1
|
De Rose DU, Campi F, Maddaloni C, Ronci S, Caoci S, Savarese I, Bersani I, Ronchetti MP, Auriti C, Capolupo I, Merli P, Insalaco A, De Benedetti F, Dotta A. Off-Label Use of Anakinra in Inflammatory Conditions in Neonates and Infants Up to 3 Months of Age: A Case Series and a Review of the Literature. Paediatr Drugs 2025; 27:293-305. [PMID: 39804459 PMCID: PMC12031743 DOI: 10.1007/s40272-024-00679-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 04/26/2025]
Abstract
BACKGROUND Anakinra is an interleukin-1 receptor antagonist (IL-1Ra). Since IL-1 has been shown to play a key role in the etiology of different autoinflammatory diseases, blocking its pathway has become an important therapeutic target, even in neonates. AIMS We aimed to report our experience in using anakinra to treat specific neonatal inflammatory conditions. METHODS We described the clinical management with anakinra of five cases of neonates or infants up to 3 months of age admitted to the neonatal intensive care unit (NICU) of Bambino Gesù Children's Hospital IRCCS in Rome (Italy) from 2020 onwards. Medical history and clinical data concerning NICU hospitalization were collected from the electronic medical records. Furthermore, we performed a literature review of off-label anakinra in the first 3 months of life, up to 5 April 2024. We excluded from this review cases of cryopyrin-associated periodic syndrome, deficiency of the interleukin-1 receptor antagonist, and mevalonate kinase deficiency, for which anakinra is a known treatment. RESULTS We reported three off-label cardiorespiratory reasons to use IL-1Ra from our series: (i) chronic lung disease with pulmonary hypertension, (ii) interstitial lung disease with pulmonary hypertension to facilitate the weaning from respiratory support, and (iii) post-surgical polyserositis if effusions accumulate despite drainage. In all our patients, the drug was administered at a dosage of 10 mg/kg/day. The route of administration was chosen based on the patient's clinical characteristics, with the subcutaneous and intravenous routes being comparable in efficacy. The duration of therapy was modulated based on the patient's clinical response, with a minimum duration of 4 months. A total of 308 retrieved articles were screened, and then full texts of records deemed eligible for inclusion were assessed. Based on the literature search and our five cases, a total of 17 infants were treated with anakinra outside its approved indications. The major off-label use was for hemophagocytic lymphohistiocytosis/macrophage activation syndrome, followed by multisystem inflammatory syndrome in children and Kawasaki disease, as in two of our cases. CONCLUSIONS According to the results of our case series and review of the literature, the off-label use of anakinra in neonates with inflammatory conditions refractory to first-line therapy could be considered. Prospective, multicenter research is necessary to determine whether anakinra is a safe treatment option for these infants to prevent early inflammatory illnesses and in which situations it could enhance clinical results.
Collapse
Affiliation(s)
- Domenico Umberto De Rose
- Neonatal Intensive Care Unit-"Bambino Gesù" Children's Hospital IRCCS, 00165, Rome, Italy
- Microbiology, Immunology, Infectious Diseases, and Transplants (MIMIT), Faculty of Medicine and Surgery, "Tor Vergata" University of Rome, Rome, Italy
| | - Francesca Campi
- Neonatal Intensive Care Unit-"Bambino Gesù" Children's Hospital IRCCS, 00165, Rome, Italy
| | - Chiara Maddaloni
- Neonatal Intensive Care Unit-"Bambino Gesù" Children's Hospital IRCCS, 00165, Rome, Italy
| | - Sara Ronci
- Neonatal Intensive Care Unit-"Bambino Gesù" Children's Hospital IRCCS, 00165, Rome, Italy.
