1
|
Zhu Y, Yuan H, Qu H. Serum alpha 1 antitrypsin potent act as an early diagnostic biomarker for cardiac amyloidosis. Heart Vessels 2024; 39:803-809. [PMID: 38580850 DOI: 10.1007/s00380-024-02396-4] [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: 12/08/2022] [Accepted: 03/13/2024] [Indexed: 04/07/2024]
Abstract
Cardiac amyloidosis is a refractory cardiomyopathy with a poor prognosis and lacks effective treatments. N-terminal pro-brain natriuretic peptide (NT-proBNP) and troponin T are poor prognostic factors for myocardial amyloidosis. However, NT-proBNP and troponin also serve as markers of heart failure and myocardial infarction, lacking specificity. Whether abnormal elevation of alpha-1 antitrypsin in myocardial amyloidosis also predicts the poor prognosis of patients remains unknown. We conducted a retrospective single-center case-control study to analyze the serological and physical examination data of 83 cardiac amyloidosis patients and 68 healthy controls matched by gender and age. We aimed to explore the onset and prognostic factors of cardiac amyloidosis. The serum alpha-1 antitrypsin level (169.78 ± 39.59 mg/dl) in patients with cardiac amyloidosis was significantly higher than that in the normal control (125.92 ± 18.26 mg/dl). Logistic regression results showed that alpha-1 antitrypsin, free sialic acid, high-density lipoprotein cholesterol, apolipoprotein A/B ratio, and homocysteine were predictors of cardiac amyloidosis. Multivariable logistic regression showed that only alpha 1 antitrypsin was an independent risk factor for cardiac amyloidosis. Receiver operating characteristic curve analysis based on the Mayo stage and troponin level showed the cut-off value of 140.55 mg/dl for alpha-1 antitrypsin in predicting cardiac amyloidosis with 81.7% sensitivity and 83.9% specificity. Elevated alpha-1 antitrypsin levels may be an early diagnostic biomarker for cardiac amyloidosis.
Collapse
Affiliation(s)
- Ye Zhu
- Department of Cardiac Medicine, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Internal Medicine, The Fifth People's Hospital of Jinan, Jinan, Shandong, China
| | - Haitao Yuan
- Department of Cardiac Medicine, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Huiting Qu
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
2
|
Ma Y, Chen Y, Zhan L, Dong Q, Wang Y, Li X, He L, Zhang J. CEBPB-mediated upregulation of SERPINA1 promotes colorectal cancer progression by enhancing STAT3 signaling. Cell Death Discov 2024; 10:219. [PMID: 38710698 DOI: 10.1038/s41420-024-01990-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024] Open
Abstract
Colorectal cancer (CRC) is a highly malignant carcinoma associated with poor prognosis, and metastasis is one of the most common causes of death in CRC. Serpin Family A Member 1 (SERPINA1) is a serine protease inhibitor from the Serpin family. Till now, the function and mechanism of SERPINA1 in CRC progression have not been fully illustrated. We established highly metastatic colorectal cancer cells named as RKO-H and Caco2-H by mice liver metastasis model. By integrative bioinformatic approaches, we analyzed the prognostic value and clinical significance of SERPINA1 in CRC, and predicted potential transcription factors. Colony formation, EDU, MTS, Transwell and wound healing assay were performed to evaluate the biological functions of SERPINA1 in CRC in vitro. Experiments in vivo were conducted to explore the effects of SERPINA1 on liver metastasis of CRC. ChIP and luciferase reporter gene assays were performed to identify the transcriptional regulatory mechanism of SERPINA1 by CEBPB. Our results show that SERPINA1 is highly expressed in CRC and correlated with poor clinical outcomes. SERPINA1 promotes the proliferation, migration by activating STAT3 pathway. Mechanistically, CEBPB binds SERPINA1 gene promoter sequence and promotes the transcription of SERPINA1. SERPINA1 drives CEBPB-induced tumor cell growth and migration via augmenting STAT3 signaling. Our results suggest that SERPINA1 is a potential prognostic marker and may serve as a novel treatment target for CRC.
Collapse
Affiliation(s)
- Yiming Ma
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning Province, China
| | - Ying Chen
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Lei Zhan
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning Province, China
| | - Qian Dong
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning Province, China
| | - Yuanhe Wang
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning Province, China
| | - Xiaoyan Li
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning Province, China
- Department of Pathology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Lian He
- Department of Pathology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Jingdong Zhang
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China.
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning Province, China.
| |
Collapse
|
3
|
Ishii S, Sakaguchi W, Yamamura M, Nagumo T, Koeda S, Akiyama H, Kinuta M, Nishikubo S, Tsukinoki K. Association between salivary proteases and protease inhibitors linked with viral infections and oral inflammatory diseases. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2023; 124:101572. [PMID: 37495185 DOI: 10.1016/j.jormas.2023.101572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 07/24/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION Despite the role of transmembrane protease, serine 2 (TMPRSS2) in facilitating the entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the primary cause of the global COVID-19 pandemic, the interaction of extracellular and intracellular proteases in this process remains poorly elucidated. Thus, we monitored the salivary expression concentration (SEC) of TMPRSS2 and its inhibitor, alpha-1 antitrypsin (A1AT), and investigated whether oral inflammatory diseases affected the SEC of both proteins. MATERIALS AND METHODS We collected saliva samples before and after surgical treatment of inflammatory cystic diseases (radicular and inflammatory dentigerous cysts) in 25 patients. The SEC of TMPRSS2 and A1AT was measured using enzyme-linked immunosorbent assay. SEC in multiple patient status groups and subgroups of each status were investigated. Finally, the correlation between TMPRSS2 and A1AT SEC was analyzed. RESULTS The TMPRSS2 and A1AT SEC did not significantly change pre- or post-treatment. The TMPRSS2 SEC was significantly higher before and after treatment in patients aged >50 years, patients with radicular cysts, and patients with the basic disease. A1AT SEC was significantly decreased after treatment in the acute inflammation, large-sized, and patients without basic disease groups. No significant correlation was observed between the SEC of either protein before and after treatment. DISCUSSION Individual-specific SEC for TMPRSS2 may be influenced by age, lesion type, and basic disease; however, oral inflammatory diseases may not have a direct effect. Moreover, the extent of oral inflammatory diseases and the presence of basic diseases may be associated with A1AT SEC. Furthermore, the SEC between the two proteins may be independent.
