1
|
Zhang S, Zhao X, Lv Y, Niu J, Wei X, Luo Z, Wang X, Chen XL. Exosomes of different cellular origins: prospects and challenges in the treatment of acute lung injury after burns. J Mater Chem B 2025; 13:1531-1547. [PMID: 39704476 DOI: 10.1039/d4tb02351j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Acute lung injury (ALI) is a critical clinical disease caused by direct factors (inhalation injury, gastroesophageal reflux, etc.) or indirect factors (including infection, sepsis, burn, shock, trauma, acute pancreatitis, fat embolism, drug overdose, etc.). ALI is characterized mainly by diffuse interstitial and alveolar edema caused by an uncontrolled inflammatory response and damage to the alveoli-capillary barrier and has very high morbidity and mortality rates. Currently, there is no effective treatment strategy other than mechanical ventilation, fluid management or other supportive treatments. Exosomes are nanovesicle-like vesicles with double-membrane structures detached from the cell membrane or secreted by cells. These vesicles can be used as drug carriers because of their unique biological properties, such as anti-inflammatory, anti-apoptotic, pro-cell growth and immunomodulatory functions, and have been applied in the treatment of ALI in recent years. In this study, the mechanism and pathophysiological characteristics of ALI were first systematically described. The different cellular sources and characteristics of exosomes are summarized, and their functions and value as drug carriers in the treatment of ALI are discussed, as are the challenges that may be faced in the treatment of ALI with exosomes.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Xinyu Zhao
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Yang Lv
- Plastic Surgery Department, The Second Affiliated Hospital of Anhui Medical University, 230061, P. R. China
| | - Jianguo Niu
- School of Biomedical Engineering, Anhui Medical University, Hefei 230022, China.
| | - Xiaolong Wei
- School of Biomedical Engineering, Anhui Medical University, Hefei 230022, China.
| | - Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P. R. China.
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230022, China.
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
2
|
Lotter CR, Sherratt JA. Pulmonary epithelial wound healing and the immune system. Mathematical modeling and bifurcation analysis of a bistable system. J Theor Biol 2025; 596:111968. [PMID: 39455020 DOI: 10.1016/j.jtbi.2024.111968] [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: 06/18/2024] [Revised: 09/15/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Respiratory diseases such as asthma, acute respiratory distress syndrome (ARDS), influenza or COVID-19 often directly target the epithelium. Elevated immune levels and a 'cytokine storm' are directly associated with defective healing dynamics of lung diseases such as COVID-19 or ARDS. The infected cells leave wounded regions in the epithelium which must be healed for the lung to return to a healthy state and carry out its main function of gas-exchange. Due to the complexity of the various interactions between cells of the lung epithelium and surrounding tissue, it is necessary to develop models that can complement experiments to fully understand the healing dynamics. In this mathematical study we model the mechanism of epithelial regeneration. We assume that healing is exclusively driven by progenitor cell proliferation, induced by a chemical activator such as epithelial growth factor (EGF) and cytokines such as interleukin-22 (IL22). Contrary to previous studies of wound healing, we consider the immune system, specifically the T effector cells TH1, TH17, TH22 and Treg to strongly contribute to the healing process, by producing IL22 or regulating the immune response. We therefore obtain a coupled system of two ordinary differential equations for the epithelial and immune cell densities and two functions for the levels of chemicals that either induce epithelial proliferation or recruit immune cells. These functions link the two cell equations. We find that to allow the epithelium to regenerate to a healthy state, the immune system must not exceed a threshold value at the onset of the healing phase. This immune threshold is supported experimentally but was not explicitly built into our equations. Our assumptions are therefore sufficient to reproduce experimental results concerning the ratio TH17/Treg cells as a threshold to predict higher mortality rates in patients. This immune threshold can be controlled by parameters of the model, specifically the base-level growth factor concentration. This conclusion is based on a mathematical bifurcation analysis and linearization of the model equations. Our results suggest treatment of severe cases of lung injury by reducing or suppressing the immune response, in an individual patient, assessed by their disease parameters such as course of lung injury and immune response levels.