| | - Stefano Caoci
- Neonatal Intensive Care Unit-"Bambino Gesù" Children's Hospital IRCCS, 00165, Rome, Italy
| | - Immacolata Savarese
- Neonatal Intensive Care Unit-"Bambino Gesù" Children's Hospital IRCCS, 00165, Rome, Italy
| | - Iliana Bersani
- Neonatal Intensive Care Unit-"Bambino Gesù" Children's Hospital IRCCS, 00165, Rome, Italy
| | - Maria Paola Ronchetti
- Neonatal Intensive Care Unit-"Bambino Gesù" Children's Hospital IRCCS, 00165, Rome, Italy
| | - Cinzia Auriti
- Saint Camillus International University of Health Sciences, Rome, Italy
| | - Irma Capolupo
- Neonatal Intensive Care Unit-"Bambino Gesù" Children's Hospital IRCCS, 00165, Rome, Italy
| | - Pietro Merli
- Hematology and Oncology Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | | | | | - Andrea Dotta
- Neonatal Intensive Care Unit-"Bambino Gesù" Children's Hospital IRCCS, 00165, Rome, Italy
| |
Collapse
|
2
|
Guo Y, Deng F, Jiang Y, Cao G, Zhang Y, Liu G, Alimujiang M, Ayati M, Chen Y, Chen L, Lv S, Dou X. IL-37 Alleviates Sepsis-Induced Lung Injury by Inhibiting Inflammatory Response Through the TGF-β/Smad3 Pathway. Immunol Invest 2025:1-15. [PMID: 40270428 DOI: 10.1080/08820139.2025.2495958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Introduction: Sepsis is caused by an uncontrolled inflammatory response and immune dysfunction, with lung injury being the most common complication and one of the leading causes of death in clinically ill patients. Interleukin 37 (IL-37) is a multifunctional cytokine that plays a vital role in various pathophysiological processes, including inflammation, infection, and immunity.Methods: The study involved both clinical and animal experiments (establishing an animal model of sepsis-induced lung injury). Firstly, 50 patients with sepsis-induced lung injury and 50 healthy controls were included. In addition, a more in-depth study was conducted using animal models.Results: IL-37, IL-6, PCT, and CRP levels were significantly higher in the sepsis-induced lung injury group. Correlation analysis revealed that IL-37 significantly correlated with IL-6, PCT, and CRP levels. In animal experiments, IL-37 significantly attenuated CLP-induced pulmonary edema and cellular injury while reducing the levels of inflammatory factors IL-6 and TNF-α, as well as sepsis-related inflammatory markers PCT and CRP. Moreover, IL-37 significantly downregulated the expression levels of genes and proteins of apoptosis-related molecules Caspase-3 and Bax and pathway molecules TGF-β and Smad3. Discussion: The TGF-β/Smad3 pathway is involved in the process of IL-37 inhibiting inflammatory response and ameliorating sepsis-induced lung injury.
Collapse
Affiliation(s)
- Yufang Guo
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
| | - Feifei Deng
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Yali Jiang
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- Ili & Jiangsu Joint Institute of Health, Yili, Xinjiang, P.R. China
| | - Guodong Cao
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
- Ili & Jiangsu Joint Institute of Health, Yili, Xinjiang, P.R. China
| | - Yixin Zhang
- Department of Critical Care Medicine, Medical School of Shihezi University, Shihezi, Xinjiang, P.R. China
| | - Gaowu Liu
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Mayinur Alimujiang
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Mairhaba Ayati
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Yufeng Chen
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Lili Chen
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Su Lv
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Xueqin Dou
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| |
Collapse
|
3
|
Su X, Sun Y, Dai A. New insights into pulmonary arterial hypertension: interaction between PANoptosis and perivascular inflammatory responses. Apoptosis 2025:10.1007/s10495-025-02086-0. [PMID: 39979525 DOI: 10.1007/s10495-025-02086-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
Pulmonary arterial hypertension (PAH) is a heterogeneous disease characterized by various etiologies, with pulmonary vascular remodeling recognized as a main pathological change. Currently, it is widely accepted that vascular remodeling is closely associated with abnormal pulmonary vascular cell death and perivascular inflammation. The simultaneous activation of various pulmonary vascular cell death leads to immune cell adhesion and inflammatory mediator releases; And in turn, the inflammatory response may also trigger cell death and jointly promote the progression of vascular remodeling. Recently, PANoptosis has been identified as a phenomenon that describes the simultaneous activation and interaction of multiple forms of programmed cell death (PCD). Therefore, the relationship between PANoptosis and inflammation in PAH warrants further investigation. This review examines the mechanisms underlying apoptosis, necroptosis, pyroptosis, and inflammatory responses in PAH, with a focus on PANoptosis and its interactions with inflammation. And it aims to elucidate the significance of this emerging form of cell death and inflammation in the pathophysiology of PAH and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xianli Su
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Yinhui Sun
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China.
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China.
| |
Collapse
|
4
|
Kim Y, Marić I, Kashiwagi CM, Han L, Chung P, Reiss JD, Butcher LD, Caoili KJ, Berson E, Xue L, Espinosa C, James T, Shome S, Xie F, Ghanem M, Seong D, Chang AL, Reincke SM, Mataraso S, Shu CH, De Francesco D, Becker M, Kumar WM, Wong R, Gaudilliere B, Angst MS, Shaw GM, Bateman BT, Stevenson DK, Prince LS, Aghaeepour N. PregMedNet: Multifaceted Maternal Medication Impacts on Neonatal Complications. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.13.25322242. [PMID: 39990567 PMCID: PMC11844599 DOI: 10.1101/2025.02.13.25322242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
While medication intake is common among pregnant women, medication safety remains underexplored, leading to unclear guidance for patients and healthcare professionals. PregMedNet addresses this gap by providing a multifaceted maternal medication safety framework based on systematic analysis of 1.19 million mother-baby dyads from U.S. claims databases. A novel confounding adjustment pipeline was applied to systematically control confounders for multiple medication-disease pairs, robustly identifying both known and novel maternal medication effects. Notably, one of the newly discovered associations was experimentally validated, demonstrating the reliability of claims data and machine learning for perinatal medication safety studies. Additionally, potential biological mechanisms of newly identified associations were generated using a graph learning method. These findings highlight PregMedNet's value in promoting safer medication use during pregnancy and maternal-neonatal outcomes.