Collapse
Affiliation(s)
- Shigeru Ishii
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Wakako Sakaguchi
- Department of Environmental Pathology, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka City, Kanagawa, 238-8580, Japan.
| | - Makiko Yamamura
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Tatsuhito Nagumo
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Satoko Koeda
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Hiroki Akiyama
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Mikihisa Kinuta
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Shuichi Nishikubo
- Department of Advanced Oral Surgery, Kanagawa Dental University, Yokohama Clinic, 3-31-6 Tsuruya-cho, Kanagawa-ku, Yokohama City, Kanagawa, 221-0835, Japan.
| | - Keiichi Tsukinoki
- Department of Environmental Pathology, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka City, Kanagawa, 238-8580, Japan.
| |
Collapse
|
4
|
Sun R, Xu Z, Zhu C, Chen T, Muñoz LE, Dai L, Zhao Y. Alpha-1 antitrypsin in autoimmune diseases: Roles and therapeutic prospects. Int Immunopharmacol 2022; 110:109001. [PMID: 35803133 DOI: 10.1016/j.intimp.2022.109001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 02/05/2023]
Abstract
Alpha-1 antitrypsin (A1AT) is a protease inhibitor in the serum. Its primary function is to inhibit the activity of a series of proteases, including proteinase 3, neutrophil elastase, metalloproteases, and cysteine-aspartate proteases. In addition, A1AT also has anti-inflammatory, anti-apoptotic, anti-oxidative stress, anti-viral, and anti-bacterial activities and plays essential roles in the regulation of tissue repair and lymphocyte differentiation and activation. The overactivation of the immune system characterizes the pathogenesis of autoimmune diseases. A1AT treatment shows beneficial effects on patients and animal models with autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus. This review summarizes the functions and therapeutic prospects of A1AT in autoimmune diseases.
Collapse
Affiliation(s)
- Rui Sun
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiqiang Xu
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics and Department of General Practice, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chenxi Zhu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Luis E Muñoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lunzhi Dai
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics and Department of General Practice, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
5
|
Mellati A, Lo Faro L, Dumbill R, Meertens P, Rozenberg K, Shaheed S, Snashall C, McGivern H, Ploeg R, Hunter J. Kidney Normothermic Machine Perfusion Can Be Used as a Preservation Technique and a Model of Reperfusion to Deliver Novel Therapies and Assess Inflammation and Immune Activation. Front Immunol 2022; 13:850271. [PMID: 35720316 PMCID: PMC9198253 DOI: 10.3389/fimmu.2022.850271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Ischaemia-reperfusion injury (IRI) is an inevitable process in transplantation and results in inflammation and immune system activation. Alpha-1 antitrypsin (AAT) has anti-inflammatory properties. Normothermic machine perfusion (NMP) can be used to deliver therapies and may help in assessing the effects of IRI and immunity. This study investigated the effects of AAT on IRI and inflammation in pig kidneys when administered during preservation, followed by normothermic reperfusion (NR) with autologous whole blood, as a surrogate for transplant. Two different models were used to deliver AAT or placebo to paired slaughterhouse pig kidneys: Model 1: 7-h static cold storage (SCS) + 3-h NR (n = 5 pairs), where either AAT (10 mg/ml) or placebo was delivered in the flush following retrieval; Model 2: 4-h SCS + 3-h NMP + 3-h NR (n = 5 pairs), where either AAT or placebo was delivered during NMP. Injury markers and cytokines levels were analysed in the perfusate, and heat shock protein 70 KDa (HSP-70) was analysed in biopsies. AAT delivered to kidneys showed no adverse effects on perfusion parameters. HSP-70 fold changes were significantly lower in the AAT group during NMP (P < 0.01, paired t-test) but not during NR. Interleukin-1 receptor antagonist (IL-1ra) fold changes were significantly higher in the AAT group during NR model 1 (p < 0.05, two-way ANOVA). In contrast to the AAT group, significant upregulation of interleukin-1 beta (IL-1β) and interleukin-6 (IL-6) between t = 90 min and t = 180 min and interleukin-8 (IL-8) between baseline and t = 90 min was observed in the control group in NR model 2 (p < 0.05, Tukey's multiple comparison test). However, overall inflammatory cytokines and injury markers showed similar levels between groups. Delivery of AAT to pig kidneys was safe without any detrimental effects. NMP and NR provided excellent methods for comparison of inflammation and immune activation in the delivery of a novel therapy.
