1
|
Abd El-Fattah AA, Hamid Sadik NA, Shahin AM, Shahin NN. Simvastatin and eugenol restore autophagic flux and alleviate oxidative, inflammatory, and fibrotic perturbations in an arginine-induced chronic pancreatitis rat model. Arch Biochem Biophys 2025; 768:110357. [PMID: 40015469 DOI: 10.1016/j.abb.2025.110357] [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: 10/25/2023] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Chronic pancreatitis (CP), a progressive inflammatory disease characterized by pancreatic tissue destruction and fibrosis, is considered a challenging health burden due to insufficiencies of current management procedures. Autophagy impairment has emerged as a major triggering event in pancreatitis, raising interest in exploring the potential of targeting autophagy as a possible interventional strategy. This study aimed to evaluate the possible ameliorative effect of two autophagy modulators, simvastatin and eugenol, on CP-related perturbations in an arginine-induced rat model. Repeated l-arginine administration (5 g/kg divided into 2 doses with a 1 h interval, given intraperitoneally every 3rd day for a total of 10 times) provoked CP features, demonstrated by acinar damage, oxidative stress, inflammation, and fibrosis. Arginine-triggered pancreatitis was accompanied by hampered pancreatic autophagic flux, evidenced by overexpression of pancreatic p62 and LC3-Ⅱ and downregulation of pancreatic AMPK and LAMP-1 mRNA expression. Treatment with simvastatin (20 mg/kg, intraperitoneally 24 h, before each arginine dose) and eugenol (50 mg/kg/day orally for 30 days) achieved significant anti-oxidative, anti-inflammatory, and anti-fibrotic effects, and reversed the arginine-instigated autophagic blockade, with superior ameliorative effects attained by eugenol. Altogether, simvastatin and eugenol provide a promising interventional approach for CP, at least partly, by restoring the impaired autophagic flux associated with CP.
Collapse
Affiliation(s)
| | | | - Ahmad Mustafa Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Nancy Nabil Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
2
|
Sanati M, Pieterman I, Levy N, Akbari T, Tavakoli M, Hassani Najafabadi A, Amin Yavari S. Osteoimmunomodulation by bone implant materials: harnessing physicochemical properties and chemical composition. Biomater Sci 2025; 13:2836-2870. [PMID: 40289736 DOI: 10.1039/d5bm00357a] [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: 04/30/2025]
Abstract
Chronic inflammation at bone defect sites can impede regenerative processes, but local immune responses can be adjusted to promote healing. Regulating the osteoimmune microenvironment, particularly through macrophage polarization, has become a key focus in bone regeneration research. While bone implants are crucial for addressing significant bone defects, they are often recognized by the immune system as foreign, triggering inflammation that leads to bone resorption and implant issues like fibrous encapsulation and aseptic loosening. Developing osteoimmunomodulatory implants offers a promising approach to transforming destructive inflammation into healing processes, enhancing implant integration and bone regeneration. This review explores strategies based on tuning the physicochemical attributes and chemical composition of materials in engineering osteoimmunomodulatory and pro-regenerative bone implants.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Ines Pieterman
- Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Natacha Levy
- Metabolic Diseases Pediatrics Division, University Medical Centre Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tayebeh Akbari
- Department of Microbiology, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Mohamadreza Tavakoli
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands.
- Regenerative Medicine Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
3
|
Lecky DA, Sheriff L, Rouvray ST, George LS, Copland A, Drummond RA, Wraith DC, Bending D. Interferon-γ and IL-27 positively regulate type 1 regulatory T cell development during adaptive tolerance. iScience 2025; 28:112308. [PMID: 40276760 PMCID: PMC12018090 DOI: 10.1016/j.isci.2025.112308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/31/2024] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
Strong T cell receptor (TCR) and interleukin (IL)-27 signaling influence type 1 regulatory (Tr1) T cell development, but whether other signals determine their differentiation is unclear. Utilizing Tg4 TCR transgenic mice, we established a model for rapid Tr1 cell induction. A single high dose of [4Y]-MBP peptide drove the differentiation of Il10 + T cells with Tr1 cell mRNA and protein signatures. Kinetic transcriptional and phenotypic analyses revealed that the Tr1 cell module was transient and preceded by Ifng transcription in other CD4+ T cells. Changes in Tr1 cell frequency correlated with altered macrophage activation, while neutralization of interferon (IFN)γ reduced Tr1 cell frequency and the TCR signal strength markers Nur77, inducible T cell costimulator (ICOS), and OX40. Antibody depletion experiments inferred that the relevant source of IFNγ was not natural killer (NK) cell derived. Additionally, blocking IL-27 in combination with IFNγ neutralization additively reduced Tr1 cell frequency in vivo. These findings reveal that IFNγ has a non-redundant role in augmenting Tr1 cell differentiation in vivo.