Collapse
Affiliation(s)
- Yeasul Kim
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
| | - Ivana Marić
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
| | - Chloe M Kashiwagi
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Lichy Han
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
| | - Philip Chung
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
| | | | | | | | - Eloïse Berson
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lei Xue
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
| | - Camilo Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Tomin James
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
| | - Sayane Shome
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
| | - Feng Xie
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
| | - Marc Ghanem
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
| | - David Seong
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan L Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
| | - S Momsen Reincke
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
| | - Samson Mataraso
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
| | - Chi-Hung Shu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
| | - Davide De Francesco
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
| | - Martin Becker
- Institute for Visual and Analytic Computing, University of Rostock, Rostock, Germany
| | - Wasan M Kumar
- Medical Doctor Program, Stanford University School of Medicine, Stanford, CA, USA
- Graduate School of Business, Stanford University School of Medicine, Stanford, CA, USA
| | - Ron Wong
- Department of Pediatrics, Stanford School of Medicine
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
| | - Gary M Shaw
- Department of Pediatrics, Stanford School of Medicine
| | - Brian T Bateman
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
| | | | | | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine
- Department of Pediatrics, Stanford School of Medicine
- Department of Biomedical Data Science, Stanford University
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
5
|
Paton MCB, Benders M, Blatch-Williams R, Dallimore E, Edwards A, Elwood N, Facer K, Finch-Edmondson M, Garrity N, Gordon A, Hunt RW, Jenkin G, McDonald CA, Moore J, Nold MF, Novak I, Popat H, Salomon C, Sato Y, Tolcos M, Wixey JA, Yawno T, Zhou L, Malhotra A. Updates on neonatal cell and novel therapeutics: Proceedings of the Second Neonatal Cell Therapies Symposium (2024). Pediatr Res 2025:10.1038/s41390-025-03856-x. [PMID: 39815092 DOI: 10.1038/s41390-025-03856-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/31/2024] [Indexed: 01/18/2025]
Abstract
Cell therapies as treatments for neonatal conditions have attracted significant research and parent interest over the last two decades. Mesenchymal stromal cells, umbilical cord blood cells and neural stem cells translate from lab, to preclinical and into clinical trials, with contributions being made from all over the world. Effective and timely translation involves frequent reflection and consultation from research-adjacent fields (i.e. cell therapies for cerebral palsy, adult neurology, companies, and regulatory bodies) as well as meaningful involvement of people with lived experience. Progress to date suggests that aligning outcome and data reporting in later phase clinical trials will support our sector, as well as involving industry partners for streamlined solutions in cell manufacturing, commercialisation and regulatory processes. Importantly, our field can also benefit from resource sharing and research collaboration in novel drug therapies, small molecules and extracellular vesicles as we attempt to bridge preclinical and clinical research. In this review, we present highlights and learnings from the second Neonatal Cell Therapies Symposium (2024), held in Sydney, Australia. IMPACT: Multiple cell therapy candidates have advanced through preclinical and clinical trials in neonatology, showing promising feasibility, safety and efficacy. Effective and timely translation is enabled by collaboration across research-adjacent fields, commercial partnerships, harmonising research outcomes and meaningful involvement of people with lived experience. Progress on the potential utility of cell therapies for neonatal conditions and further translational considerations are discussed in this paper.
Collapse
Affiliation(s)
- Madison C B Paton
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Manon Benders
- Wilhemina Children's Hospital, Neonatology Department, Utrecht Brain Center, University Medical Centre, University Utrecht, Utrecht, The Netherlands
| | - Remy Blatch-Williams
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | | | - Adam Edwards
- Argenica Therapeutics LTD, Nedlands, WA, Australia
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, WA, Australia
| | - Ngaire Elwood
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- BMDI Cord Blood Bank, Melbourne, VIC, Australia
| | - Kylie Facer
- Parent with Lived Experience, Sydney, Australia
| | - Megan Finch-Edmondson
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Natasha Garrity
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Adrienne Gordon
- Discipline of Obstetrics, Gynaecology, and Neonatology, The University of Sydney, Sydney, NSW, Australia
| | - Rod W Hunt
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Justin Moore
- Department of Neurosurgery, Monash Health, Melbourne, VIC, Australia
| | - Marcel F Nold
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Iona Novak
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Himanshu Popat
- The Children's Hospital at Westmead, Sydney, NSW, Australia
- NHMRC Clinical Trial Centre, University of Sydney, Camperdown, VIC, Australia
| | - Carlos Salomon
- Exosome Biology Laboratory, University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, QLD, Australia
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Julie A Wixey
- Perinatal Research Centre, University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, QLD, Australia
| | - Tamara Yawno
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Lindsay Zhou
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia.
