antioxid Redox Signal 2021; 34:891-914. [PMID: 32746619 PMCID: PMC8035923 DOI: 10.1089/ars.2020.8169] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/14/2022]
Abstract
Endothelial-to-mesenchymal transition (EndMT) is a process that encompasses extensive transcriptional reprogramming of activated endothelial cells leading to a shift toward mesenchymal cellular phenotypes and functional responses. Initially observed in the context of embryonic development, in the last few decades EndMT is increasingly recognized as a process that contributes to a variety of pathologies in the adult organism. Within the settings of cardiovascular biology, EndMT plays a role in various diseases, including atherosclerosis, heart valvular disease, cardiac fibrosis, and myocardial infarction. EndMT is also being progressively implicated in development and progression of pulmonary hypertension (PH) and pulmonary arterial hypertension (PAH). This review covers the current knowledge about EndMT in PH and PAH, and provides comprehensive overview of seminal discoveries. Topics covered include evidence linking EndMT to factors associated with PAH development, including hypoxia responses, inflammation, dysregulation of bone-morphogenetic protein receptor 2 (BMPR2), and redox signaling. This review amalgamates these discoveries into potential insights for the identification of underlying mechanisms driving EndMT in PH and PAH, and discusses future directions for EndMT-based therapeutic strategies in disease management.
Collapse
Affiliation(s)
- Anastasia Gorelova
- Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mariah Berman
- Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Imad Al Ghouleh
- Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
31
|
Briceño O, Peralta-Prado A, Garrido-Rodríguez D, Romero-Mora K, Chávez-Torres M, de la Barrera CA, Reyes-Terán G, Ávila-Ríos S. Characterization of CD31 expression in CD4+ and CD8+T cell subpopulations in chronic untreated HIV infection. Immunol Lett 2021; 235:22-31. [PMID: 33852965 DOI: 10.1016/j.imlet.2021.04.004] [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: 09/23/2020] [Revised: 03/09/2021] [Accepted: 04/07/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND The platelet endothelial cell adhesion molecule-1 (PECAM-1) or CD31 has been involved in regulation of T-cell tolerance, activation, survival and homing in mice cells. However, there is limited knowledge about the expression pattern and role of this molecule in human T cells, particularly in conditions of chronic immune activation. OBJECTIVES We explored CD31 expression in T cell differentiation subsets of individuals with untreated HIV infection and in non-HIV-infected controls. We also assessed phenotypic differences between CD31+ and CD31- subsets in memory and terminally differentiated (TEMRA) CD4+ and CD8 + T cells. METHODS Forty-one individuals with untreated HIV infection and 34 non-HIV-infected controls were included in the study. We compared the expression of CD31 in CD4+ and CD8 + T cells across stages of differentiation in the two study groups by flow cytometry. We also analyzed the expression of CD57 (a marker of senescence), Ki67 (a marker of cycling cells), PD-1 (a marker of exhaustion), and CD38/HLA-DR (a marker of immune activation) on memory and TEMRA CD31+ and CD31- T cells. RESULTS CD31 expression was significantly higher in CD8 + T cells than in CD4 + T cells, measured as frequency, absolute numbers and median fluorescence intensity (MFI), in both study groups (p < 0.0001 in all cases). Intermediate differentiation subsets of CD4+ and CD8 + T cells expressed higher levels of CD31 in the context of HIV infection (p < 0.001 in all cases). CD31 expression frequency decreased with cellular differentiation of CD4+ and CD8 + T cells in both groups, but this decrease was steeper in individuals without HIV infection (CD4+: p < 0.001 and CD8+: p < 0.0001). As expected, memory and TEMRA CD4+ and CD8 + T cells expressed significantly higher levels of CD57, PD-1, Ki67 and CD38/HLA-DR in HIV-infected compared to non-HIV-infected individuals (p < 0.01 in all cases). CD31 expression was associated with lower activation of memory (but not TEMRA) CD4 + T cells in non-HIV-infected persons, an effect not observed in the HIV-infected group. CD31 expression on memory CD8 + T cells of HIV-infected individuals was associated higher levels of PD-1 (p = 0.0019) and CD38/HLADR (p = 0.0345), and higher PD-1 expression on CD8 + TEMRA (p = 0.0024), an effect not observed in non-HIV-infected individuals. CONCLUSION In the context of HIV-associated chronic immune activation, specifically on memory CD8 + T cells, CD31 expression was associated with higher PD-1 and CD38/HLA-DR co-expression, suggesting that CD31 expression may result from an insufficient attempt to contain T cell exhaustion and activation. CD31-targeted therapies may contribute to modulate these cellular responses.
Collapse
Affiliation(s)
- Olivia Briceño
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico.
| | - Amy Peralta-Prado
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| | - Daniela Garrido-Rodríguez
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| | - Karla Romero-Mora
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| | - Monserrat Chávez-Torres
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| | - Claudia-Alvarado de la Barrera
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| | - Gustavo Reyes-Terán
- Coordinating Commission of the Mexican National Institutes of Health, Mexico City, Mexico
| | - Santiago Ávila-Ríos
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| |
Collapse
|
32
|
Aleksonienė R, Besusparis J, Gruslys V, Jurgauskienė L, Laurinavičienė A, Laurinavičius A, Malickaitė R, Norkūnienė J, Zablockis R, Žurauskas E, Danila E. CD31 +, CD38 +, CD44 +, and CD103 + lymphocytes in peripheral blood, bronchoalveolar lavage fluid and lung biopsy tissue in sarcoid patients and controls. J Thorac Dis 2021; 13:2300-2318. [PMID: 34012580 PMCID: PMC8107533 DOI: 10.21037/jtd-20-2396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background The mechanisms driving the transition from inflammation to fibrosis in sarcoidosis patients are poorly understood; prognostic features are lacking. Immune cell profiling may provide insights into pathogenesis and prognostic factors of the disease. This study aimed to establish associations in simultaneous of lymphocyte subset profiles in the blood, bronchoalveolar lavage fluid (BALF), and lung biopsy tissue in the patients with newly diagnosed sarcoidosis. Methods A total of 71 sarcoid patients (SPs) and 20 healthy controls (HCs) were enrolled into the study. CD31, CD38, CD44, CD103 positive T lymphocytes in blood and BALF were analysed. Additionally, the densities of CD4, CD8, CD38, CD44, CD103 positive cells in lung tissue biopsies were estimated by digital image analysis. Results Main findings: (I) increase of percentage of CD3+CD4+CD38+ in BALF and blood, and increase of percentage of CD3+CD4+CD44+ in BALF in Löfgren syndrome patients comparing with patients without Löfgren syndrome, (II) increase of percentage of CD3+CD4+103+ in BALF and in blood in patients without Löfgren syndrome (comparing with Löfgren syndrome patients) and increase of percentage of CD3+CD4+103+ in BALF and in blood in more advanced sarcoidosis stage. (III) Increasing percentage of BALF CD3+CD4+CD31+ in sarcoidosis patients when comparing with controls independently of presence of Löfgren syndrome, smoking status or stage of sarcoidosis. Several significant correlations were found. Conclusions Lymphocyte subpopulations in blood, BALF, and lung tissue were substantially different in SPs at the time of diagnosis compared to HCs. CD3+CD4+CD31+ in BALF might be a potential supporting marker for the diagnosis of sarcoidosis. CD3+CD4+CD38+ in BALF and blood and CD3+CD4+CD44+ in BALF may be markers of the acute immune response in sarcoidosis patients. CD4+CD103+ T-cells in BALF and in blood are markers of the persistent immune response in sarcoidosis patients and are potential prognostic features of the chronic course of this disease.