Collapse
Affiliation(s)
- Azita Mellati
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Letizia Lo Faro
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Richard Dumbill
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Pommelien Meertens
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Leiden University Medical Centre, Leiden University, Leiden, Netherlands
| | - Kaithlyn Rozenberg
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Sadr Shaheed
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Corinna Snashall
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Hannah McGivern
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Rutger Ploeg
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Leiden University Medical Centre, Leiden University, Leiden, Netherlands
- Oxford University Hospital National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - James Hunter
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- University Hospitals of Coventry and Warwickshire National Health Service (NHS) Trust, Coventry, United Kingdom
| |
Collapse
|
6
|
van Bilsen JHM, van den Brink W, van den Hoek AM, Dulos R, Caspers MPM, Kleemann R, Wopereis S, Verschuren L. Mechanism-Based Biomarker Prediction for Low-Grade Inflammation in Liver and Adipose Tissue. Front Physiol 2021; 12:703370. [PMID: 34858196 PMCID: PMC8631400 DOI: 10.3389/fphys.2021.703370] [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: 04/30/2021] [Accepted: 10/18/2021] [Indexed: 01/12/2023] Open
Abstract
Metabolic disorders, such as obesity and type 2 diabetes have a large impact on global health, especially in industrialized countries. Tissue-specific chronic low-grade inflammation is a key contributor to complications in metabolic disorders. To support therapeutic approaches to these complications, it is crucial to gain a deeper understanding of the inflammatory dynamics and to monitor them on the individual level. To this end, blood-based biomarkers reflecting the tissue-specific inflammatory dynamics would be of great value. Here, we describe an in silico approach to select candidate biomarkers for tissue-specific inflammation by using a priori mechanistic knowledge from pathways and tissue-derived molecules. The workflow resulted in a list of candidate markers, in part consisting of literature confirmed biomarkers as well as a set of novel, more innovative biomarkers that reflect inflammation in the liver and adipose tissue. The first step of biomarker verification was on murine tissue gene-level by inducing hepatic inflammation and adipose tissue inflammation through a high-fat diet. Our data showed that in silico predicted hepatic markers had a strong correlation to hepatic inflammation in the absence of a relation to adipose tissue inflammation, while others had a strong correlation to adipose tissue inflammation in the absence of a relation to liver inflammation. Secondly, we evaluated the human translational value by performing a curation step in the literature using studies that describe the regulation of the markers in human, which identified 9 hepatic (such as Serum Amyloid A, Haptoglobin, and Interleukin 18 Binding Protein) and 2 adipose (Resistin and MMP-9) inflammatory biomarkers at the highest level of confirmation. Here, we identified and pre-clinically verified a set of in silico predicted biomarkers for liver and adipose tissue inflammation which can be of great value to study future development of therapeutic/lifestyle interventions to combat metabolic inflammatory complications.
Collapse
Affiliation(s)
- Jolanda H M van Bilsen
- Department of Risk Assessment for Products in Development, The Netherlands Organization for Applied Scientific Research (TNO), Utrecht, Netherlands
| | - Willem van den Brink
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, Netherlands
| | - Anita M van den Hoek
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Remon Dulos
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, Netherlands
| | - Martien P M Caspers
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Suzan Wopereis
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, Netherlands
| |
Collapse
|
7
|
Nakagiri T, Wrenger S, Sivaraman K, Ius F, Goecke T, Zardo P, Grau V, Welte T, Haverich A, Knöfel AK, Janciauskiene S. α1-Antitrypsin attenuates acute rejection of orthotopic murine lung allografts. Respir Res 2021; 22:295. [PMID: 34789247 PMCID: PMC8597316 DOI: 10.1186/s12931-021-01890-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/08/2021] [Indexed: 01/04/2023] Open
Abstract
Background α1-Antitrypsin (AAT) is an acute phase glycoprotein, a multifunctional protein with proteinase inhibitory, anti-inflammatory and cytoprotective properties. Both preclinical and clinical experiences show that the therapy with plasma purified AAT is beneficial for a broad spectrum of inflammatory conditions. The potential effects of AAT therapy have recently been highlighted in lung transplantation (LuTx) as well. Methods We used a murine fully mismatched orthotopic single LuTx model (BALB/CJ as donors and C57BL/6 as recipients). Human AAT preparations (5 mg, n = 10) or vehicle (n = 5) were injected to the recipients subcutaneously prior to and intraperitoneally immediately after the LuTx. No immune suppressive drugs were administered. Three days after the transplantation, the mice were sacrificed, and biological samples were assessed. Results Histological analysis revealed significantly more severe acute rejection in the transplanted lungs of controls than in AAT treated mice (p < 0.05). The proportion of neutrophil granulocytes, B cells and the total T helper cell populations did not differ between two groups. There was no significant difference in serum CXCL1 (KC) levels. However, when compared to controls, human AAT was detectable in the serum of mice treated with AAT and these mice had a higher serum anti-elastase activity, and significantly lower proportion of Th1 and Th17 among all Th cells. Cleaved caspase-3-positive cells were scarce but significantly less abundant in allografts from recipients treated with AAT as compared to those treated with vehicle. Conclusion Therapy with AAT suppresses the acute rejection after LuTx in a mouse model. The beneficial effects seem to involve anti-protease and immunomodulatory activities of AAT.
Collapse
Affiliation(s)
- Tomoyuki Nakagiri
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Sabine Wrenger
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | | | - Fabio Ius
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Tobias Goecke
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Patrick Zardo
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Veronika Grau
- Department of General and Thoracic Surgery, Laboratory of Experimental Surgery, Justus-Liebig-University Giessen, German Center for Lung Research, Giessen, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Axel Haverich
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Ann-Kathrin Knöfel
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany. .,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.
| |
Collapse
|
8
|
Aust AC, Benesova E, Vidova V, Coufalikova K, Smetanova S, Borek I, Janku P, Budinska E, Klanova J, Thon V, Spacil Z. Profiling Tryptophan Catabolites of Human Gut Microbiota and Acute-Phase Protein Levels in Neonatal Dried Blood Specimens. Front Microbiol 2021; 12:665743. [PMID: 34777268 PMCID: PMC8581761 DOI: 10.3389/fmicb.2021.665743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 09/16/2021] [Indexed: 11/21/2022] Open
Abstract
National screening programs use dried blood specimens to detect metabolic disorders or aberrant protein functions that are not clinically evident in the neonatal period. Similarly, gut microbiota metabolites and immunological acute-phase proteins may reveal latent immune aberrations. Microbial metabolites interact with xenobiotic receptors (i.e., aryl hydrocarbon and pregnane-X) to maintain gastrointestinal tissue health, supported by acute-phase proteins, functioning as sensors of microbial immunomodulation and homeostasis. The delivery (vaginal or cesarean section) shapes the microbial colonization, which substantially modulates both the immune system’s response and mucosal homeostasis. This study profiled microbial metabolites of the kynurenine and tryptophan pathway and acute-phase proteins in 134 neonatal dried blood specimens. We newly established neonatal blood levels of microbial xenobiotic receptors ligands (i.e., indole-3-aldehyde, indole-3-butyric acid, and indole-3-acetamide) on the second day of life. Furthermore, we observed diverse microbial metabolic profiles in neonates born vaginally and via cesarean section, potentially due to microbial immunomodulatory influence. In summary, these findings suggest the supportive role of human gut microbiota in developing and maintaining immune system homeostasis.