Collapse
Affiliation(s)
- David A.J. Lecky
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Lozan Sheriff
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Sophie T. Rouvray
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Lorna S. George
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Alastair Copland
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Rebecca A. Drummond
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - David C. Wraith
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - David Bending
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
4
|
Li W, Zhang L, Xu Y, Li H, Li B, Sun S, Zhang X, Duan G, Chen Y, Zhang J, Cao Y, Li X, Liu Q, Wu Y, Zhang S, Leavenworth JW, Wang X, Zhu C. Altered monocyte subpopulations and their association with autism spectrum disorder risk in children. Brain Behav Immun 2025; 126:315-326. [PMID: 40010548 DOI: 10.1016/j.bbi.2025.02.028] [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: 07/12/2024] [Revised: 02/06/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025] Open
Abstract
OBJECTIVE Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficits in social communication, restricted interests, and repetitive behaviors. Emerging evidence suggests a link between immune dysregulation and ASD. This study investigates alterations in monocyte subpopulations and cytokine production in children with ASD and their potential associations with ASD risk and severity. METHODS Initially, the immune status of peripheral blood mononuclear cells was assessed in cohort-I of 96 typically developing (TD) children and 92 children diagnosed with ASD using flow cytometry. Subsequently, the secretion of cytokines IL-6 and IL-10 by monocytes was evaluated following stimulation with a leukocyte activation mixture and intracellular protein staining technique in cohort-II. RESULTS Children with ASD exhibited significantly higher levels of total monocytes, classical monocytes (CD14hi/CD16-), and non-classical monocytes (CD14low/CD16+) compared to TD children (p < 0.001). Elevated levels of classical monocytes (β: 0.395; 95 %CI: 0.260-0.530; p < 0.001) and non-classical monocytes (β: 0.629; 95 %CI: 0.516-0.742; p < 0.001) were significantly associated with ASD after adjusting for age, sex and body mass index. Furthermore, increased production of IL-6 by monocytes was observed in children with ASD (p = 0.001). Logistic regression analysis revealed that classical monocytes (OR: 1.104; 95 %CI: 1.062-1.147; p < 0.001), non-classical monocytes (OR: 2.913; 95 %CI: 2.130-3.986; p < 0.001) and IL-6 production by monocytes (OR: 1.306; 95 %CI: 1.096-1.557; p = 0.003) are risk factors for ASD. Spearman correlation analysis revealed a negative correlation between classical monocyte levels and adaptive behavior developmental quotient (DQ) (r = - 0.377; p = 0.001), fine motor DQ (r = - 0.329; p = 0.003) and personal-social DQ (r = - 0.247; p = 0.029) in children with ASD. CONCLUSION Elevated classical and non-classical monocytes are potential risk factors for ASD and may influence neurodevelopmental outcomes. Further research is needed to elucidate the precise mechanisms and therapeutic implications.