Collapse
Affiliation(s)
- Regina Aleksonienė
- Clinic of Chest Diseases, Immunology and Allergology of Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Justinas Besusparis
- National Center of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania.,Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Vygantas Gruslys
- Clinic of Chest Diseases, Immunology and Allergology of Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | | | - Aida Laurinavičienė
- National Center of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania.,Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Arvydas Laurinavičius
- National Center of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania.,Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Jolita Norkūnienė
- Department of Mathematical Statistics, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Rolandas Zablockis
- Clinic of Chest Diseases, Immunology and Allergology of Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Edvardas Žurauskas
- National Center of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania.,Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Edvardas Danila
- Clinic of Chest Diseases, Immunology and Allergology of Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| |
Collapse
|
33
|
Falleni M, Tosi D, Savi F, Chiumello D, Bulfamante G. Endothelial-Mesenchymal Transition in COVID-19 lung lesions. Pathol Res Pract 2021; 221:153419. [PMID: 33857718 PMCID: PMC7997691 DOI: 10.1016/j.prp.2021.153419] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 01/13/2023]
Abstract
Sars-Cov-2 infection is still a healthcare emergency and acute respiratory distress failure with Diffuse Alveolar Damage (DAD) features is the main causes of patients’ death. Pathogenic mechanisms of the disease are not clear yet, but new insights are necessary to improve therapeutic management, to prevent fatal irreversible multi-organ damage and to adequately follow up those patients who survive. Here we investigated, by histochemistry and immunohistochemistry, a wide number of mapped lung specimens taken from whole body autopsies of 7 patients dead of COVID-19 disease. Our data confirm morphological data of other authors, and enlarge recent reports of the literature suggesting that Endothelial–Mesenchymal Transition might be central to COVID-19 lung fibrosing lesions. Furthermore, based upon recent acquisition of new roles in immunity and vascular pathology of the CD31 molecule, we hypothesize that this molecule might be important in the development and treatment of COVID-19 pulmonary lesions. These preliminary findings need further investigations to shed light on the complexity of Sars-Cov-2 disease.
Collapse
Affiliation(s)
- Monica Falleni
- Unità di Anatomia Patologica, Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142, Milan, Italy; Struttura Complessa Dipartimentale di Anatomia Patologica e Genetica Medica, ASST Santi Paolo e Carlo, 20142, Milan, Italy.
| | - Delfina Tosi
- Unità di Anatomia Patologica, Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142, Milan, Italy
| | - Federica Savi
- Struttura Complessa Dipartimentale di Anatomia Patologica e Genetica Medica, ASST Santi Paolo e Carlo, 20142, Milan, Italy
| | - Davide Chiumello
- Anestesia e Rianimazione, Ospedale, Centro ricerca coordinata di insufficienza respiratoria, Dipartimento di Scienze della Salute, Università degli Studi di Milano ASST Santi Paolo e Carlo, 20142, Milan, Italy
| | - Gaetano Bulfamante
- Unità di Anatomia Patologica, Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142, Milan, Italy; Struttura Complessa Dipartimentale di Anatomia Patologica e Genetica Medica, ASST Santi Paolo e Carlo, 20142, Milan, Italy
| |
Collapse
|
34
|
Sluiter TJ, van Buul JD, Huveneers S, Quax PHA, de Vries MR. Endothelial Barrier Function and Leukocyte Transmigration in Atherosclerosis. Biomedicines 2021; 9:328. [PMID: 33804952 PMCID: PMC8063931 DOI: 10.3390/biomedicines9040328] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/24/2022] Open
Abstract
The vascular endothelium is a highly specialized barrier that controls passage of fluids and migration of cells from the lumen into the vessel wall. Endothelial cells assist leukocytes to extravasate and despite the variety in the specific mechanisms utilized by different leukocytes to cross different vascular beds, there is a general principle of capture, rolling, slow rolling, arrest, crawling, and ultimately diapedesis via a paracellular or transcellular route. In atherosclerosis, the barrier function of the endothelium is impaired leading to uncontrolled leukocyte extravasation and vascular leakage. This is also observed in the neovessels that grow into the atherosclerotic plaque leading to intraplaque hemorrhage and plaque destabilization. This review focuses on the vascular endothelial barrier function and the interaction between endothelial cells and leukocytes during transmigration. We will discuss the role of endothelial dysfunction, transendothelial migration of leukocytes and plaque angiogenesis in atherosclerosis.
Collapse
Affiliation(s)
- Thijs J. Sluiter
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jaap D. van Buul
- Sanquin Research and Landsteiner Laboratory, Leeuwenhoek Centre for Advanced Microscopy, Swammerdam Institute for Life Sciences, University of Amsterdam, 1066 CX Amsterdam, The Netherlands;
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Paul H. A. Quax
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Margreet R. de Vries
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
35
|
Cao S, Wang Y, Li J, Ling X, Zhang Y, Zhou Y, Zhong H. Prognostic Implication of the Expression Level of PECAM-1 in Non-small Cell Lung Cancer. Front Oncol 2021; 11:587744. [PMID: 33828969 PMCID: PMC8019905 DOI: 10.3389/fonc.2021.587744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 02/08/2021] [Indexed: 01/14/2023] Open
Abstract
Background: Lung cancer is a malignant disease that threatens human health. Hence, it is crucial to identify effective prognostic factors and treatment targets. Single-cell RNA sequencing can quantify the expression profiles of transcripts in individual cells. Methods: GSE117570 profiles were downloaded from the Gene Expression Omnibus database. Key ligand-receptor genes in the tumor and the normal groups were screened to identify integrated differentially expressed genes (DEGs) from the GSE118370 and The Cancer Genome Atlas Lung Adenocarcinoma databases. DEGs associated with more ligand-receptor pairs were selected as candidate DEGs for Gene Ontology (GO) functional annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and survival analysis. In addition, we conducted validation immunohistochemical experiments on postoperative specimens of 30 patients with lung cancer. Results: A total of 18 candidate DEGs were identified from the tumor and the normal groups. The analysis of the GO biological process revealed that these DEGs were mainly enriched in wound healing, in response to wounding, cell migration, cell motility, and regulation of cell motility, while the KEGG pathway analysis found that these DEGs were mainly enriched in proteoglycans in cancer, bladder cancer, malaria, tyrosine kinase inhibitor resistance in Epidermal Growth Factor Receptor (EGFR), and the ERBB signaling pathway. Survival analysis showed that a high, rather than a low, expression of platelet endothelial cell adhesion molecule-1 (PECAM-1) was associated with improved survival. Similarly, in postoperative patients with lung cancer, we found that the overall survival of the PECAM-1 high-expression group shows a better trend than the PECAM-1 low-expression group (p = 0.172). Conclusions: The candidate DEGs identified in this study may play some important roles in the occurrence and development of lung cancer, especially PECAM-1, which may present potential prognostic biomarkers for the outcome.