- Monash Newborn, Monash Children's Hospital, Melbourne, VIC, Australia.
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia.
| |
Collapse
|
6
|
Li Y, Yu J, Zeng Z, Lin W. Regulation of ubiquitination in sepsis: from PAMP versus DAMP to peripheral inflammation and cell death. Front Immunol 2024; 15:1513206. [PMID: 39720715 PMCID: PMC11666442 DOI: 10.3389/fimmu.2024.1513206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Sepsis (sepsis) is a systemic inflammatory response triggered by infection, and its pathologic features include overproduction of peripheral inflammatory factors (e.g., IL-1β, IL-6, TNF-α), which ultimately leads to cytokine storm and multiple organ dysfunction syndrome (MODS). Pathogen-associated molecular patterns (PAMP) and damage-associated molecular patterns (DAMP) induce strong immune responses and exacerbate inflammation by activating pattern recognition receptors (PRRs) in the host. Ubiquitination, as a key protein post-translational modification, dynamically regulates the activity of several inflammation-associated proteins (e.g., RIPK1, NLRP3) through the coordinated action of the E1, E2, and E3 enzymes, affects cell death pathways such as necroptosis and pyroptosis, and ultimately regulates the release of peripheral inflammatory factors. Deubiquitinating enzymes (DUBs), on the other hand, influence the intensity of the inflammatory response in sepsis by counter-regulating the ubiquitination process and balancing pro- and anti-inflammatory signals. This review focuses on how PAMP and DAMP activate inflammatory pathways via PRRs, and the central role of ubiquitination and deubiquitination in the development of sepsis, especially the mechanisms in regulating the secretion of peripheral inflammatory factors and cell death. By deeply dissecting the impact of the balance of ubiquitination and deubiquitination on inflammatory regulation, we further envision its potential as a therapeutic target in sepsis.
Collapse
Affiliation(s)
| | | | | | - Weixiong Lin
- Department of Anesthesiology I, Meizhou People’s Hospital,
Meizhou, Guangdong, China
| |
Collapse
|
7
|
Xie W, Ding B, Lou J, Wang X, Guo X, Zhu J. Metformin attenuates white matter injury in neonatal mice through activating NRF2/HO-1/NF-κB pathway. Int Immunopharmacol 2024; 141:112961. [PMID: 39163687 DOI: 10.1016/j.intimp.2024.112961] [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: 05/29/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024]
Abstract
White matter injury (WMI) is a major form of brain injury that occurs in preterm infants and develops into lifelong disabilities, including cerebral palsy, impaired cognitive function, and psychiatric disorders. Metformin (MET) has been reported to have neuroprotective effects. However, whether MET is responsible for neuroprotection against WMI remains unclear. In this study, we established a WMI model in neonatal mice to explore the neuroprotective effects of MET and attempted to elucidate its potential mechanisms. Our results showed that MET increased the expression of myelin basic protein (MBP), oligodendrocyte transcription factor 2 (Olig2), and CC1, improved the thickness and density of the myelin sheath, and reduced oxidative stress and microglial infiltration after chronic hypoxia induction. Moreover, MET improved memory, learning, and motor abilities as well as relieved anxiety-like behaviors in mice with WMI. These protective effects of MET may involve the upregulation of the nuclear factor erythroid 2-related factor 2 (NRF2)/heme oxygenase-1(HO-1)/NF-κB pathway related protein expressions. In addition, the NRF2 inhibitor ML385 could significantly reverse the effects of MET. In conclusion, this study suggested that MET attenuated chronic hypoxia-induced WMI through activating the NRF2/HO-1/NF-κB pathway, indicating that MET might be a promising therapeutic option for WMI.
Collapse
Affiliation(s)
- Weiwei Xie
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Pediatrics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 150 Ximen Street, Linhai, Zhejiang, China
| | - Bingqing Ding
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jia Lou
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinyi Wang
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoling Guo
- Scientific Research Department, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China.
| | - Jianghu Zhu
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China.