Collapse
Affiliation(s)
- Clara R Lotter
- Department of Mathematics, Heriot-Watt University and Maxwell Institute for Mathematical Sciences, Edinburgh, EH14 4AS, UK.
| | - Jonathan A Sherratt
- Department of Mathematics, Heriot-Watt University and Maxwell Institute for Mathematical Sciences, Edinburgh, EH14 4AS, UK
| |
Collapse
|
3
|
Huang H, Shi Y, Zhou Y. The Protective Effects of Annexin A1 in Acute Lung Injury Mediated by Nrf2. Immun Inflamm Dis 2025; 13:e70111. [PMID: 39807748 PMCID: PMC11729740 DOI: 10.1002/iid3.70111] [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: 04/02/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Acute lung injury (ALI), one of the most severe respiratory system diseases, is prevalent worldwide. Annexin A1 (AnxA1) is an important member of the annexin superfamily, known for its wide range of physiological functions. However, its potential protective effect against lipopolysaccharide (LPS)-induced ALI remains unclear. MATERIALS AND METHODS Mice were divided into four groups: Sham, LPS + vehicle, LPS + 0.1 μg AnxA1, and LPS + 0.5 μg AnxA1. Lung injury was assessed through histopathology, pulmonary wet-to-dry (W/D) ratio, cell counting of bronchoalveolar lavage fluid (BALF), oxidative stress analysis, and noninvasive pulmonary function testing. Gene and protein expression levels were measured using RT-PCR, ELISA, and western blot analysis. RESULTS AnxA1 alleviated LPS-induced ALI by protecting lung tissue from damage, reducing the lung wet/dry (W/D) weight ratio, and improving LPS-induced impaired lung function. Interestingly, administration of AnxA1 was found to repress the infiltration of inflammatory cells by decreasing the total cell count, neutrophils, and protein concentrations in bronchoalveolar lavage fluid (BALF). AnxA1 mitigated the inflammatory response in the pulmonary tissue by lowering the levels of IL-1β, IL-6, and TNF-α in BALF of ALI mice. Additionally, AnxA1 attenuated oxidative stress in lung tissues of ALI mice by restoring the activity of catalase (CAT), SOD, and glutathione (GSH) but reducing the levels of malondialdehyde (MDA). We also found that AnxA1 suppressed activation of the NLRP3 signaling pathway. Mechanistically, AnxA1 activated the Nrf2/HO-1 signaling pathway while preventing the activation of NF-κB. CONCLUSION Collectively, these findings suggest that AnxA1 alleviates LPS-induced ALI and might be a promising novel therapeutic agent against LPS-induced ALI.
Collapse
Affiliation(s)
- Hui Huang
- Department of StomatologyLiyuan Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Yuqin Shi
- Department of Respiratory and Critical Care MedicineLiyuan Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Yuequan Zhou
- Department of Respiratory and Critical Care MedicineLiyuan Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
4
|
Silva RCMC. The dichotomic role of cytokines in aging. Biogerontology 2024; 26:17. [PMID: 39621124 DOI: 10.1007/s10522-024-10152-4] [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: 09/04/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024]
Abstract
The chronic inflammation present in aged individuals is generally depicted as a detrimental player for longevity. Here, it is discussed several beneficial effects associated with the cytokines that are chronically elevated in inflammaging. These cytokines, such as IL-1β, type I interferons, IL-6 and TNF positively regulate macroautophagy, mitochondrial function, anti-tumor immune responses and skeletal muscle biogenesis, possibly contributing to longevity. On the other side, the detrimental and antagonistic role of these cytokines including the induction of sarcopenia, tissue damage and promotion of tumorigenesis are also discussed, underscoring the dichotomy associated with inflammaging and its players. In addition, it is discussed the role of the anti-inflammatory cytokine IL-10 and other cytokines that affect aging in a more linear way, such as IL-11, which promotes senescence, and IL-4 and IL-15, which promotes longevity. It is also discussed more specific regulators of aging that are downstream cytokines-mediated signaling.