Collapse
Affiliation(s)
| | | | | | | | | | - Yan Zhou
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Hua Zhong
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
36
|
Bavle RM, Paremala K, Venugopal R, Rudramuni AS, Khan N, Hosthor SS. Grading of Oral Leukoplakia: Can It be Improvised Using Immunohistochemical Markers p63 and CD31. Contemp Clin Dent 2021; 12:37-43. [PMID: 33967536 PMCID: PMC8092094 DOI: 10.4103/ccd.ccd_493_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 05/17/2020] [Accepted: 07/03/2020] [Indexed: 11/04/2022] Open
Abstract
Introduction Oral squamous cell carcinoma is usually preceded by potentially malignant disorders (PMDs), the most common being oral leukoplakia. A conservative management protocol is followed for milder dysplastic cases, while severe dysplastic lesions are surgically excised. Several classification systems have been developed based mainly on histopathological features with a lot of inter-observer variations. The present study was done to assess the use of immunohistochemical (IHC) markers in grading leukoplakic lesions in addition to histopathological grading. Aims and Objectives To grade leukoplakia using different grading systems and assess if IHC markers can aid in categorizing leukoplakia. Materials and Methods Thirty-five cases of leukoplakia were graded using Ljubljana, 2005 World Health Organization (WHO), and Binary System followed by IHC staining with p63 and CD31. Results and Statistics Variation was noted in 12 cases while using WHO, 11 using Ljubljana, and 7 using Binary System and was significant on Cohen-Kappa statistics, with the least significant variation noted on Binary System. p63 staining assisted to group doubtful cases and even identify variation in cases graded positively on histopathology. In total, 17 cases stained one-third (mild/low), while 15 cases stained one-half or more (higher grade) epithelial thickness. A weak correlation was observed between all grading systems and p63 on Kendall's Tau-b analysis and the weak correlation was significant for the WHO and binary grading systems. Analyzing p63 and CD31 using Kruskal-Wallis test, an increase in mean vessel density (MVD) was noted for mild/moderate cases but decreased for severe cases. Conclusion Addressing histological categorization of PMDs assisted by IHC markers to understand the biological behavior of the tissues is currently essential with studies on more markers to assist in the management protocol.
Collapse
Affiliation(s)
- Radhika Manoj Bavle
- Department of Oral and Maxillofacial Pathology, Krishnadevaraya College of Dental College and Hospital, Sir MVIT Campus, Bengaluru, Karnataka, India
| | - K Paremala
- Government Dental College and Hospital, Hyderabad, Telangana, India
| | - Reshma Venugopal
- Department of Oral and Maxillofacial Pathology, Krishnadevaraya College of Dental College and Hospital, Sir MVIT Campus, Bengaluru, Karnataka, India
| | - Amulya S Rudramuni
- Department of Oral and Maxillofacial Pathology, Krishnadevaraya College of Dental College and Hospital, Sir MVIT Campus, Bengaluru, Karnataka, India
| | - Nawal Khan
- Sher-i-Kashmir Institute of Medical Sciences, Community Medicine (Dental Unit), Srinagar, Jammu and Kashmir, India
| | - Sreenitha S Hosthor
- Department of Oral and Maxillofacial Pathology, Krishnadevaraya College of Dental College and Hospital, Sir MVIT Campus, Bengaluru, Karnataka, India
| |
Collapse
|
37
|
Núñez-Sánchez E, Flores-Espinosa MDP, Mancilla-Herrera I, González L, Cisneros J, Olmos-Ortiz A, Quesada-Reyna B, Granados-Cepeda M, Zaga-Clavellina V. Prolactin modifies the in vitro LPS-induced chemotactic capabilities in human fetal membranes at the term of gestation. Am J Reprod Immunol 2021; 86:e13413. [PMID: 33660388 PMCID: PMC8365646 DOI: 10.1111/aji.13413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/03/2021] [Accepted: 03/01/2021] [Indexed: 12/05/2022] Open
Abstract
Problem Immune responses of fetal membranes involve the production of chemoattractant mediators causing infiltration of maternal and fetal leukocytes, intrauterine inflammation and potentially the disruption of maternal‐fetal tolerance. Prolactin (PRL) has deep immunoregulatory effects in the fetal‐maternal interface. We aimed to test the in vitro PRL effect upon chemotactic capacities of human fetal membranes. Method of Study Fetal membranes and umbilical cord blood were collected from healthy non‐laboring caesarean deliveries at term. Fetal membranes were cultured in Transwell® frames to mimic the barrier function between choriodecidual and amniotic sides. Tissues were treated with PRL, Lipopolysaccharide (LPS), or both simultaneously. Then, RANTES, MCP‐1, MIP‐1α, IP‐10, and PECAM‐1 were quantified in a conditioned medium by choriodecidual or amniotic sides. The chemotaxis of subsets of migrating mononuclear cells from umbilical cord blood was evaluated in a Boyden Chamber in response to the conditioned medium by both sides. Results Lipopolysaccharide stimulates the production of RANTES, MCP‐1, MIP‐1α, and PECAM‐1 in choriodecidua, while MIP‐1α and PECAM‐1 only increase in amnion. PRL decrease RANTES, MCP‐1, and MIP‐1 only in choriodecidua, but PECAM‐1 was decreased mainly in amnion. The leukocyte migration was regulated significantly in response to the conditioned medium by the amnion, increase in the conditioned medium after LPS treatment, contrary with, the leukocyte migration decreased in a significant manner in response to conditioned medium after PRL and LPS‐PRL co‐treatment. Finally, T cells were the most responsive subset of cells. Conclusions Prolactin modified in a tissue‐specific manner the chemotactic factor and the leukocyte migration differentially in fetal membranes.
Collapse
Affiliation(s)
- Estefanía Núñez-Sánchez
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Ciudad de México, México
| | - María Del Pilar Flores-Espinosa
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Ciudad de México, México
| | - Ismael Mancilla-Herrera
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Ciudad de México, México
| | - Leticia González
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Ciudad de México, México
| | - José Cisneros
- Laboratorio de Biopatología Pulmonar, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Ciudad de México, México
| | - Andrea Olmos-Ortiz
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Ciudad de México, México
| | - Braulio Quesada-Reyna
- División de Gineco-Obstetricia, UMAE Hospital de Gineco-Obstetricia No. 4 "Luis Castelazo Ayala" IMSS, Ciudad de México, México
| | - Martha Granados-Cepeda
- Departamento de Neonatología, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Ciudad de México, México
| | - Veronica Zaga-Clavellina
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes", Ciudad de México, México
| |
Collapse
|
38
|
Ning W, Acharya A, Sun Z, Ogbuehi AC, Li C, Hua S, Ou Q, Zeng M, Liu X, Deng Y, Haak R, Ziebolz D, Schmalz G, Pelekos G, Wang Y, Hu X. Deep Learning Reveals Key Immunosuppression Genes and Distinct Immunotypes in Periodontitis. Front Genet 2021; 12:648329. [PMID: 33777111 PMCID: PMC7994531 DOI: 10.3389/fgene.2021.648329] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/18/2021] [Indexed: 02/02/2023] Open
Abstract
Background Periodontitis is a chronic immuno-inflammatory disease characterized by inflammatory destruction of tooth-supporting tissues. Its pathogenesis involves a dysregulated local host immune response that is ineffective in combating microbial challenges. An integrated investigation of genes involved in mediating immune response suppression in periodontitis, based on multiple studies, can reveal genes pivotal to periodontitis pathogenesis. Here, we aimed to apply a deep learning (DL)-based autoencoder (AE) for predicting immunosuppression genes involved in periodontitis by integrating multiples omics datasets. Methods Two periodontitis-related GEO transcriptomic datasets (GSE16134 and GSE10334) and immunosuppression genes identified from DisGeNET and HisgAtlas were included. Immunosuppression genes related to periodontitis in GSE16134 were used as input to build an AE, to identify the top disease-representative immunosuppression gene features. Using K-means clustering and ANOVA, immune subtype labels were assigned to disease samples and a support vector machine (SVM) classifier was constructed. This classifier was applied to a validation set (Immunosuppression genes related to periodontitis in GSE10334) for predicting sample labels, evaluating the accuracy of the AE. In addition, differentially expressed genes (DEGs), signaling pathways, and transcription factors (TFs) involved in immunosuppression and periodontitis were determined with an array of bioinformatics analysis. Shared DEGs common to DEGs differentiating periodontitis from controls and those differentiating the immune subtypes were considered as the key immunosuppression genes in periodontitis. Results We produced representative molecular features and identified two immune subtypes in periodontitis using an AE. Two subtypes were also predicted in the validation set with the SVM classifier. Three “master” immunosuppression genes, PECAM1, FCGR3A, and FOS were identified as candidates pivotal to immunosuppressive mechanisms in periodontitis. Six transcription factors, NFKB1, FOS, JUN, HIF1A, STAT5B, and STAT4, were identified as central to the TFs-DEGs interaction network. The two immune subtypes were distinct in terms of their regulating pathways. Conclusion This study applied a DL-based AE for the first time to identify immune subtypes of periodontitis and pivotal immunosuppression genes that discriminated periodontitis from the healthy. Key signaling pathways and TF-target DEGs that putatively mediate immune suppression in periodontitis were identified. PECAM1, FCGR3A, and FOS emerged as high-value biomarkers and candidate therapeutic targets for periodontitis.