| |
Collapse
|
8
|
Glaser K, Jensen EA, Wright CJ. Prevention of Inflammatory Disorders in the Preterm Neonate: An Update with a Special Focus on Bronchopulmonary Dysplasia. Neonatology 2024; 121:636-645. [PMID: 38870912 PMCID: PMC11444906 DOI: 10.1159/000539303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/08/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND The rates of major neonatal morbidities, such as bronchopulmonary dysplasia, necrotizing enterocolitis, preterm white matter disease, and retinopathy of prematurity, remain high among surviving preterm infants. Exposure to inflammatory stimuli and the subsequent host innate immune response contribute to the risk of developing these complications of prematurity. Notably, the burden of inflammation and associated neonatal morbidity is inversely related to gestational age - leaving primarily but not exclusively the tiniest babies at highest risk. SUMMARY Avoidance, prevention, and treatment of inflammation to reduce this burden remain a major goal for neonatologists worldwide. In this review, we discuss the link between the host response to inflammatory stimuli and the disease state. We argue that inflammatory exposures play a key role in the pathobiology of preterm birth and that preterm neonates hereafter are highly susceptible to immune stimulation not only from their surrounding environment but also from therapeutic interventions employed in clinical care. Using bronchopulmonary dysplasia as an example, we report clinical studies demonstrating the potential utility of targeting inflammation to prevent this neonatal morbidity. On the contrary, we highlight limitations in our current understanding of how inflammation contributes to disease prevention and treatment. KEY MESSAGE To be successful in preventing and treating inflammation-driven morbidity in neonatal intensive care, it may be necessary to better identify at-risk patients and pair therapeutic interventions to key pathways and mediators of inflammation-associated neonatal morbidity identified in pre-clinical and translational studies.
Collapse
Affiliation(s)
- Kirsten Glaser
- Division of Neonatology, Department of Women's and Children's Health, University of Leipzig Medical Center, Leipzig, Germany
| | - Erik A Jensen
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado and University of Colorado School of Medicine Aurora, Aurora, Colorado, USA
| |
Collapse
|
9
|
Perrone S, Carloni S, Dell'Orto VG, Filonzi L, Beretta V, Petrolini C, Lembo C, Buonocore G, Esposito S, Nonnis Marzano F. Hypoxic ischemic brain injury: animal models reveal new mechanisms of melatonin-mediated neuroprotection. Rev Neurosci 2024; 35:331-339. [PMID: 38153803 DOI: 10.1515/revneuro-2023-0126] [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: 10/13/2023] [Accepted: 11/24/2023] [Indexed: 12/30/2023]
Abstract
Oxidative stress (OS) and inflammation play a key role in the development of hypoxic-ischemic (H-I) induced brain damage. Following H-I, rapid neuronal death occurs during the acute phase of inflammation, and activation of the oxidant-antioxidant system contributes to the brain damage by activated microglia. So far, in an animal model of perinatal H-I, it was showed that neuroprostanes are present in all brain damaged areas, including the cerebral cortex, hippocampus and striatum. Based on the interplay between inflammation and OS, it was demonstrated in the same model that inflammation reduced brain sirtuin-1 expression and affected the expression of specific miRNAs. Moreover, through proteomic approach, an increased expression of genes and proteins in cerebral cortex synaptosomes has been revealed after induction of neonatal H-I. Administration of melatonin in the experimental treatment of brain damage and neurodegenerative diseases has produced promising therapeutic results. Melatonin protects against OS, contributes to reduce the generation of pro-inflammatory factors and promotes tissue regeneration and repair. Starting from the above cited aspects, this educational review aims to discuss the inflammatory and OS main pathways in H-I brain injury, focusing on the role of melatonin as neuroprotectant and providing current and emerging evidence.
Collapse
Affiliation(s)
- Serafina Perrone
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Aurelio Saffi 2, 61029 Urbino, Italy
| | - Valentina Giovanna Dell'Orto
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Laura Filonzi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| | - Virginia Beretta
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Petrolini
- Neonatology Unit, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Lembo
- Department of Neonatology, APHP, Necker-Enfants, Malades Hospital, 149 Rue de Sèvres, 75015 Paris, France
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, Via Banchi di Sotto 55, 53100 Siena, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, Pietro Barilla Children's Hospital, Via Gramsci 14, 43126 Parma, Italy
| | - Francesco Nonnis Marzano
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| |
Collapse
|
10
|
Dong X, Chen X, Xue M, Zhang Y. Changes in serum inflammatory factors in group B streptococcal infection and their predictive value for premature rupture of membranes complicated by chorioamnionitis. Biomark Med 2024; 18:301-309. [PMID: 38623925 PMCID: PMC11218798 DOI: 10.2217/bmm-2023-0588] [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: 09/18/2023] [Accepted: 02/02/2024] [Indexed: 04/17/2024] Open
Abstract