Collapse
Affiliation(s)
- Wenhua Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg 40530, Sweden
| | - Lingling Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hongwei Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Department of Laboratory Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Bingbing Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shuang Sun
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Guiqin Duan
- Center for Child Behavioral Development, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yiwen Chen
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jie Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yangyang Cao
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiaoping Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qianqian Liu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yanan Wu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shan Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg 40530, Sweden
| | - Jianmei W Leavenworth
- Department of Neurosurgery and Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham AL 35233, USA
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Centre of Perinatal Medicine and Health, Institute of Clinical Science, University of Gothenburg 40530 Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg 40530, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
5
|
Stacchiotti C, Mazzella di Regnella S, Cinotti M, Spalloni A, Volpe E. Neuroinflammation and Amyotrophic Lateral Sclerosis: Recent Advances in Anti-Inflammatory Cytokines as Therapeutic Strategies. Int J Mol Sci 2025; 26:3854. [PMID: 40332510 PMCID: PMC12028049 DOI: 10.3390/ijms26083854] [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: 03/18/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Neuroinflammation is an inflammatory response occurring within the central nervous system (CNS). The process is marked by the production of pro-inflammatory cytokines, chemokines, small-molecule messengers, and reactive oxygen species. Microglia and astrocytes are primarily involved in this process, while endothelial cells and infiltrating blood cells contribute to neuroinflammation when the blood-brain barrier (BBB) is damaged. Neuroinflammation is increasingly recognized as a pathological hallmark of several neurological diseases, including amyotrophic lateral sclerosis (ALS), and is closely linked to neurodegeneration, another key feature of ALS. In fact, neurodegeneration is a pathological trigger for inflammation, and neuroinflammation, in turn, contributes to motor neuron (MN) degeneration through the induction of synaptic dysfunction, neuronal death, and inhibition of neurogenesis. Importantly, resolution of acute inflammation is crucial for avoiding chronic inflammation and tissue destruction. Inflammatory processes are mediated by soluble factors known as cytokines, which are involved in both promoting and inhibiting inflammation. Cytokines with anti-inflammatory properties may exert protective roles in neuroinflammatory diseases, including ALS. In particular, interleukin (IL)-10, transforming growth factor (TGF)-β, IL-4, IL-13, and IL-9 have been shown to exert an anti-inflammatory role in the CNS. Other recently emerging immune regulatory cytokines in the CNS include IL-35, IL-25, IL-37, and IL-27. This review describes the current understanding of neuroinflammation in ALS and highlights recent advances in the role of anti-inflammatory cytokines within CNS with a particular focus on their potential therapeutic applications in ALS. Furthermore, we discuss current therapeutic strategies aimed at enhancing the anti-inflammatory response to modulate neuroinflammation in this disease.
Collapse
Affiliation(s)
- Costanza Stacchiotti
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Simona Mazzella di Regnella
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
| | - Miriam Cinotti
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
| | - Alida Spalloni
- Molecular Neurobiology Unit, Santa Lucia Foundation, 00143 Rome, Italy
| | - Elisabetta Volpe
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
| |
Collapse
|
6
|
Yeshna, Singh M, Monika, Kumar A, Garg V, Jhawat V. Pathophysiology and emerging therapeutic strategies for cervical spondylosis: The role of pro-inflammatory mediators, kinase inhibitors, and Organogel based drug delivery systems. Int Immunopharmacol 2025; 151:114350. [PMID: 40010157 DOI: 10.1016/j.intimp.2025.114350] [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: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Cervical spondylosis is a prevalent ailment characterized by chronic wear and degenerative changes affecting the cervical spine, leading to various clinical syndromes such as axial neck pain, cervical myelopathy, and cervical radiculopathy. The pathophysiology of the development of cervical alterations is multifaceted, with alterations in the normal physiology and pathogenesis of intervertebral disc degeneration. The involvement of pro-inflammatory mediators, such as interleukin-1, tumor necrosis factor-α, interleukin-4, interleukin-6, and interleukin-10, in the pathological processes associated with intervertebral disc degeneration offers potential therapeutic targets. The review also introduces kinase inhibitors as potential treatments for cervical spondylosis. Protein kinase inhibitors, including mitogen-activated protein kinase (MAPK), Janus kinase (JAK), and spleen tyrosine kinase (SYK), are explored for their anti-inflammatory properties. The article discusses their potential in modulating inflammatory signaling cascades and presents them as attractive candidates for treating immune-mediated disorders. Inhibitors of Nuclear Factor-κB, p38 MAPK, Jun-N terminal kinase (JNK), and Extracellular signal-regulated kinase (ERK) have shown efficacy in suppressing inflammatory responses, offering potential avenues for intervention in this prevalent condition. Organogels are semi-solid materials formed by trapping an organic solvent within a three-dimensional cross-linked network. They hold considerable potential in drug delivery, especially in enhancing drug solubility, facilitating controlled release, and improving skin penetration. These properties of organogels can help treat or alleviate the symptoms of cervical spondylosis.