Collapse
Affiliation(s)
- Wanchen Ning
- Department of Conservative Dentistry and Periodontology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Aneesha Acharya
- Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, India.,Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Zhengyang Sun
- Faculty of Mechanical Engineering, Chemnitz University of Technology, Chemnitz, Germany
| | | | - Cong Li
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shiting Hua
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qianhua Ou
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Muhui Zeng
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiangqiong Liu
- Laboratory of Cell and Molecular Biology, Beijing Tibetan Hospital, China Tibetology Research Center, Beijing, China
| | - Yupei Deng
- Laboratory of Cell and Molecular Biology, Beijing Tibetan Hospital, China Tibetology Research Center, Beijing, China
| | - Rainer Haak
- Department of Cariology, Endodontology and Periodontology, University of Leipzig, Leipzig, Germany
| | - Dirk Ziebolz
- Department of Cariology, Endodontology and Periodontology, University of Leipzig, Leipzig, Germany
| | - Gerhard Schmalz
- Department of Cariology, Endodontology and Periodontology, University of Leipzig, Leipzig, Germany
| | - George Pelekos
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Yang Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, School of Life Sciences, Hubei University, Wuhan, China
| | - Xianda Hu
- Laboratory of Cell and Molecular Biology, Beijing Tibetan Hospital, China Tibetology Research Center, Beijing, China
| |
Collapse
|
39
|
Cortese J, Rasser C, Even G, Bardet SM, Choqueux C, Mesnier J, Perrin ML, Janot K, Caroff J, Nicoletti A, Michel JB, Spelle L, Caligiuri G, Rouchaud A. CD31 Mimetic Coating Enhances Flow Diverting Stent Integration into the Arterial Wall Promoting Aneurysm Healing. Stroke 2021; 52:677-686. [PMID: 33412905 DOI: 10.1161/strokeaha.120.030624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Beyond aneurysmal occlusion, metallic flow diverters (FDs) can induce an adverse endovascular reaction due to the foreignness of metal devices, hampering FD endothelialization across the aneurysm neck, and arterial healing of intracranial aneurysms. Here, we evaluated the potential benefits of an FD coating mimicking CD31, a coreceptor critically involved in endothelial function and endovascular homeostasis, on the endothelialization of FDs implanted in vivo. METHODS Nitinol FD (Silk Vista Baby) and flat disks were dip-coated with a CD31-mimetic peptide via an intermediate layer of polydopamine. Disks were used to assess the reaction of endothelial cells and blood elements in vitro. An aneurysm rabbit model was used to compare in vivo effects on the arterial wall of CD31-mimetic-coated (CD31-mimetic, n=6), polydopamine-coated (polydopamine, n=6), and uncoated FDs (bare, n=5) at 4 weeks post-FD implantation. In addition, long-term safety was assessed at 12 weeks. RESULTS In vitro, CD31-mimetic coated disks displayed reduced adhesion of blood elements while favoring endothelial cell attachment and confluence, compared to bare and polydopamine disks. Strikingly, in vivo, the neoarterial wall formed over the CD31-mimetic-FD struts at the aneurysm neck was characteristic of an arterial tunica media, with continuous differentiated endothelium covering a significantly thicker layer of collagen and smooth muscle cells as compared to the controls. The rates of angiographic complete occlusion and covered branch arterial patency were similar in all 3 groups. CONCLUSIONS CD31-mimetic coating favors the colonization of metallic endovascular devices with endothelial cells displaying a physiological phenotype while preventing the adhesion of platelets and leukocytes. These biological properties lead to a rapid and improved endothelialization of the neoarterial wall at the aneurysm neck. CD31-mimetic coating could therefore represent a valuable strategy for FD biocompatibility improvement and aneurysm healing.
Collapse
Affiliation(s)
- Jonathan Cortese
- NEURI Center, Hôpital Bicêtre, APHP-Université Paris Sud, Kremlin-Bicêtre, France (J. Cortese, J. Caroff, L.S.).,Laboratory for Vascular Translational Science, Université de Paris, INSERM U1148, France (J. Cortese, C.R., G.E., C.C., J.M., A.N., J.-B.M., G.C.)
| | - Charlotte Rasser
- Laboratory for Vascular Translational Science, Université de Paris, INSERM U1148, France (J. Cortese, C.R., G.E., C.C., J.M., A.N., J.-B.M., G.C.)
| | - Guillaume Even
- Laboratory for Vascular Translational Science, Université de Paris, INSERM U1148, France (J. Cortese, C.R., G.E., C.C., J.M., A.N., J.-B.M., G.C.)
| | - Sylvia M Bardet
- University of Limoges, XLIM UMR CNRS 7252, France (S.M.B., M.-L.P., A.R.)
| | - Christine Choqueux
- Laboratory for Vascular Translational Science, Université de Paris, INSERM U1148, France (J. Cortese, C.R., G.E., C.C., J.M., A.N., J.-B.M., G.C.)
| | - Jules Mesnier
- Laboratory for Vascular Translational Science, Université de Paris, INSERM U1148, France (J. Cortese, C.R., G.E., C.C., J.M., A.N., J.-B.M., G.C.)
| | - Marie-Laure Perrin
- University of Limoges, XLIM UMR CNRS 7252, France (S.M.B., M.-L.P., A.R.)
| | - Kevin Janot
- Department of Interventional Neuroradiology, Limoges University Hospital, France (K.J., A.R.)
| | - Jildaz Caroff
- NEURI Center, Hôpital Bicêtre, APHP-Université Paris Sud, Kremlin-Bicêtre, France (J. Cortese, J. Caroff, L.S.)
| | - Antonino Nicoletti
- Laboratory for Vascular Translational Science, Université de Paris, INSERM U1148, France (J. Cortese, C.R., G.E., C.C., J.M., A.N., J.-B.M., G.C.)
| | - Jean-Baptiste Michel
- Laboratory for Vascular Translational Science, Université de Paris, INSERM U1148, France (J. Cortese, C.R., G.E., C.C., J.M., A.N., J.-B.M., G.C.)
| | - Laurent Spelle
- NEURI Center, Hôpital Bicêtre, APHP-Université Paris Sud, Kremlin-Bicêtre, France (J. Cortese, J. Caroff, L.S.)