Objective: The aim of this study as to unveil changes in serum inflammatory factors in pregnant women with genital tract group B Streptococcus (GBS) infection and their predictive value for premature rupture of membranes (PROM) complicated by chorioamnionitis (CS) and adverse pregnancy outcomes. Methods: The value of serum inflammatory factor levels in predicting PROM complicating CS and adverse pregnancy outcomes in GBS-infected pregnant women was evaluated by ELISA. Results: Serum IL-6, TNF-α, PCT and hs-CRP levels were higher in pregnant women with GBS infection. The combined diagnosis of these factors had excellent diagnostic value in PROM complicating CS and adverse pregnancy outcomes. Conclusion: Joint prediction of IL-6, TNF-α, PCT and hs-CRP has the best predictive value for PROM complicating CS and adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Xiaorui Dong
- Department of Obstetrics, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, 310000, China
| | - Xixi Chen
- Department of Obstetrics, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, 310000, China
| | - Mengling Xue
- Department of Obstetrics, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, 310000, China
| | - Yina Zhang
- Department of Obstetrics, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, 310000, China
| |
Collapse
|
11
|
Zhu Y, He L, Zhu Y, Yao H, Jiang J, Lu H. IRF4 affects the protective effect of regulatory T cells on the pulmonary vasculature of a bronchopulmonary dysplasia mouse model by regulating FOXP3. Mol Med 2024; 30:6. [PMID: 38195465 PMCID: PMC10777489 DOI: 10.1186/s10020-023-00770-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/12/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in preterm infants, characterised by compromised alveolar development and pulmonary vascular abnormalities. Emerging evidence suggests that regulatory T cells (Tregs) may confer protective effects on the vasculature. Knockdown of their transcription factor, interferon regulatory factor 4 (IRF4), has been shown to promote vascular endothelial hyperplasia. However, the involvement of Tregs and IRF4 in the BPD pathogenesis remains unclear. This study aimed to investigate the regulation of Tregs by IRF4 and elucidate its potential role in pulmonary vasculature development in a BPD mouse model. METHODS The BPD model was established using 85% hyperoxia exposure, with air exposure as the normal control. Lung tissues were collected after 7 or 14 days of air or hyperoxia exposure, respectively. Haematoxylin-eosin staining was performed to assess lung tissue pathology. Immunohistochemistry was used to measure platelet endothelial cell adhesion molecule-1 (PECAM-1) level, flow cytometry to quantify Treg numbers, and Western blot to assess vascular endothelial growth factor (VEGFA), angiopoietin-1 (Ang-1), forkhead box protein P3 (FOXP3), and IRF4 protein levels. We also examined the co-expression of IRF4 and FOXP3 proteins using immunoprecipitation and immunofluorescence double staining. Furthermore, we employed CRISPR/Cas9 technology to knock down the IRF4 gene and observed changes in the aforementioned indicators to validate its effect on pulmonary vasculature development in mice. RESULTS Elevated IRF4 levels in BPD model mice led to FOXP3 downregulation, reduced Treg numbers, and impaired pulmonary vascular development. Knockdown of IRF4 resulted in improved pulmonary vascular development and upregulated FOXP3 level. CONCLUSION IRF4 may affect the protective role of Tregs in the proliferation of pulmonary vascular endothelial cells and pulmonary vascular development in BPD model mice by inhibiting the FOXP3 level.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Langyue He
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yue Zhu
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Huici Yao
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jianfeng Jiang
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Hongyan Lu
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
12
|
Hawkes JE, Visvanathan S, Krueger JG. The role of the interleukin-36 axis in generalized pustular psoriasis: a review of the mechanism of action of spesolimab. Front Immunol 2023; 14:1292941. [PMID: 38077370 PMCID: PMC10703363 DOI: 10.3389/fimmu.2023.1292941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/31/2023] [Indexed: 12/18/2023] Open
Abstract
Generalized pustular psoriasis (GPP) is a rare, chronic, inflammatory skin disorder characterized by recurrent flares associated with skin erythema, desquamation, and widespread superficial sterile pustules, which may be severe ("lakes of pus"). Systemic symptoms are often present, including malaise, fever, and skin pain. In GPP, innate immune responses are driven by abnormal activation of the interleukin (IL)-36-chemokine-neutrophil axis and excessive neutrophil infiltration. This review highlights the IL-36 pathway in the context of the IL-1 superfamily and describes how unopposed IL-36 signaling can lead to the development of GPP. Targeted inhibition of the IL-36 receptor (IL-36R) is an attractive therapeutic strategy in the treatment of GPP, including flare prevention and sustained disease control. Spesolimab is a first-in-class, humanized, monoclonal antibody that binds specifically to the IL-36R and antagonizes IL-36 signaling. Spesolimab was approved by the US Food and Drug Administration in September 2022 to treat GPP flares in adults and was subsequently approved for GPP flare treatment in other countries across the world. Anti-IL-36R therapy, such as spesolimab, can mitigate flares and address flare prevention in GPP, presumably through rebalancing IL-36 signaling and modulating the pro-inflammatory response of the downstream effectors.