Collapse
Affiliation(s)
| | - Eliska Benesova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Veronika Vidova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | | | - Sona Smetanova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Ivo Borek
- Department of Pediatrics, University Hospital Brno and Masaryk University Medical School, Brno, Czechia
| | - Petr Janku
- Department of Gynecology and Obstetrics, University Hospital Brno and Masaryk University Medical School, Brno, Czechia
| | - Eva Budinska
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Jana Klanova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Vojtech Thon
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Zdenek Spacil
- RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| |
Collapse
|
9
|
Saren G, Wong A, Lu YB, Baciu C, Zhou W, Zamel R, Soltanieh S, Sugihara J, Liu M. Ischemia-Reperfusion Injury in a Simulated Lung Transplant Setting Differentially Regulates Transcriptomic Profiles between Human Lung Endothelial and Epithelial Cells. Cells 2021; 10:cells10102713. [PMID: 34685693 PMCID: PMC8534993 DOI: 10.3390/cells10102713] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/01/2021] [Accepted: 10/07/2021] [Indexed: 11/30/2022] Open
Abstract
Current understanding of mechanisms of ischemia-reperfusion-induced lung injury during lung preservation and transplantation is mainly based on clinical observations and animal studies. Herein, we used cell and systems biology approaches to explore these mechanisms at transcriptomics levels, especially by focusing on the differences between human lung endothelial and epithelial cells, which are crucial for maintaining essential lung structure and function. Human pulmonary microvascular endothelial cells and human lung epithelial cells were cultured to confluent, subjected to different cold ischemic times (CIT) to mimic static cold storage with preservation solution, and then subjected to warm reperfusion with a serum containing culture medium to simulate lung transplantation. Cell morphology, viability, and transcriptomic profiles were studied. Ischemia-reperfusion injury induced a CIT time-dependent cell death, which was associated with dramatic changes in gene expression. Under normal control conditions, endothelial cells showed gene clusters enriched in the vascular process and inflammation, while epithelial cells showed gene clusters enriched in protein biosynthesis and metabolism. CIT 6 h alone or after reperfusion had little effect on these phenotypic characteristics. After CIT 18 h, protein-biosynthesis-related gene clusters disappeared in epithelial cells; after reperfusion, metabolism-related gene clusters in epithelial cells and multiple gene clusters in the endothelial cells also disappeared. Human pulmonary endothelial and epithelial cells have distinct phenotypic transcriptomic signatures. Severe cellular injury reduces these gene expression signatures in a cell-type-dependent manner. Therapeutics that preserve these transcriptomic signatures may represent new treatment to prevent acute lung injury during lung transplantation.
Collapse
Affiliation(s)
- Gaowa Saren
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (G.S.); (A.W.); (Y.-B.L.); (C.B.); (W.Z.); (R.Z.); (S.S.); (J.S.)
| | - Aaron Wong
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (G.S.); (A.W.); (Y.-B.L.); (C.B.); (W.Z.); (R.Z.); (S.S.); (J.S.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1X8, Canada
| | - Yun-Bi Lu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (G.S.); (A.W.); (Y.-B.L.); (C.B.); (W.Z.); (R.Z.); (S.S.); (J.S.)
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Cristina Baciu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (G.S.); (A.W.); (Y.-B.L.); (C.B.); (W.Z.); (R.Z.); (S.S.); (J.S.)
| | - Wenyong Zhou
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (G.S.); (A.W.); (Y.-B.L.); (C.B.); (W.Z.); (R.Z.); (S.S.); (J.S.)
| | - Ricardo Zamel
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (G.S.); (A.W.); (Y.-B.L.); (C.B.); (W.Z.); (R.Z.); (S.S.); (J.S.)
| | - Sahar Soltanieh
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (G.S.); (A.W.); (Y.-B.L.); (C.B.); (W.Z.); (R.Z.); (S.S.); (J.S.)
| | - Junichi Sugihara
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (G.S.); (A.W.); (Y.-B.L.); (C.B.); (W.Z.); (R.Z.); (S.S.); (J.S.)
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (G.S.); (A.W.); (Y.-B.L.); (C.B.); (W.Z.); (R.Z.); (S.S.); (J.S.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1X8, Canada
- Department of Surgery, Medicine and Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1X8, Canada
- Correspondence:
| |
Collapse
|
10
|
Bianchera A, Alomari E, Bruno S. Augmentation therapy with alpha 1-antitrypsin: present and future of production, formulation, and delivery. Curr Med Chem 2021; 29:385-410. [PMID: 34036902 DOI: 10.2174/0929867328666210525161942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/24/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
Alpha 1-antitrypsin is one of the first protein therapeutics introduced on the market - more than 30 years ago - and, to date, it is indicated only for the treatment of the severe forms of a genetic condition known as alpha-1 antitrypsin deficiency. The only approved preparations are derived from plasma, posing potential problems associated with its limited supply and high processing costs. Moreover, augmentation therapy with alpha 1-antitrypsin is still limited to intravenous infusions, a cumbersome regimen for patients. Here, we review the recent literature on its possible future developments, focusing on i) the recombinant alternatives to the plasma-derived protein, ii) novel formulations, and iii) novel administration routes. Regulatory issues and the still unclear noncanonical functions of alpha 1-antitrypsin - possibly associated with the glycosylation pattern found only in the plasma-derived protein - have hindered the introduction of new products. However, potentially new therapeutic indications other than the treatment of alpha-1 antitrypsin deficiency might open the way to new sources and new formulations.