| | - Giuseppina Caligiuri
- Laboratory for Vascular Translational Science, Université de Paris, INSERM U1148, France (J. Cortese, C.R., G.E., C.C., J.M., A.N., J.-B.M., G.C.)
| | - Aymeric Rouchaud
- Department of Interventional Neuroradiology, Limoges University Hospital, France (K.J., A.R.).,University of Limoges, XLIM UMR CNRS 7252, France (S.M.B., M.-L.P., A.R.)
| |
Collapse
|
40
|
Henrot P, Laurent P, Levionnois E, Leleu D, Pain C, Truchetet ME, Cario M. A Method for Isolating and Culturing Skin Cells: Application to Endothelial Cells, Fibroblasts, Keratinocytes, and Melanocytes From Punch Biopsies in Systemic Sclerosis Skin. Front Immunol 2020; 11:566607. [PMID: 33117350 PMCID: PMC7575752 DOI: 10.3389/fimmu.2020.566607] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/31/2020] [Indexed: 11/13/2022] Open
Abstract
Systemic Sclerosis (SSc) is a complex auto-immune connective tissue disease combining inflammatory, vasculopathic and fibrotic manifestations. Skin effectively recapitulates the main pathogenic processes and therefore is a good organ to decipher the disease pathophysiology, which remains unclear. However, culturing primary skin cells is SSc can be a major issue due to small sample size combined to skin fibrosis. Here, we present a protocol allowing to isolate and culture the four main types of skin cells: dermal cells (microvascular dermal endothelial cells—HDMECs—and fibroblasts) and epidermal cells (keratinocytes and melanocytes), from a single 4 mm-punch biopsy, at a low cost. The present protocol has been optimized to fit SSc skin cells particularities. Such technique allows to culture primary cells, crucial to study the disease pathophysiology, as well as to isolate cells in order to perform immediate molecular biology experiments such as single-cell transcriptomic. Cells grown from biopsies are also suitable for various types of experiments such as immunocytochemistry, Western blot, RT-qPCR or functional in vitro assays (angiogenesis, migration, etc.). Ultimately, they can be used for experimental 3D cell culture models such as reconstructed skin.
Collapse
Affiliation(s)
- Pauline Henrot
- Univ. Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France.,Department of Rheumatology, National Reference Center for Systemic Autoimmune Rare Diseases, Hopital Pellegrin, Bordeaux, France
| | - Paôline Laurent
- Univ. Bordeaux, CNRS, Immunoconcept, UMR 5164, Bordeaux, France
| | | | - Damien Leleu
- Univ. Bordeaux, CNRS, Immunoconcept, UMR 5164, Bordeaux, France
| | - Catherine Pain
- Univ. Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France
| | - Marie-Elise Truchetet
- Department of Rheumatology, National Reference Center for Systemic Autoimmune Rare Diseases, Hopital Pellegrin, Bordeaux, France.,Univ. Bordeaux, CNRS, Immunoconcept, UMR 5164, Bordeaux, France
| | - Muriel Cario
- Univ. Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France.,Department of Dermatology and Pediatric Dermatology, National Center for Rare Skin Disorders, Hôpital Saint André, Bordeaux, France.,AquiDerm, Univ. Bordeaux, Bordeaux, France
| |
Collapse
|
41
|
Luo L, Xu M, Liao D, Deng J, Mei H, Hu Y. PECAM-1 protects against DIC by dampening inflammatory responses via inhibiting macrophage pyroptosis and restoring vascular barrier integrity. Transl Res 2020; 222:1-16. [PMID: 32417429 DOI: 10.1016/j.trsl.2020.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/12/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022]
Abstract
Disseminated intravascular coagulation (DIC) is a frequent complication of sepsis that affects patient outcomes due to accompanying thrombo-inflammation and microvascular permeability changes. Platelet endothelial cell adhesion molecule-1 (PECAM-1), a cellular adhesion and signaling receptor that is expressed on both hematopoietic and endothelial cells, plays an important anti-inflammatory role in acute and chronic inflammatory disease models. Little is known, however, about role and mechanism of PECAM-1 in septic DIC. Here, we investigated whether PECAM-1 might play a protective role in hindering the development of septic DIC. Plasma levels of soluble PECAM-1 were markedly elevated in septic patients that developed DIC, with a correspondingly poorer outcome. PECAM-1 knockout exhibited more severe DIC and poorer outcome in the LPS induced- and cecal ligation and puncture-induced DIC model, which could be alleviated by tissue factor inhibitor. This phenomenon seemed to be equally linked to PECAM-1 expression by both endothelial and blood cells. Furthermore, PECAM-1 was found to exert its protective effect on developing septic DIC by the following 2 distinct mechanisms: the inhibition of macrophage pyroptosis and the acceleration of the restoration of the endothelial cell barrier. Taken together, these results implicate PECAM-1 as a potentially attractive target for the development of novel therapeutics to manage and treat septic DIC.
Collapse
Affiliation(s)
- Lili Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Min Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Danying Liao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| |
Collapse
|
42
|
McGuire HM, Rizzetto S, Withers BP, Clancy LE, Avdic S, Stern L, Patrick E, Fazekas de St Groth B, Slobedman B, Gottlieb DJ, Luciani F, Blyth E. Mass cytometry reveals immune signatures associated with cytomegalovirus (CMV) control in recipients of allogeneic haemopoietic stem cell transplant and CMV-specific T cells. Clin Transl Immunology 2020; 9:e1149. [PMID: 32642063 PMCID: PMC7332355 DOI: 10.1002/cti2.1149] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/22/2022] Open
Abstract
Objectives Cytomegalovirus (CMV) is known to have a significant impact on immune recovery post‐allogeneic haemopoietic stem cell transplant (HSCT). Adoptive therapy with donor‐derived or third‐party virus‐specific T cells (VST) can restore CMV immunity leading to clinical benefit in prevention and treatment of post‐HSCT infection. We developed a mass cytometry approach to study natural immune recovery post‐HSCT and assess the mechanisms underlying the clinical benefits observed in recipients of VST. Methods A mass cytometry panel of 38 antibodies was utilised for global immune assessment (72 canonical innate and adaptive immune subsets) in HSCT recipients undergoing natural post‐HSCT recovery (n = 13) and HSCT recipients who received third‐party donor‐derived CMV‐VST as salvage for unresponsive CMV reactivation (n = 8). Results Mass cytometry identified distinct immune signatures associated with CMV characterised by a predominance of innate cells (monocytes and NK) seen early and an adaptive signature with activated CD8+ T cells seen later. All CMV‐VST recipients had failed standard antiviral pharmacotherapy as a criterion for trial involvement; 5/8 had failed to develop the adaptive immune signature by study enrolment despite significant CMV antigen exposure. Of these, VST administration resulted in development of the adaptive signature in association with CMV control in three patients. Failure to respond to CMV‐VST in one patient was associated with persistent absence of the adaptive immune signature. Conclusion The clinical benefit of CMV‐VST may be mediated by the recovery of an adaptive immune signature characterised by activated CD8+ T cells.