Collapse
Affiliation(s)
- Jason E. Hawkes
- Integrative Skin Science and Research and Pacific Skin Institute, Sacramento, CA, United States
| | - Sudha Visvanathan
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, United States
| | - James G. Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, United States
| |
Collapse
|
13
|
Kelly SB, Tran NT, Polglase GR, Hunt RW, Nold MF, Nold-Petry CA, Olson DM, Chemtob S, Lodygensky GA, Robertson SA, Gunn AJ, Galinsky R. A systematic review of immune-based interventions for perinatal neuroprotection: closing the gap between animal studies and human trials. J Neuroinflammation 2023; 20:241. [PMID: 37864272 PMCID: PMC10588248 DOI: 10.1186/s12974-023-02911-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/28/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Perinatal infection/inflammation is associated with a high risk for neurological injury and neurodevelopmental impairment after birth. Despite a growing preclinical evidence base, anti-inflammatory interventions have not been established in clinical practice, partly because of the range of potential targets. We therefore systematically reviewed preclinical studies of immunomodulation to improve neurological outcomes in the perinatal brain and assessed their therapeutic potential. METHODS We reviewed relevant studies published from January 2012 to July 2023 using PubMed, Medline (OvidSP) and EMBASE databases. Studies were assessed for risk of bias using the SYRCLE risk of bias assessment tool (PROSPERO; registration number CRD42023395690). RESULTS Forty preclinical publications using 12 models of perinatal neuroinflammation were identified and divided into 59 individual studies. Twenty-seven anti-inflammatory agents in 19 categories were investigated. Forty-five (76%) of 59 studies reported neuroprotection, from all 19 categories of therapeutics. Notably, 10/10 (100%) studies investigating anti-interleukin (IL)-1 therapies reported improved outcome, whereas half of the studies using corticosteroids (5/10; 50%) reported no improvement or worse outcomes with treatment. Most studies (49/59, 83%) did not control core body temperature (a known potential confounder), and 25 of 59 studies (42%) did not report the sex of subjects. Many studies did not clearly state whether they controlled for potential study bias. CONCLUSION Anti-inflammatory therapies are promising candidates for treatment or even prevention of perinatal brain injury. Our analysis highlights key knowledge gaps and opportunities to improve preclinical study design that must be addressed to support clinical translation.
Collapse
Affiliation(s)
- Sharmony B Kelly
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Nhi T Tran
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, VIC, 3168, Australia
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, VIC, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Rodney W Hunt
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, VIC, 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children's Hospital, Melbourne, Australia
| | - Marcel F Nold
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, VIC, 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children's Hospital, Melbourne, Australia
| | - Claudia A Nold-Petry
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, VIC, 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - David M Olson
- Department of Obstetrics and Gynaecology, University of Alberta, Edmonton, Canada
| | - Sylvain Chemtob
- Department of Paediatrics, CHU Sainte Justine Research Centre, University of Montreal, Quebec, Canada
| | - Gregory A Lodygensky
- Department of Paediatrics, CHU Sainte Justine Research Centre, University of Montreal, Quebec, Canada
| | - Sarah A Robertson
- The University of Adelaide, Robinson Research Institute, North Adelaide, SA, Australia
| | - Alistair J Gunn
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Melbourne, VIC, 3168, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Chen W, Xi S, Ke Y, Lei Y. The emerging role of IL-38 in diseases: A comprehensive review. Immun Inflamm Dis 2023; 11:e991. [PMID: 37647430 PMCID: PMC10461426 DOI: 10.1002/iid3.991] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/30/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023] Open
Abstract
INTRODUCTION Interleukin-38 (IL-38) is a new type of anti-inflammatory cytokine, which is mainly expressed in the immunity-related organs and is involved in various diseases including cardiovascular and cerebrovascular diseases, lung diseases, viral infectious diseases and autoimmune diseases. AIM This review aims to detail the biological function, receptors and signaling of IL-38, which highlights its therapeutic potential in related diseases. CONCLUSION This article provides a comprehensive review of the association between interleukin-38 and related diseases, using interleukin-38 as a keyword and searching the relevant literature through Pubmed and Web of science up to July 2023.