Collapse
|
5
|
Ziaka M, Exadaktylos A. Gut-derived immune cells and the gut-lung axis in ARDS. Crit Care 2024; 28:220. [PMID: 38965622 PMCID: PMC11225303 DOI: 10.1186/s13054-024-05006-x] [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: 04/20/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024] Open
Abstract
The gut serves as a vital immunological organ orchestrating immune responses and influencing distant mucosal sites, notably the respiratory mucosa. It is increasingly recognized as a central driver of critical illnesses, with intestinal hyperpermeability facilitating bacterial translocation, systemic inflammation, and organ damage. The "gut-lung" axis emerges as a pivotal pathway, where gut-derived injurious factors trigger acute lung injury (ALI) through the systemic circulation. Direct and indirect effects of gut microbiota significantly impact immune responses. Dysbiosis, particularly intestinal dysbiosis, termed as an imbalance of microbial species and a reduction in microbial diversity within certain bodily microbiomes, influences adaptive immune responses, including differentiating T regulatory cells (Tregs) and T helper 17 (Th17) cells, which are critical in various lung inflammatory conditions. Additionally, gut and bone marrow immune cells impact pulmonary immune activity, underscoring the complex gut-lung interplay. Moreover, lung microbiota alterations are implicated in diverse gut pathologies, affecting local and systemic immune landscapes. Notably, lung dysbiosis can reciprocally influence gut microbiota composition, indicating bidirectional gut-lung communication. In this review, we investigate the pathophysiology of ALI/acute respiratory distress syndrome (ARDS), elucidating the role of immune cells in the gut-lung axis based on recent experimental and clinical research. This exploration aims to enhance understanding of ALI/ARDS pathogenesis and to underscore the significance of gut-lung interactions in respiratory diseases.
Collapse
Affiliation(s)
- Mairi Ziaka
- Clinic of Geriatric Medicine, Center of Geriatric Medicine and Rehabilitation, Kantonsspital Baselland, Bruderholz, Switzerland.
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland.
| | - Aristomenis Exadaktylos
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
6
|
Pan T, Lee JW. A crucial role of neutrophil extracellular traps in pulmonary infectious diseases. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:34-41. [PMID: 39170960 PMCID: PMC11332830 DOI: 10.1016/j.pccm.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Indexed: 08/23/2024]
Abstract
Neutrophil extracellular traps (NETs), extrusions of intracellular DNA with attached granular material that exert an antibacterial effect through entangling, isolating, and immobilizing microorganisms, have been extensively studied in recent decades. The primary role of NETs is to entrap and facilitate the killing of bacteria, fungi, viruses, and parasites, preventing bacterial and fungal dissemination. NET formation has been described in many pulmonary diseases, including both infectious and non-infectious. NETs are considered a double-edged sword. As innate immune cells, neutrophils release NETs to kill pathogens and remove cellular debris. However, the deleterious effects of excessive NET release in lung disease are particularly important because NETs and by-products of NETosis can directly induce epithelial and endothelial cell death while simultaneously inducing inflammatory cytokine secretion and immune-mediated thrombosis. Thus, NET formation must be tightly regulated to preserve the anti-microbial capability of NETs while minimizing damage to the host. In this review, we summarized the recent updates on the mechanism of NETs formation and pathophysiology associated with excessive NETs, aiming to provide insights for research and treatment of pulmonary infectious diseases.