Collapse
Affiliation(s)
- Helen M McGuire
- Ramaciotti Facility for Human Systems Biology The University of Sydney Sydney NSW Australia.,Charles Perkins Centre The University of Sydney Sydney NSW Australia.,Discipline of Pathology Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia
| | - Simone Rizzetto
- Kirby Institute for Infection and Immunity University of New South Wales Sydney NSW Australia.,School of Medical Sciences University of New South Wales Kensington NSW Australia
| | - Barbara P Withers
- Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,St Vincent's Hospital Darlinghurst NSW Australia
| | - Leighton E Clancy
- Sydney Cellular Therapies Laboratory Westmead NSW Australia.,BMT and Cell Therapies Program Westmead Hospital Sydney NSW Australia.,Westmead Institute for Medical Research The University of Sydney Sydney NSW Australia
| | - Selmir Avdic
- Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Westmead Institute for Medical Research The University of Sydney Sydney NSW Australia
| | - Lauren Stern
- Charles Perkins Centre The University of Sydney Sydney NSW Australia.,Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Discipline of Infectious Diseases and Immunology Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia
| | - Ellis Patrick
- Westmead Institute for Medical Research The University of Sydney Sydney NSW Australia.,School of Mathematics and Statistics Faculty of Science The University of Sydney Sydney NSW Australia
| | - Barbara Fazekas de St Groth
- Ramaciotti Facility for Human Systems Biology The University of Sydney Sydney NSW Australia.,Charles Perkins Centre The University of Sydney Sydney NSW Australia.,Discipline of Pathology Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia
| | - Barry Slobedman
- Charles Perkins Centre The University of Sydney Sydney NSW Australia.,Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Discipline of Infectious Diseases and Immunology Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia
| | - David J Gottlieb
- Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,BMT and Cell Therapies Program Westmead Hospital Sydney NSW Australia.,Westmead Institute for Medical Research The University of Sydney Sydney NSW Australia
| | - Fabio Luciani
- Kirby Institute for Infection and Immunity University of New South Wales Sydney NSW Australia.,School of Medical Sciences University of New South Wales Kensington NSW Australia
| | - Emily Blyth
- Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Sydney Cellular Therapies Laboratory Westmead NSW Australia.,BMT and Cell Therapies Program Westmead Hospital Sydney NSW Australia.,Westmead Institute for Medical Research The University of Sydney Sydney NSW Australia
| |
Collapse
|
43
|
Saul J, Hutchins E, Reiman R, Saul M, Ostrow LW, Harris BT, Van Keuren-Jensen K, Bowser R, Bakkar N. Global alterations to the choroid plexus blood-CSF barrier in amyotrophic lateral sclerosis. Acta Neuropathol Commun 2020; 8:92. [PMID: 32586411 PMCID: PMC7318439 DOI: 10.1186/s40478-020-00968-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/15/2020] [Indexed: 01/08/2023] Open
Abstract
The choroid plexus (CP) is a highly vascularized structure located in the ventricles that forms the blood-CSF barrier (BCSFB) and separates the blood from the cerebrospinal fluid (CSF). In addition to its role as a physical barrier, the CP functions in CSF secretion, transport of nutrients into the central nervous system (CNS) and a gated point of entry of circulating immune cells into the CNS. Aging and neurodegeneration have been reported to affect CP morphology and function and increase protein leakage from blood to the CSF. Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease associated with both upper and lower motor neuron loss, as well as altered proteomic and metabolomic signatures in the CSF. The role of the BCSFB and the CP in ALS is unknown. Here we describe a transcriptomic and ultrastructural analysis of BCSFB and CP alterations in human postmortem tissues from ALS and non-neurologic disease controls. ALS-CP exhibited widespread disruptions in tight junctional components of the CP epithelial layer and vascular integrity. In addition, we detected loss of pericytes around ALS blood vessels, accompanied by activation of platelet aggregation markers vWF and Fibrinogen, reminiscent of vascular injury. To investigate the immune component of ALS-CP, we conducted a comprehensive analysis of cytokines and chemokine panels in CP lysates and found a significant down-regulation of M-CSF and V-CAM1 in ALS, as well as up-regulation of VEGF-A protein. This phenotype was accompanied by an infiltration of MERTK positive macrophages into the parenchyma of the ALS-CP when compared to controls. Taken together, we demonstrate widespread structural and functional disruptions of the BCSFB in human ALS increasing our understanding of the disease pathology and identifying potential new targets for ALS therapeutic development.
Collapse
|
44
|
Truffault F, Nazzal D, Verdier J, Gradolatto A, Fadel E, Roussin R, Eymard B, Le Panse R, Berrih-Aknin S. Comparative Analysis of Thymic and Blood Treg in Myasthenia Gravis: Thymic Epithelial Cells Contribute to Thymic Immunoregulatory Defects. Front Immunol 2020; 11:782. [PMID: 32435245 PMCID: PMC7218102 DOI: 10.3389/fimmu.2020.00782] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/06/2020] [Indexed: 11/13/2022] Open
Abstract
The thymus is involved in autoimmune Myasthenia gravis (MG) associated with anti-acetylcholine (AChR) antibodies. In MG, thymic regulatory T cells (Treg) are not efficiently suppressive, and conventional T cells (Tconv) are resistant to suppression. To better understand the specific role of the thymus in MG, we compared the phenotype and function of peripheral and thymic Treg and Tconv from controls and MG patients. Suppression assays with thymic or peripheral CD4 + T cells showed that the functional impairment in MG was more pronounced in the thymus than in the periphery. Phenotypic analysis of Treg showed a significant reduction of resting and effector Treg in the thymus but not in the periphery of MG patients. CD31, a marker lost with excessive immunoreactivity, was significantly reduced in thymic but not blood resting Treg. These results suggest that an altered thymic environment may explain Treg differences between MG patients and controls. Since thymic epithelial cells (TECs) play a major role in the generation of Treg, we co-cultured healthy thymic CD4 + T cells with control or MG TECs and tested their suppressive function. Co-culture with MG TECs consistently hampers regulatory activity, as compared with control TECs, suggesting that MG TECs contribute to the immune regulation defects of MG CD4 + T cells. MG TECs produced significantly higher thymic stromal lymphopoietin (TSLP) than control TECs, and a neutralizing anti-TSLP antibody partially restored the suppressive capacity of Treg derived from co-cultures with MG TECs, suggesting that TSLP contributed to the defect of thymic Treg in MG patients. Finally, a co-culture of MG CD4 + T cells with control TECs restored numbers and function of MG Treg, demonstrating that a favorable environment could correct the immune regulation defects of T cells in MG. Altogether, our data suggest that the severe defect of thymic Treg is at least partially due to MG TECs that overproduce TSLP. The Treg defects could be corrected by replacing dysfunctional TECs by healthy TECs. These findings highlight the role of the tissue environment on the immune regulation.
Collapse
Affiliation(s)
- Frédérique Truffault
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Dani Nazzal
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Julien Verdier
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Angeline Gradolatto
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Elie Fadel
- Marie Lannelongue Hospital, Le Plessis-Robinson, France
| | | | - Bruno Eymard
- AP-HP, Referral Center for Neuromuscular Disorders, Pitié-Salpêtrière Hospital, Institute of Myology, Paris, France
| | - Rozen Le Panse
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Sonia Berrih-Aknin
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| |
Collapse
|
45
|
Abstract
The potential of CD31 as a therapeutic target in atherosclerosis has been considered ever since its cloning in the 1990s, but the exact role played by this molecule in the biologic events underlying atherosclerosis has remained controversial, resulting in the stalling of any therapeutic perspective. Due to the supposed cell adhesive properties of CD31, specific monoclonal antibodies and recombinant proteins were regarded as blocking agents because their use prevented the arrival of leukocytes at sites of acute inflammation. However, the observed effect of those compounds likely resulted from the engagement of the immunomodulatory function of CD31 signaling. This was acknowledged only later though, upon the discovery of CD31's 2 intracytoplasmic tyrosine residues called immunoreceptor tyrosine inhibitory motifs. A growing body of evidence currently points at a therapeutic potential for CD31 agonists in atherothrombosis. Clinical observations show that CD31 expression is altered at the surface of leukocytes infiltrating unhealed atherothrombotic lesions and that the physiological immunomodulatory functions of CD31 are lost at the surface of blood leukocytes in patients with acute coronary syndromes. On the contrary, translational studies using candidate therapeutic molecules in laboratory animals have provided encouraging results: synthetic peptides administered to atherosclerotic mice as systemic drugs in the acute phases of atherosclerotic complications favor the healing of wounded arteries, whereas the immobilization of CD31 agonist peptides onto coronary stents implanted in farm pigs favors their peaceful integration within the coronary arterial wall.