Collapse
Affiliation(s)
- Annalisa Bianchera
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| | - Esraa Alomari
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| | - Stefano Bruno
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| |
Collapse
|
11
|
Gou W, Wang J, Song L, Kim DS, Cui W, Strange C, Wang H. Alpha-1 antitrypsin suppresses macrophage activation and promotes islet graft survival after intrahepatic islet transplantation. Am J Transplant 2021; 21:1713-1724. [PMID: 33047509 PMCID: PMC8082666 DOI: 10.1111/ajt.16342] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/13/2020] [Accepted: 09/23/2020] [Indexed: 01/25/2023]
Abstract
Alpha-1 antitrypsin (AAT) has protective functions in animal islet transplantation models. While the therapeutic effect of AAT therapy is currently being tested in clinical trials, we investigated the mechanism of AAT protection in a clinically relevant marginal intrahepatic human islet transplantation model. In recipients receiving islets and AAT, 68.9% (20/29) reached normoglycemia, compared to 35.7% (10/28) in those receiving islets only, at 60 days posttransplant (PT). AAT-treated mice had lower serum levels of inflammatory cytokines immediately PT. Reduced M1 macrophages were observed in livers of AAT-treated recipients compared to controls as evidenced by flow cytometry and RNA-seq transcriptional profiling analysis. In vitro AAT suppressed IFN-γ-induced M1 macrophage activation/polarization via suppression of STAT1 phosphorylation and iNOS production. AAT inhibits macrophage activation induced by cytokines or dying islets, and consequently leads to islet cell survival. In a macrophage depletion mouse model, the presence of M1 macrophages in the liver contributed to graft death. AAT, through suppressing macrophage activation, protected transplanted islets from death and dysfunction in the human islet and NOD-SCID mouse model. The protective effect of AAT was confirmed in a major mismatch allogeneic islet transplantation model. Taken together, AAT suppresses liver macrophage activation that contributes to graft survival after transplantation.
Collapse
Affiliation(s)
- Wenyu Gou
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Jingjing Wang
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Lili Song
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Do-Sung Kim
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Wanxing Cui
- MedStar Georgetown University, Washington, District of Columbia
| | - Charlie Strange
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
12
|
Ataya A. Recurrence of emphysema post-lung transplantation in a patient with alpha 1 antitrypsin deficiency (AATD). Respir Med Case Rep 2020; 31:101309. [PMID: 33304809 PMCID: PMC7718162 DOI: 10.1016/j.rmcr.2020.101309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/13/2020] [Accepted: 11/22/2020] [Indexed: 11/17/2022] Open
Abstract
The genetic disorder alpha 1 antitrypsin deficiency (AATD) results in reduced levels of alpha 1 antitrypsin (AAT) in the lung and an imbalance between AAT anti-protease activity and the activity of proteases that degrade elastin and connective tissues. This imbalance commonly leads to the excessive proteolysis of structural tissue of the alveoli, causing chronic obstructive pulmonary disease (COPD)/emphysema. While patients with AATD are encouraged to make lifestyle changes, including stopping smoking, and can be treated with alpha 1 antitrypsin therapy (AAT therapy) to slow progression of COPD/emphysema, damage to the lungs is irreparable, and therefore, lung transplantation is required in severe cases. However, following lung transplant, the genetic cause of AATD-related COPD/emphysema remains, and patients may continue to be at risk of redeveloping COPD/emphysema. Recurrence of COPD/emphysema was observed in a patient with AATD 2 years after initial successful lung transplantation and cessation of AAT therapy who recommenced smoking after no signs of disease at the 1-year assessment. This case demonstrates that smoking cessation is critical in patients with AATD, even after lung transplant, and it highlights that patients with AATD may benefit from AAT therapy post-lung transplant. Patients with AATD can redevelop emphysema following lung transplantation. Abstinence from smoking remains crucial in all patients with AATD. AAT therapy may be required in patients who redevelop emphysema post-lung transplant.
Collapse
Affiliation(s)
- Ali Ataya
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida Health, Gainesville, FL, 32610-0225, USA
| |
Collapse
|
13
|
Lim JH, Oh EJ, Oh SH, Jung HY, Choi JY, Cho JH, Park SH, Kim YL, Kim CD. Renoprotective Effects of Alpha-1 Antitrypsin against Tacrolimus-Induced Renal Injury. Int J Mol Sci 2020; 21:ijms21228628. [PMID: 33207690 PMCID: PMC7696546 DOI: 10.3390/ijms21228628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/09/2020] [Accepted: 11/13/2020] [Indexed: 11/16/2022] Open
Abstract
The protective effects of alpha-1 antitrypsin (AAT) in tacrolimus (TAC)-induced renal injury was evaluated in a rat model. The TAC group rats were subcutaneously injected with 2 mg/kg TAC every day for four weeks. The TAC with AAT group was cotreated with daily subcutaneous injections of TAC and intraperitoneal injections of AAT (80 mg/kg) for four weeks. The effects of AAT on TAC-induced renal injury were evaluated using serum biochemistry, histopathology, and Western blotting. The TAC injection significantly increased renal interstitial fibrosis, inflammation, and apoptosis as compared to the control treatment. The histopathological examination showed that cotreatment of TAC and AAT attenuated interstitial fibrosis (collagen, fibronectin, and α-SMA staining), and α-SMA expression in Western blotting was also decreased. Immunohistochemical staining for inflammation (osteopontin and ED-1 staining) revealed improved interstitial inflammation in the TAC with AAT group compared to that in the TAC group. The TAC treatment increased renal apoptosis compared to the control treatment, based on the results of increased immunohistochemical staining of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL), increased caspase-3 activity, and lower Bcl-2 to Bad expression ratio. However, AAT cotreatment significantly changed these markers and consequently showed decreased apoptosis. AAT protects against TAC-induced renal injury via antifibrotic, anti-inflammatory, and antiapoptotic effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Chan-Duck Kim
- Correspondence: ; Tel.: +82-53-200-5560; Fax: +82-53-426-2046
| |
Collapse
|
14
|
Wong A, Zamel R, Yeung J, Bader GD, Dos Santos CC, Bai X, Wang Y, Keshavjee S, Liu M. Potential therapeutic targets for lung repair during human ex vivo lung perfusion. Eur Respir J 2020; 55:13993003.02222-2019. [DOI: 10.1183/13993003.02222-2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 01/15/2020] [Indexed: 12/20/2022]
Abstract
IntroductionThe ex vivo lung perfusion (EVLP) technique has been developed to assess the function of marginal donor lungs and has significantly increased donor lung utilisation. EVLP has also been explored as a platform for donor lung repair through injury-specific treatments such as antibiotics or fibrinolytics. We hypothesised that actively expressed pathways shared between transplantation and EVLP may reveal common mechanisms of injury and potential therapeutic targets for lung repair prior to transplantation.Materials and methodsRetrospective transcriptomics analyses were performed with peripheral tissue biopsies from “donation after brain death” lungs, with 46 pre-/post-transplant pairs and 49 pre-/post-EVLP pairs. Pathway analysis was used to identify and compare the responses of donor lungs to transplantation and to EVLP.Results22 pathways were enriched predominantly in transplantation, including upregulation of lymphocyte activation and cell death and downregulation of metabolism. Eight pathways were enriched predominantly in EVLP, including downregulation of leukocyte functions and upregulation of vascular processes. 27 pathways were commonly enriched, including activation of innate inflammation, cell death, heat stress and downregulation of metabolism and protein synthesis. Of the inflammatory clusters, Toll-like receptor/innate immune signal transduction adaptor signalling had the greatest number of nodes and was central to inflammation. These mechanisms have been previously speculated as major mechanisms of acute lung injury in animal models.ConclusionEVLP and transplantation share common molecular features of injury including innate inflammation and cell death. Blocking these pathways during EVLP may allow for lung repair prior to transplantation.