Collapse
Affiliation(s)
- Ting Pan
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jae Woo Lee
- Department of Anesthesiology, University of California Los Angeles, Los Angeles, CA 90230, USA
| |
Collapse
|
7
|
Kahrizi MS, Nasiri K, Ebrahimzadeh F, Yaseri AF, Ghodratizadeh S, Gholamrezaei M, Rahat Dahmardeh A, Adili A, Amjidifar R, Hemmatzadeh M, Arabi M, Maghsoudi MR, Mohammadi H. Lymphopenia associated with survivin and its downstream pathway in COVID-19 serving as a potential route in COVID-19 pathogenesis. Adv Med Sci 2024; 69:190-197. [PMID: 38521459 DOI: 10.1016/j.advms.2024.03.006] [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: 12/14/2022] [Revised: 02/16/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
PURPOSE Starting in 2019, coronavirus disease 2019 (COVID-19) caused an epidemic that was growing rapidly and has harmed millions of people globally. It has been demonstrated that survivin regulates lymphocyte survival, a main route involved in COVID-19 pathogenesis. Survivin belongs to the inhibitor of apoptosis protein (IAP) family, and its primary functions comprise regulating mitosis and inhibiting apoptosis. Since lower survivin expression has been shown to increase the sensitivity of lymphocytes to apoptotic induction, we looked into the function of survivin and its corresponding pathways in COVID-19 pathogenesis. MATERIALS AND METHODS The expression of survivin, X-linked inhibitor of apoptosis protein (XIAP), caspases 3, 7, 9, and poly (ADP-ribose) polymerase (PARP) was evaluated at both mRNA and protein levels in peripheral blood mononuclear cells (PBMCs) derived from healthy donors and patients with severe and moderate COVID-19 by qRT-PCR and Western blotting, respectively. Then, we enforced apoptosis to COVID-19 patient-derived lymphocytes, and the percent was assessed by flow cytometry. RESULTS Survivin and XIAP were less expressed in PBMCs derived from COVID-19 patients as apoptosis inhibitors than PARP, cleaved-PARP, caspase 9, and cleaved caspases 3 and 7, according to the results of real-time PCR and Western blot analysis. Additionally, according to the flow cytometry results, the down-regulation of survivin served as a potential factor in the lymphocyte depletion observed in patients with COVID-19. CONCLUSION The role of survivin and its related pathway was first discovered in the development of COVID-19 and may serve as a potential prognostic and therapeutic target.
Collapse
Affiliation(s)
| | - Kamyar Nasiri
- Department of Dentistry, Islamic Azad University, Tehran, Iran
| | - Farnoosh Ebrahimzadeh
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Soroush Ghodratizadeh
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mostafa Gholamrezaei
- Department of Parasitology and Mycology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alireza Rahat Dahmardeh
- Department of Anesthesiology and Critical Care, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ali Adili
- Department of Oncology, Tabriz University of Medical Sciences, Tabriz, Iran; Senior Adult Oncology Department, Moffitt Cancer Center, University of South, Florida, USA
| | - Rosita Amjidifar
- Department of Microbiology, Islamic Azad University of Iran, Ahar, Iran
| | - Maryam Hemmatzadeh
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Arabi
- Department of Physiology, Pharmacology and Medical Physics, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Reza Maghsoudi
- Faculty of Emergency Medicine & Toxicology, Emergency Department, Alborz University of Medical Sciences, Karaj, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