Collapse
Affiliation(s)
- Giuseppina Caligiuri
- From the Laboratory for Vascular Translational Science, Inserm U1148, Université de Paris, France; and Department of Cardiology, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Nord Val-de-Seine, Site Bichat, France
| |
Collapse
|
46
|
Figueiredo CR, Kalirai H, Sacco JJ, Azevedo RA, Duckworth A, Slupsky JR, Coulson JM, Coupland SE. Loss of BAP1 expression is associated with an immunosuppressive microenvironment in uveal melanoma, with implications for immunotherapy development. J Pathol 2020; 250:420-439. [PMID: 31960425 PMCID: PMC7216965 DOI: 10.1002/path.5384] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/28/2019] [Accepted: 01/14/2020] [Indexed: 12/22/2022]
Abstract
Immunotherapy using immune checkpoint inhibitors (ICIs) induces durable responses in many metastatic cancers. Metastatic uveal melanoma (mUM), typically occurring in the liver, is one of the most refractory tumours to ICIs and has dismal outcomes. Monosomy 3 (M3), polysomy 8q, and BAP1 loss in primary uveal melanoma (pUM) are associated with poor prognoses. The presence of tumour-infiltrating lymphocytes (TILs) within pUM and surrounding mUM - and some evidence of clinical responses to adoptive TIL transfer - strongly suggests that UMs are indeed immunogenic despite their low mutational burden. The mechanisms that suppress TILs in pUM and mUM are unknown. We show that BAP1 loss is correlated with upregulation of several genes associated with suppressive immune responses, some of which build an immune suppressive axis, including HLA-DR, CD38, and CD74. Further, single-cell analysis of pUM by mass cytometry confirmed the expression of these and other markers revealing important functions of infiltrating immune cells in UM, most being regulatory CD8+ T lymphocytes and tumour-associated macrophages (TAMs). Transcriptomic analysis of hepatic mUM revealed similar immune profiles to pUM with BAP1 loss, including the expression of IDO1. At the protein level, we observed TAMs and TILs entrapped within peritumoural fibrotic areas surrounding mUM, with increased expression of IDO1, PD-L1, and β-catenin (CTNNB1), suggesting tumour-driven immune exclusion and hence the immunotherapy resistance. These findings aid the understanding of how the immune response is organised in BAP1 - mUM, which will further enable functional validation of detected biomarkers and the development of focused immunotherapeutic approaches. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Carlos R Figueiredo
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Department of the Faculty of Medicine, MediCity Research Laboratory and Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Helen Kalirai
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Joseph J Sacco
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Department of Medical OncologyThe Clatterbridge Cancer CentreWirralUK
| | - Ricardo A Azevedo
- Department of Cancer BiologyThe University of Texas–MD Anderson Cancer CenterHoustonTXUSA
| | - Andrew Duckworth
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Joseph R Slupsky
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Judy M Coulson
- Department of Cellular and Molecular PhysiologyUniversity of LiverpoolLiverpoolUK
| | - Sarah E Coupland
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Liverpool Clinical LaboratoriesRoyal Liverpool University HospitalLiverpoolUK
| |
Collapse
|
47
|
Lipski DA, Foucart V, Dewispelaere R, Caspers LE, Defrance M, Bruyns C, Willermain F. Retinal endothelial cell phenotypic modifications during experimental autoimmune uveitis: a transcriptomic approach. BMC Ophthalmol 2020; 20:106. [PMID: 32183784 PMCID: PMC7076950 DOI: 10.1186/s12886-020-1333-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 02/03/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Blood-retinal barrier cells are known to exhibit a massive phenotypic change during experimental autoimmune uveitis (EAU) development. In an attempt to investigate the mechanisms of blood-retinal barrier (BRB) breakdown at a global level, we studied the gene regulation of total retinal cells and retinal endothelial cells during non-infectious uveitis. METHODS Retinal endothelial cells were isolated by flow cytometry either in Tie2-GFP mice (CD31+ CD45- GFP+ cells), or in wild type C57BL/6 mice (CD31+ CD45- endoglin+ cells). EAU was induced in C57BL/6 mice by adoptive transfer of IRBP1-20-specific T cells. Total retinal cells and retinal endothelial cells from naïve and EAU mice were sorted and their gene expression compared by RNA-Seq. Protein expression of selected genes was validated by immunofluorescence on retinal wholemounts and cryosections and by flow cytometry. RESULTS Retinal endothelial cell sorting in wild type C57BL/6 mice was validated by comparative transcriptome analysis with retinal endothelial cells sorted from Tie2-GFP mice, which express GFP under the control of the endothelial-specific receptor tyrosine kinase promoter Tie2. RNA-Seq analysis of total retinal cells mainly brought to light upregulation of genes involved in antigen presentation and T cell activation during EAU. Specific transcriptome analysis of retinal endothelial cells allowed us to identify 82 genes modulated in retinal endothelial cells during EAU development. Protein expression of 5 of those genes (serpina3n, lcn2, ackr1, lrg1 and lamc3) was validated at the level of inner BRB cells. CONCLUSION Those data not only confirm the involvement of known pathogenic molecules but further provide a list of new candidate genes and pathways possibly implicated in inner BRB breakdown during non-infectious posterior uveitis.