Collapse
Affiliation(s)
- Weijun Chen
- Center of Forensic ExpertiseAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
- School of Forensic MedcineZunyi Medical UniversityZunyiGuizhouChina
| | - Shuangyun Xi
- Center of Forensic ExpertiseAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
- School of Forensic MedcineZunyi Medical UniversityZunyiGuizhouChina
| | - Yong Ke
- Center of Forensic ExpertiseAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
- School of Forensic MedcineZunyi Medical UniversityZunyiGuizhouChina
| | - Yinlei Lei
- Center of Forensic ExpertiseAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
- School of Forensic MedcineZunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
15
|
Borțea CI, Enatescu I, Dima M, Pantea M, Iacob ER, Dumitru C, Popescu A, Stoica F, Heredea RE, Iacob D. A Prospective Analysis of the Retinopathy of Prematurity Correlated with the Inflammatory Status of the Extremely Premature and Very Premature Neonates. Diagnostics (Basel) 2023; 13:2105. [PMID: 37371000 DOI: 10.3390/diagnostics13122105] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Retinopathy of Prematurity (ROP) is a major cause of blindness in premature infants. This study aimed to evaluate the association between inflammatory markers and ROP development in extremely premature and very premature neonates and identify potential inflammatory biomarkers for ROP risk prediction. This prospective study was conducted from January 2021 to January 2023 in two clinical hospitals associated with the "Victor Babes" University of Medicine and Pharmacy Timisoara. The study population comprised neonates with a gestational age of less than 32 weeks. Various inflammatory markers, including total white blood cell count, polymorphonuclear leukocytes, C-reactive protein, interleukin-6, and lactate dehydrogenase, were analyzed from blood samples collected at birth and three days postnatally. ROP was diagnosed and classified following the International Classification of Retinopathy of Prematurity. The study included 48 neonates, 12 Extremely Premature Infants (EPI), and 36 Very Premature Infants (VPI). The EPI group had significantly higher mean interleukin-6 and lactate dehydrogenase levels at birth and three days postnatally than the VPI group. C-reactive protein levels at three days were significantly higher in the VPI group. Umbilical cord inflammation and ROP severity were found to have a statistically significant positive correlation. Half of the EPIs had moderate to severe ROP, significantly more than in the VPI group. The duration of oxygen supplementation, mechanical ventilation, Continuous Positive Airway Pressure (CPAP), gestational age less than 28 weeks, and umbilical cord inflammation at or above stage 3 were significant risk factors for developing ROP stage 2 or above. Elevated CRP and IL-6 were also significantly associated with an increased risk of developing ROP stage 2 or above, highlighting their potential as biomarkers for ROP risk prediction. This study suggests a significant association between inflammatory markers and ROP development in extremely premature and very premature neonates. These findings could contribute to the identification of potential inflammatory biomarkers for ROP risk prediction, improving early diagnosis and intervention strategies for this condition.
Collapse
Affiliation(s)
- Claudia Ioana Borțea
- Department of Neonatology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Doctoral School, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Ileana Enatescu
- Department of Neonatology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Mirabela Dima
- Department of Neonatology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Manuela Pantea
- Department of Neonatology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Doctoral School, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Emil Radu Iacob
- Department of Pediatric Surgery, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Catalin Dumitru
- Department of Obstetrics and Gynecology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Alin Popescu
- Department of Obstetrics and Gynecology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Florina Stoica
- Department of Ophthalmology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Rodica Elena Heredea
- Department of Pathology, "Louis Turcanu" Children's Clinical Emergency Hospital, 300041 Timisoara, Romania
- Department of Clinical Practical Skills, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Daniela Iacob
- Department of Neonatology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| |
Collapse
|
16
|
Owen JC, Garrick SP, Peterson BM, Berger PJ, Nold MF, Sehgal A, Nold-Petry CA. The role of interleukin-1 in perinatal inflammation and its impact on transitional circulation. Front Pediatr 2023; 11:1130013. [PMID: 36994431 PMCID: PMC10040554 DOI: 10.3389/fped.2023.1130013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/31/2023] Open
Abstract
Preterm birth is defined as delivery at <37 weeks of gestational age (GA) and exposes 15 million infants worldwide to serious early life diseases. Lowering the age of viability to 22 weeks GA entailed provision of intensive care to a greater number of extremely premature infants. Moreover, improved survival, especially at extremes of prematurity, comes with a rising incidence of early life diseases with short- and long-term sequelae. The transition from fetal to neonatal circulation is a substantial and complex physiologic adaptation, which normally happens rapidly and in an orderly sequence. Maternal chorioamnionitis or fetal growth restriction (FGR) are two common causes of preterm birth that are associated with impaired circulatory transition. Among many cytokines contributing to the pathogenesis of chorioamnionitis-related perinatal inflammatory diseases, the potent pro-inflammatory interleukin (IL)-1 has been shown to play a central role. The effects of utero-placental insufficiency-related FGR and in-utero hypoxia may also be mediated, in part, via the inflammatory cascade. In preclinical studies, blocking such inflammation, early and effectively, holds great promise for improving the transition of circulation. In this mini-review, we outline the mechanistic pathways leading to abnormalities in transitional circulation in chorioamnionitis and FGR. In addition, we explore the therapeutic potential of targeting IL-1 and its influence on perinatal transition in the context of chorioamnionitis and FGR.
Collapse
Affiliation(s)
- Josephine C. Owen
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Steven P. Garrick
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Briana M. Peterson
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Philip J. Berger
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Marcel F. Nold
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Arvind Sehgal
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Claudia A. Nold-Petry
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- Correspondence: Claudia A. Nold-Petry
| |
Collapse
|