8
|
Zurera-Egea C, Teniente-Serra A, Fuster D, Martínez-Cáceres E, Muga R, Zuluaga P. Cytotoxic NK cells phenotype and activated lymphocytes are the main characteristics of patients with alcohol-associated liver disease. Clin Exp Med 2023; 23:3539-3547. [PMID: 37392250 PMCID: PMC10618338 DOI: 10.1007/s10238-023-01121-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/13/2023] [Indexed: 07/03/2023]
Abstract
T cells, natural killer (NK) and NKT cells have opposing actions in the development of alcohol-associated liver fibrosis. We aimed to evaluate the phenotype of NK cells, NKT cells and activated T cells in patients with alcohol use disorder (AUD) according to the presence of advanced liver fibrosis (ALF). Totally, 79 patients (51-years, 71% males) were admitted to treatment of AUD. ALF was defined as FIB4-score > 2.67. Immunophenotyping of NK cells (CD3-CD56+CD16+, CD3-CD56+CD16-, CD3-CD56-CD16+), NKT-like (CD3+CD56+), and the activation status of CD4+, CD8+ and regulatory T cells (Tregs) were evaluated according to the HLA-DR expression. Patients had an AUD duration of 18 ± 11 years with a daily alcohol consumption of 155 ± 77 gr/day prior to hospital admission. The values of absolute cells were 2 ± 0.9 cells/L for total lymphocytes, 1054 ± 501 cells/µL for CD4+, 540 ± 335 cells/µL for CD8+, 49.3 ± 24.8 cells/µL for Tregs, 150.3 ± 97.5 cells/µL for NK cells and 69.8 ± 78.3 cells/µL for NKT-like. The percentage of total NK cells (11.3 ± 5.5% vs. 7 ± 4.3%, p < 0.01), CD3-CD56+CD16+ regarding total lymphocytes (9.7 ± 5.1% vs. 5.8 ± 3.9%, p < 0.01), activated CD4+ cells (5.2 ± 3.2% vs. 3.9 ± 3%, p = 0.04) and activated CD8+ cells (15.7 ± 9.1% vs. 12.2 ± 9%, p = 0.05) were significantly higher in patients with ALF. The percentage of CD3-CD56+CD16- regarding NK cells (5.1 ± 3.4% vs. 7.6 ± 6.2%, p = 0.03) was significantly lower in patients with ALF. Activated Tregs (39.9 ± 11.5 vs. 32.4 ± 9.2, p = 0.06) showed a tendency to be higher in patients with ALF. The proportion of activated CD4+ cells (r = 0.40, p < 0.01) and activated CD8+ cells (r = 0.51, p < 0.01) was correlated with the proportion of NKT-like in patients without ALF. Patients with ALF presented an increased NK cytotoxic phenotype and activated T cells concomitant with a decreased NK cytokine-secreting phenotype.
Collapse
Affiliation(s)
- Coral Zurera-Egea
- Genetics of Male Fertility Group, Unitat de Biologia Cel·lular (Facultat de Biociències), Departament de Biologia Cel·lular Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Aina Teniente-Serra
- Department of Inmunology, Hospital Universitari Germans Trias I Pujol, IGTP, Badalona, Barcelona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Daniel Fuster
- Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Internal Medicine, Hospital Universitari Germans Trias I Pujol, IGTP, Ctra. Canyet S/N, 08916, Badalona, Barcelona, Spain
| | - Eva Martínez-Cáceres
- Department of Inmunology, Hospital Universitari Germans Trias I Pujol, IGTP, Badalona, Barcelona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Roberto Muga
- Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Internal Medicine, Hospital Universitari Germans Trias I Pujol, IGTP, Ctra. Canyet S/N, 08916, Badalona, Barcelona, Spain
| | - Paola Zuluaga
- Universitat Autònoma de Barcelona, Barcelona, Spain.
- Department of Internal Medicine, Hospital Universitari Germans Trias I Pujol, IGTP, Ctra. Canyet S/N, 08916, Badalona, Barcelona, Spain.