Collapse
Affiliation(s)
- Deborah A. Lipski
- Ophthalmology Group, IRIBHM (Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire), Université Libre de Bruxelles (ULB), Erasme Campus, Building C, Room C6.117, 808 Route de Lennik, 1070 Brussels, Belgium
- Ophthalmology Department of Erasme Hospital, Université Libre de Bruxelles (ULB), 808 Route de Lennik, 1070 Brussels, Belgium
| | - Vincent Foucart
- Ophthalmology Group, IRIBHM (Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire), Université Libre de Bruxelles (ULB), Erasme Campus, Building C, Room C6.117, 808 Route de Lennik, 1070 Brussels, Belgium
- Ophthalmology Department of CHU Saint-Pierre, 322 Rue Haute, 1000 Brussels, Belgium
- Ophthalmology Department of CHU Brugmann, 4 Place Van Gehuchten, 1020 Brussels, Belgium
| | - Rémi Dewispelaere
- Ophthalmology Group, IRIBHM (Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire), Université Libre de Bruxelles (ULB), Erasme Campus, Building C, Room C6.117, 808 Route de Lennik, 1070 Brussels, Belgium
- Ophthalmology Department of CHU Saint-Pierre, 322 Rue Haute, 1000 Brussels, Belgium
| | - Laure E. Caspers
- Ophthalmology Department of CHU Saint-Pierre, 322 Rue Haute, 1000 Brussels, Belgium
| | - Matthieu Defrance
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles - Vrije Universiteit Brussel, La Plaine Campus, BC building, 6th floor, CP 263, Triomflaan, 1050 Brussels, Belgium
| | - Catherine Bruyns
- Ophthalmology Group, IRIBHM (Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire), Université Libre de Bruxelles (ULB), Erasme Campus, Building C, Room C6.117, 808 Route de Lennik, 1070 Brussels, Belgium
| | - François Willermain
- Ophthalmology Group, IRIBHM (Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire), Université Libre de Bruxelles (ULB), Erasme Campus, Building C, Room C6.117, 808 Route de Lennik, 1070 Brussels, Belgium
- Ophthalmology Department of CHU Saint-Pierre, 322 Rue Haute, 1000 Brussels, Belgium
- Ophthalmology Department of CHU Brugmann, 4 Place Van Gehuchten, 1020 Brussels, Belgium
| |
Collapse
|
48
|
Zhan S, Liu Z, Zhang M, Guo T, Quan Q, Huang L, Guo L, Cao L, Zhang X. Overexpression of B7-H3 in α-SMA-Positive Fibroblasts Is Associated With Cancer Progression and Survival in Gastric Adenocarcinomas. Front Oncol 2020; 9:1466. [PMID: 31998637 PMCID: PMC6966326 DOI: 10.3389/fonc.2019.01466] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/06/2019] [Indexed: 12/14/2022] Open
Abstract
Background: B7-H3 promotes tumor immune escape and is highly expressed in tumor tissues. Stromal cells in tumors, including fibroblasts, play an important role in this process; however, the role of B7-H3 in tumor fibroblasts has not been fully clarified. Methods: We examined B7-H3, CD31, and alpha-smooth muscle actin (α-SMA) protein expression in 268 gastric adenocarcinomas (GACs) by immunohistochemistry. The coexpression of B7-H3 with CD31 or α-SMA was examined using immunofluorescence double staining. Cytokine expression from fibroblasts treated with B7-H3 small interfering RNA (siRNA) was analyzed by a Quantitative reverse transcription-polymerase chain reaction (qPCR) and Enzyme-linked immunosorbent assay (ELISA). The transwell tests were conducted to assess the migration and invasion ability of fibroblasts. The overall survival was analyzed by a Kaplan-Meier analysis. Associations between categorical variables were assessed using the Pearson's Chi-square test or Fisher's exact test. Results: GAC patients with B7-H3 expression showed significantly poorer survival (P = 0.012). The overall survival of the group with high B7-H3 expression was significantly worse than the group with low B7-H3 expression in both tumor cells and in stromal cells (P = 0.007 and P = 0.048, respectively). B7-H3 expression correlated with many clinicopathological data, including tumor stage, tumor depth, lymph node involvement, and survival. Immunofluorescence staining showed that B7-H3 was expressed in tumor cells and α-SMA-positive fibroblasts. Remarkably, high expression of α-SMA was associated with a poor prognosis (P = 0.007), and the prognoses of patients with high stromal expression of B7-H3 and α-SMA were significantly worse than that of other combination types (P = 0.001). Additionally, the absence of B7-H3 led to decreased secretion of cytokines, such as interleukin (IL)-6 and vascular endothelial growth factor (VEGF), as well as a decline in migration and invasion ability in cancer-associated fibroblasts (CAFs). Conclusions: Patients with high B7-H3 expression either in tumor cells or in stromal cells had significantly poorer overall survival. Stromal B7-H3 expression was mostly detected in α-SMA-positive CAFs. GAC patients with both stromal B7-H3-high and α-SMA-high expression had significantly poorer overall survival, suggesting that stromal B7-H3 and α-SMA expression status can serve as an indicator of poor prognosis for GAC patients.
Collapse
Affiliation(s)
- Shenghua Zhan
- Department of Pathology, Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiju Liu
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Zhang
- Department of Pathology, Children's Hospital of Soochow University, Suzhou, China
| | - Tianwei Guo
- Department of Pathology, Changshu Hospital of Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Qiuying Quan
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lili Huang
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, China
| | - Lingchuan Guo
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lei Cao
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| |
Collapse
|
49
|
Zhang X, Hubal MJ, Kraus VB. Immune cell extracellular vesicles and their mitochondrial content decline with ageing. IMMUNITY & AGEING 2020; 17:1. [PMID: 31911808 PMCID: PMC6942666 DOI: 10.1186/s12979-019-0172-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/29/2019] [Indexed: 12/30/2022]
Abstract
Background Although the mechanisms of action are not fully understood, extracellular vesicles (EVs) have emerged as key indicators and effectors of immune function. Characterizing circulating EVs associated with stem and immune cells across the lifespan of healthy individuals could aid an understanding of immunosenescence, a process of age-related decline of cells in both adaptive and innate immune systems. Results Using high resolution multicolor flow cytometry, we identified three major subsets of EVs of varying sizes in healthy control (HC) plasma. Multiple plasma EVs associated with immune cells declined with ageing in HCs. In addition, we observed age-associated declines of respiring mitochondria cargo in EVs of several types of immune cells, suggesting that these parent cells may experience a decline in mitophagy or a mitochondrial dysfunction-induced immunosenescence. By contrast, the number of CD34+ hematopoietic stem cell-associated EVs were high and carried respiring mitochondria, which did not decline with age. Conclusion As demonstrated here, multicolor flow cytometry simultaneously measures plasma EV size, surface markers and cargo that reflect biological processes of specific cell types. The distinct surface markers and cytokine cargo of plasma EVs suggest that they may carry different bio-messages and originate by different biogenesis pathways.
Collapse
Affiliation(s)
- Xin Zhang
- 1Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, North Carolina 27701 USA.,2Department of Orthopaedic Surgery, Duke University School of Medicine, Duke University, Durham, North Carolina USA
| | - Monica Jeanne Hubal
- 3School of Health and Human Sciences, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana USA
| | - Virginia Byers Kraus
- 1Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, North Carolina 27701 USA.,4Department of Medicine, Duke University School of Medicine, Duke University, Durham, North Carolina USA
| |
Collapse
|
50
|
Cleaved CD31 as a target for in vivo molecular imaging of inflammation. Sci Rep 2019; 9:19560. [PMID: 31863037 PMCID: PMC6925130 DOI: 10.1038/s41598-019-56163-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/02/2019] [Indexed: 01/16/2023] Open
Abstract
There is a need for new targets to specifically localize inflammatory foci, usable in a wide range of organs. Here, we hypothesized that the cleaved molecular form of CD31 is a suitable target for molecular imaging of inflammation. We evaluated a bioconjugate of D-P8RI, a synthetic peptide that binds all cells with cleaved CD31, in an experimental rat model of sterile acute inflammation. Male Wistar rats were injected with turpentine oil into the gastrocnemius muscle two days before 99mTc-HYNIC-D-P8RI (or its analogue with L-Proline) SPECT/CT or [18F]FDG PET/MRI. Biodistribution, stability study, histology, imaging and autoradiography of 99mTc-HYNIC-D-P8RI were further performed. Biodistribution studies revealed rapid elimination of 99mTc-HYNIC-D-P8RI through renal excretion with almost no uptake from most organs and excellent in vitro and in vivo stability were observed. SPECT/CT imaging showed a significant higher 99mTc-HYNIC-D-P8RI uptake compared with its analogue with L-Proline (negative control) and no significant difference compared with [18F]FDG (positive control). Moreover, autoradiography and histology revealed a co-localization between 99mTc-HYNIC-D-P8RI uptake and inflammatory cell infiltration. 99mTc-HYNIC-D-P8RI constitutes a new tool for the detection and localization of inflammatory sites. Our work suggests that targeting cleaved CD31 is an attractive strategy for the specific in vivo imaging of inflammatory processes.
Collapse
|