Collapse
|
15
|
Zamora M. Surgery for patients with Alpha 1 Antitrypsin Deficiency: A review. Am J Surg 2019; 218:639-647. [DOI: 10.1016/j.amjsurg.2018.10.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/12/2018] [Indexed: 12/01/2022]
|
16
|
Schuster R, Bar-Nathan O, Tiosano A, Lewis EC, Silberstein E. Enhanced Survival and Accelerated Perfusion of Skin Flap to Recipient Site Following Administration of Human α1-Antitrypsin in Murine Models. Adv Wound Care (New Rochelle) 2019; 8:281-290. [PMID: 31737418 DOI: 10.1089/wound.2018.0889] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/23/2018] [Indexed: 12/18/2022] Open
Abstract
Objective: Skin flaps are routinely used in reconstructive surgery yet remain susceptible to ischemia and necrosis. Distant flaps require lengthy time to detach causing patient discomfort. Human α1-antitrypsin (hAAT) is a clinically available serum glycoprotein. hAAT was shown to support mature vessel formation and enhance tissue survival following ischemia-reperfusion injuries. The purpose of the presented study was to examine the effect of hAAT on skin flap survival and distant "tube" flap perfusion through its recipient site. Approach: Random-pattern skin flaps were performed on mice treated with clinical-grade hAAT using three unique routes of administration (transgenic, i.p. and s.c. infiltration); necrotic area and tissue perfusion were assessed. Blockade of vascular endothelial growth factor (VEGF) and nitric oxide synthase (NOS) were used to explore aspects of mechanism of action. A distant tube flap model was performed to examine time to perfusion. Results: hAAT-treated mice displayed approximately two-fold smaller necrotic flap areas versus controls across all hAAT administration routes. Flaps displayed greater perfusion as early as 3 days postsurgery (64.6% ± 4.0% vs. 43.7% ± 1.7% in controls; p = 0.007). hAAT-mediated flap survival was prominently NOS dependent, but only partially VEGF dependent. Finally, distant flaps treated with hAAT displayed significantly earlier perfusion versus controls (mean 9.6 ± 1.6 vs. 13.1 ± 1.0 days; p = 0.0005). Innovation: The established safety record of hAAT renders it an attractive candidate toward improving skin flap surgery outcomes, particularly during VEGF blockade. Conclusions: hAAT treatment enhances survival and accelerates perfusion of skin flaps in animal models in a NOS-dependent manner, partially circumventing VEGF blockade. Further mechanistic studies are required.
Collapse
Affiliation(s)
- Ronen Schuster
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Or Bar-Nathan
- Department of Plastic and Reconstructive Surgery, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Tiosano
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eldad Silberstein
- Department of Plastic and Reconstructive Surgery, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
17
|
Alpha-1 Antitrypsin Attenuates Acute Lung Allograft Injury in a Rat Lung Transplant Model. Transplant Direct 2019; 5:e458. [PMID: 31723592 PMCID: PMC6791593 DOI: 10.1097/txd.0000000000000898] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/15/2019] [Accepted: 03/22/2019] [Indexed: 11/27/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) after lung transplantation triggers a cascade of inflammatory changes that can contribute to acute allograft injury. This influences both the short- and long-term survival of the lung allograft. Alpha-1 antitrypsin (AAT) is a protease inhibitor with known anti-inflammatory and immune-regulatory properties that mitigate tissue damage. This study explores the protective effects of AAT in the setting of IRI utilizing a rat lung transplant model. Methods Orthotopic left single lung transplantation was performed from Lewis to Sprague-Dawley rats; recipients did not receive systemic immunosuppression. Before transplantation, the donor lungs were primed with either albumin (control) or AAT. Starting the day of transplantation, recipient rats also received either albumin (control) or AAT with subsequent doses administered over the next 7 days. On the eighth postoperative day, lung allografts were recovered and analyzed. Results Degree of inflammatory infiltrate, as quantified by the allograft weight (g)/body weight (kg) ratio, was significantly reduced in the AAT-treated group compared with controls (3.5 vs 7.7, respectively, P < 0.05). Treatment with AAT also significantly decreased allograft necrosis in treated animals, as measured by a semiquantitative score that ranged from 0 to 4 (1.25 vs 4, P < 0.05). In addition, lymphocytes isolated from recipients treatment group showed significant proliferative inhibition via a mixed lymphocyte response assay in response to donor antigens. Conclusions AAT attenuates acute allograft injury and necrosis in a rat model of lung transplantation, suggesting that AAT may play a role in reducing IRI-induced inflammation.