| |
Collapse
|
9
|
Zhuxiao R, Fang X, Wei W, Shumei Y, Jianlan W, Qiuping L, Jingjun P, Chuan N, Yongsheng L, Zhichun F, Jie Y. Prevention for moderate or severe BPD with intravenous infusion of autologous cord blood mononuclear cells in very preterm infants-a prospective non-randomized placebo-controlled trial and two-year follow up outcomes. EClinicalMedicine 2023; 57:101844. [PMID: 36864985 PMCID: PMC9971511 DOI: 10.1016/j.eclinm.2023.101844] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is the primary severe complication of preterm birth. Severe BPD was associated with higher risks of mortality, more postnatal growth failure, long term respiratory and neurological developmental retardation. Inflammation plays a central role in alveolar simplification and dysregulated vascularization of BPD. There is no effective treatment to improve BPD severity in clinical practice. Our previous clinical study showed autologous cord blood mononuclear cells (ACBMNCs) infusion could reduce the respiratory support duration safely and potential improved BPD severity. Abundant preclinical studies have reported the immunomodulation effect as an important mechanism underlying the beneficial results of stem cell therapies in preventing and treating BPD. However, clinical studies assessing the immunomodulatory effect after stem cells therapy were rare. This study was to investigate the effect of ACBMNCs infusion soon after birth on prevention for severe BPD and long term outcomes in very preterm neonates. The immune cells and inflammatory biomarkers were detected to investigate the underlying immunomodulatory mechanisms. METHODS This single-center, prospective, investigator-initiated, non-randomized trial with blinded outcome assessment aimed to assess the effect of a single intravenous infusion of ACBMNCs in preventing severe BPD (moderate or severe BPD at 36 weeks of gestational age or discharge home) in surviving very preterm neonates less than 32 gestational weeks. Patients admitted to the Neonatal Intensive Care Unit (NICU) of Guangdong Women and Children Hospital from July 01, 2018 to January 1, 2020 were assigned to receiving a targeted dosage of 5 × 107 cells/kg ACBMNC or normal saline intravenously within 24 h after enrollment. Incidence of moderate or severe BPD in survivors were investigated as the primary short term outcome. Growth, respiratory and neurological development were assessed as long term outcomes at corrected age of 18-24 month-old. Immune cells and inflammatory biomarkers were detected for potential mechanism investigation. The trial was registered at ClinicalTrials.gov (NCT02999373). FINDINGS Six-two infants were enrolled, of which 29 were enrolled to intervention group, 33 to control group. Moderate or severe BPD in survivors significantly decreased in intervention group (adjusted p = 0.021). The number of patients needed to treat to gain one moderate or severe BPD-free survival was 5 (95% confidence interval: 3-20). Survivors in the intervention group had a significantly higher chance to be extubated than infants in the control group (adjusted p = 0.018). There was no statistical significant difference in total BPD incidence (adjusted p = 0.106) or mortality (p = 1.000). Incidence of developmental delay reduced in intervention group in long term follow-up (adjusted p = 0.047). Specific immune cells including proportion of T cells (p = 0.04) and CD4+ T cells in lymphocytes (p = 0.03), and CD4+ CD25+ forkhead box protein 3 (FoxP3)+ regulatory T cells in CD4+ T cells increased significantly after ACBMNCs intervention (p < 0.001). Anti-inflammatory factor IL-10 was higher (p = 0.03), while pro-inflammatory factor such as TNF-a (p = 0.03) and C reactive protein (p < 0.001) level was lower in intervention group than in control group after intervention. INTERPRETATION ACBMNCs could prevent moderate or severe BPD in surviving very premature neonates and might improve neurodevelopmental outcomes in long term. An immunomodulatory effect of MNCs contributed to the improvement of BPD severity. FUNDING This work was supported by National Key R&D Program of China (2021YFC2701700), National Natural Science Foundation of China (82101817, 82171714, 8187060625), Guangzhou science and technology program (202102080104).
Collapse
Affiliation(s)
- Ren Zhuxiao
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Xu Fang
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Wei Wei
- Guang Dong Cord Blood Bank, Guangzhou, China
| | - Yang Shumei
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Wang Jianlan
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Li Qiuping
- Department of Neonatology, The 7th Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
| | - Pei Jingjun
- Department of Neonatology, Nanfang Hospital, Southern Medical University, China
| | - Nie Chuan
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | | | - Feng Zhichun
- Department of Neonatology, The 7th Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
- Corresponding author. Department of Neonatology, The 7th Medical Center of PLA General Hospital, Beijing, China.
| | - Yang Jie
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
- Department of Neonatology, Nanfang Hospital, Southern Medical University, China
- Corresponding author. Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China; Department of Neonatology, Nanfang Hospital, Southern Medical University, China.
| |
Collapse
|