Collapse
|
18
|
Lee C, Dhawan A, Iansante V, Filippi C, Mitry R, Tang J, Walker S, Fernandez DaCosta R, Sinha S, Hughes RD, Koulmanda M, Fitzpatrick E. Improving engraftment of hepatocyte transplantation using alpha-1 antitrypsin as an immune modulator. J Mol Med (Berl) 2019; 97:563-577. [PMID: 30820592 PMCID: PMC6440943 DOI: 10.1007/s00109-019-01747-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 01/11/2023]
Abstract
Abstract For patients with non-cirrhotic liver-based metabolic disorders, hepatocyte transplantation can be an effective treatment. However, long-term function of transplanted hepatocytes following infusion has not been achieved due to insufficient numbers of hepatocytes reaching the liver cell plates caused by activation of the instant blood-mediated inflammatory reaction (IBMIR). Our aim was to determine if the natural immune modulator, alpha-1 antitrypsin (AAT), could improve engraftment of transplanted hepatocytes and investigate its mechanism of action. A tubing loop model was used to analyse activation of the IBMIR when human hepatocytes were in contact with ABO-matched blood and 4 mg/ml AAT. Platelet and white cell counts, complement and cytokine expression were analysed. To determine if AAT could improve short-term engraftment, female rats underwent tail vein injection of AAT (120 mg/kg) or water (control) prior to the intrasplenic transplantation of 2 × 107 male hepatocytes. At 48 h and 1 week, livers were collected for analysis. In our loop model, human hepatocytes elicited a significant drop in platelet count with thrombus formation compared to controls. Loops containing AAT and hepatocytes showed no platelet consumption and no thrombus formation. Further, AAT treatment resulted in reduced IL-1β, IL-6 and IFN-γ and increased IL-1RA compared to untreated loops. In vivo, AAT significantly improved engraftment of rat hepatocytes compared to untreated at 48 h. AAT infusion may inhibit the IBMIR, thus improving short-term engraftment of donor hepatocytes and potentially improve the outcomes for patients with liver-based metabolic disease. Key messages • Alpha-1 antitrypsin (AAT) acts as an immune modulator to improve the efficacy of hepatocyte transplantation. • Treatment with AAT decreased thrombus formation and pro-inflammatory cytokine expression in a tubing loop model. • AAT significantly improved engraftment of donor hepatocytes within the first 48 h post transplantation. Electronic supplementary material The online version of this article (10.1007/s00109-019-01747-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charlotte Lee
- Dhawan Group at Mowat Labs, Institute of Liver Studies, King's College London at King's College Hospital, London, UK
| | - Anil Dhawan
- Paediatric Liver, GI and Nutrition Centre, King's College London School of Medicine at King's College Hospital, Denmark Hill, London, UK.
| | - Valeria Iansante
- Dhawan Group at Mowat Labs, Institute of Liver Studies, King's College London at King's College Hospital, London, UK
| | - Celine Filippi
- Dhawan Group at Mowat Labs, Institute of Liver Studies, King's College London at King's College Hospital, London, UK
| | - Ragai Mitry
- Dhawan Group at Mowat Labs, Institute of Liver Studies, King's College London at King's College Hospital, London, UK
| | - Joanne Tang
- Dhawan Group at Mowat Labs, Institute of Liver Studies, King's College London at King's College Hospital, London, UK
| | - Simon Walker
- Paediatric Liver, GI and Nutrition Centre, King's College London School of Medicine at King's College Hospital, Denmark Hill, London, UK
| | - Raquel Fernandez DaCosta
- Paediatric Liver, GI and Nutrition Centre, King's College London School of Medicine at King's College Hospital, Denmark Hill, London, UK
| | - Siddharth Sinha
- Dhawan Group at Mowat Labs, Institute of Liver Studies, King's College London at King's College Hospital, London, UK
| | - Robin D Hughes
- Dhawan Group at Mowat Labs, Institute of Liver Studies, King's College London at King's College Hospital, London, UK
| | - Maria Koulmanda
- Departments of Medicine and Surgery, The Transplant Institute, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Emer Fitzpatrick
- Paediatric Liver, GI and Nutrition Centre, King's College London School of Medicine at King's College Hospital, Denmark Hill, London, UK.
| |
Collapse
|
19
|
Repetto O, Mussolin L, Elia C, Martina L, Bianchi M, Buffardi S, Sala A, Burnelli R, Mascarin M, De Re V. Proteomic Identification of Plasma Biomarkers in Children and Adolescents with Recurrent Hodgkin Lymphoma. J Cancer 2018; 9:4650-4658. [PMID: 30588249 PMCID: PMC6299395 DOI: 10.7150/jca.27560] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/26/2018] [Indexed: 12/13/2022] Open
Abstract
The treatment of paediatric Hodgkin lymphoma (HL) has steadily improved over the years, so that 10- years survival exceed 80%. The purpose of this study was to identify prognostic markers for relapsed HL that might contribute to optimize therapeutic approaches. To this aim we retrospectively analysed differential protein expression profiles obtained from plasma of children/adolescents with HL (age ranging from 10 to 18 years) collected at diagnosis. We examined the protein profiles of 15 HL relapsed (R) patients compared with 14 HL not relapsed (NR) patients treated with the same LH-2004 protocol. Two dimensional difference in gel electrophoresis (2D-DIGE) revealed significant differences (fold change > 1.5; Student's T-test p<0.01) between R and NR patients in 10 proteins: α-1-antitrypsin chain a, apolipoprotein A-IV precursor; inter-α-trypsin inhibitor heavy chain; antithrombin-III; vitronectin; fibrinogen α, β and γ chains, complement C3, and ceruloplasmin. An up-regulation of fibrinogen α (spots 78, 196, 230, 234, 239) and β (spots 98, 291, 296, 300) chains together with a lower level of α-1-antitrypsin (spots 255, 264, 266, 272, 273) were found in R patients, and this difference was validated by immunoblotting. The functional role(s) of these proteins in the coagulation and inflammation associated with paediatric/adolescent HL progression and relapse deserves further investigations.
Collapse
Affiliation(s)
- Ombretta Repetto
- Facility of Bio-Proteomics, Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Lara Mussolin
- Clinic of Pediatric Haemato-Oncology, Department of Women's and Children's Health, University of Padua, Padua, Institute of Paediatric Research - Fondazione Città della Speranza, Padua, Italy
| | - Caterina Elia
- Pediatric Radioterapy Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Lia Martina
- Facility of Bio-Proteomics, Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Maurizio Bianchi
- Pediatric Onco-Hematology and Stem Cell Transplant Division, City of Health and Science, Regina Margherita Children's Hospital, Turin, Italy
| | - Salvatore Buffardi
- Paediatric Haemato-Oncology department, Santobono-Pausilipon Children's Hospital, Napoli, Italy
| | - Alessandra Sala
- Department of Paediatrics, Ospedale San Gerardo, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Roberta Burnelli
- Pediatric Oncology University Hospital, Sant'Anna Hospital, Ferrara, Italy
| | - Maurizio Mascarin
- Pediatric Radioterapy Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Valli De Re
- Facility of Bio-Proteomics, Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| |
Collapse
|
20
|
Huang L, Yang C, Liu M. Intracellular signal transduction pathways as potential drug targets for ischemia-reperfusion injury in lung transplantation. J Thorac Dis 2018; 10:S3965-S3969. [PMID: 30631528 DOI: 10.21037/jtd.2018.09.130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Lei Huang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, Departments of Surgery, Medicine and Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Chengliang Yang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, Departments of Surgery, Medicine and Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, Departments of Surgery, Medicine and Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
21
|
Korniotis S, Thornley TB, Kyriazis P, Theodorou E, Ma L, Li LS, Kokkotou E, Strom TB, Koulmanda M. Hematopoietic Stem/Progenitor Cell Dependent Participation of Innate Lymphoid Cells in Low-Intensity Sterile Inflammation. Front Immunol 2018; 9:2007. [PMID: 30233592 PMCID: PMC6134892 DOI: 10.3389/fimmu.2018.02007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/14/2018] [Indexed: 11/13/2022] Open
Abstract
Hematopoietic stem/progenitor cells (HSPC) are characterized by their unique capacities of self-renewal and multi-differentiation potential. This second property makes them able to adapt their differentiation profile depending on the local environment they reach. Taking advantage of an animal model of peritonitis, induced by injection of the TLR-2 ligand, zymosan, we sought to study the relationship between bone marrow-derived hematopoietic stem/progenitor cells (BM-HSPCs) and innate lymphoid cells (ILCs) regarding their emergence and differentiation at the site of inflammation. Our results demonstrate that the strength of the inflammatory signals affects the capacity of BM-derived HSPCs to migrate and give rise in situ to ILCs. Both low- and high-dose of zymosan injections trigger the appearance of mature ILCs in the peritoneal cavity where the inflammation occurs. Herein, we show that only in low-dose injected mice, the recovered ILCs are dependent on an in situ differentiation of BM-derived HSPCs and/or ILC2 precursors (ILC2P) wherein high-dose, the stronger inflammatory environment seems to be able to induce the emergence of ILCs independently of BM-derived HSPCs. We suggest that a relationship between HSPCs and ILCs seems to be affected by the strength of the inflammatory stimuli opening new perspectives in the manipulation of these early hematopoietic cells.
Collapse
Affiliation(s)
- Sarantis Korniotis
- Department of Medicine, Harvard Medical School and the Transplant Institute at Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Thomas B. Thornley
- Department of Medicine, Harvard Medical School and the Transplant Institute at Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Periklis Kyriazis
- Department of Medicine, Harvard Medical School and the Transplant Institute at Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Evangelos Theodorou
- Department of Medicine, Harvard Medical School and the Transplant Institute at Beth Israel Deaconess Medical Center, Boston, MA, United States
- Division of Gastroenterology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Lingzhi Ma
- Department of Medicine, Harvard Medical School and the Transplant Institute at Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Lisa S. Li
- Department of Surgery, Harvard Medical School and the Transplant Institute at Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Efi Kokkotou
- Department of Medicine, Harvard Medical School and the Transplant Institute at Beth Israel Deaconess Medical Center, Boston, MA, United States
- Division of Gastroenterology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Terry B. Strom
- Department of Medicine, Harvard Medical School and the Transplant Institute at Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of Surgery, Harvard Medical School and the Transplant Institute at Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Maria Koulmanda
- Department of Medicine, Harvard Medical School and the Transplant Institute at Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of Surgery, Harvard Medical School and the Transplant Institute at Beth Israel Deaconess Medical Center, Boston, MA, United States
| |
Collapse
|
22
|
Berger M, Liu M, Uknis ME, Koulmanda M. Alpha-1-antitrypsin in cell and organ transplantation. Am J Transplant 2018; 18:1589-1595. [PMID: 29607607 PMCID: PMC6055806 DOI: 10.1111/ajt.14756] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/14/2018] [Accepted: 03/13/2018] [Indexed: 01/25/2023]
Abstract
Limited availability of donor organs and risk of ischemia-reperfusion injury (IRI) seriously restrict organ transplantation. Therapeutics that can prevent or reduce IRI could potentially increase the number of transplants by increasing use of borderline organs and decreasing discards. Alpha-1 antitrypsin (AAT) is an acute phase reactant and serine protease inhibitor that limits inflammatory tissue damage. Purified plasma-derived AAT has been well tolerated in more than 30 years of use to prevent emphysema in AAT-deficient individuals. Accumulating evidence suggests that AAT has additional anti-inflammatory and tissue-protective effects including improving mitochondrial membrane stability, inhibiting apoptosis, inhibiting nuclear factor kappa B activation, modulating pro- vs anti-inflammatory cytokine balance, and promoting immunologic tolerance. Cell culture and animal studies have shown that AAT limits tissue injury and promotes cell and tissue survival. AAT can promote tolerance in animal models by downregulating early inflammation and favoring induction and stabilization of regulatory T cells. The diverse intracellular and immune-modulatory effects of AAT and its well-established tolerability in patients suggest that it might be useful in transplantation. Clinical trials, planned and/or in progress, should help determine whether the promise of the animal and cellular studies will be fulfilled by improving outcomes in human organ transplantation.
Collapse
Affiliation(s)
| | | | | | - Maria Koulmanda
- Transplant Institute at Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMAUSA
| |
Collapse
|