Collapse
Affiliation(s)
- Yeshna
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Monika Singh
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Monika
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Ashok Kumar
- Faculty of Pharmacy, Kalinga University, Naya Raipur, Chhattisgarh, India
| | - Vandana Garg
- Department of Pharmaceutical Science, MD University, Rohtak, India
| | - Vikas Jhawat
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India.
| |
Collapse
|
7
|
O'Garra A. From Cytokines to Tuberculosis and Back: My Journey to Understanding the Immune Response to Infection. Annu Rev Immunol 2025; 43:1-28. [PMID: 40279305 DOI: 10.1146/annurev-immunol-010824-041601] [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] [Indexed: 04/27/2025]
Abstract
I felt honored by the invitation to write this autobiography, although it was an arduous task to describe my journey through science: first bacterial adhesion, then cytokine function, and then immune responses in tuberculosis. Since only seven women had been authors of autobiographies for the Annual Review of Immunology, I felt I couldn't refuse to contribute to Volume 43 of the journal. Moreover, this was a good occasion to record my appreciation to all the lab members and collaborators for their contributions over the last 40 years, to remember the exciting times, and to reflect on the obstacles we faced. I often reflect on this line that is commonly attributed to Winston Churchill: Success is not final; failure is not fatal: It is the courage to continue that counts. What kept me going was a burning desire to know how things work and find enjoyment in the discovery. This passion to understand immune responses to infection remains with me to this day. I thank all those I have interacted with for the support and friendship they provided.
Collapse
Affiliation(s)
- Anne O'Garra
- Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London, United Kingdom;
| |
Collapse
|
8
|
Pamela RH, Minerva MR, Ernesto CMM, Manuel MAJ, Norberto SE, Francisco AH, de la Torre Silvia MD, Angélica RL, Elva JH, Carlos NEJ, Sara O, Juan XC, Ariadnna CC, Paula FA, José AG. Is the vIL-10 Protein from Cytomegalovirus Associated with the Potential Development of Acute Lymphoblastic Leukemia? Viruses 2025; 17:435. [PMID: 40143362 PMCID: PMC11945621 DOI: 10.3390/v17030435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/01/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Leukemia is a hematologic malignancy; acute lymphoblastic leukemia (ALL) is the most prevalent subtype among children rather than in adults. Orthoherpesviridae family members produce proteins during latent infection phases that may contribute to cancer development. One such protein, viral interleukin-10 (vIL-10), closely resembles human interleukin-10 (IL-10) in structure. Research has explored the involvement of human cytomegalovirus (hCMV) in the pathogenesis of ALL. However, the limited characterization of its latent-phase proteins restricts a full understanding of the relationship between hCMV infection and leukemia progression. Studies have shown that hCMV induces an inflammatory response during infection, marked by the release of cytokines and chemokines. Inflammation may, therefore, play a role in how hCMV contributes to oncogenesis in pediatric ALL, possibly mediated by latent viral proteins. The classification of a virus as oncogenic is based on its alignment with cancer's established hallmarks. Viruses can manipulate host cellular mechanisms, causing dysregulated cell proliferation, evasion of apoptosis, and genomic instability. These processes lead to mutations, chromosomal abnormalities, and chronic inflammation, all of which are vital for carcinogenesis. This study aims to investigate the role of vIL-10 during the latent phase of hCMV as a potential factor in leukemia development.
Collapse
Affiliation(s)
- Ruvalcaba-Hernández Pamela
- Laboratorio de Virología, Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (R.-H.P.); (M.-D.d.l.T.S.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Mata-Rocha Minerva
- Unidad de Investigación en Genética Humana, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Avenida Cuauhtémoc 330, Doctores, Ciudad de México 06720, Mexico; (M.-R.M.); (S.-E.N.)
| | | | - Mejía-Aranguré Juan Manuel
- Laboratorio de Genómica Funcional del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Sánchez-Escobar Norberto
- Unidad de Investigación en Genética Humana, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Avenida Cuauhtémoc 330, Doctores, Ciudad de México 06720, Mexico; (M.-R.M.); (S.-E.N.)
- Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca City 68120, Mexico
| | - Arenas-Huertero Francisco
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico;
| | - Melchor-Doncel de la Torre Silvia
- Laboratorio de Virología, Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (R.-H.P.); (M.-D.d.l.T.S.)
| | - Rangel-López Angélica
- Laboratorio de Virología, Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (R.-H.P.); (M.-D.d.l.T.S.)
| | - Jiménez-Hernández Elva
- Departamento de Oncología, Hospital Pediátrico Moctezuma SEDESA, Universidad Autónoma Metropolitana, Mexico City 09769, Mexico;
| | - Nuñez-Enriquez Juan Carlos
- Unidad de Investigación Médica en Epidemiología Clínica, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Ochoa Sara
- Laboratorio de Bacteriología Intestinal, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (O.S.); (X.-C.J.)
| | - Xicohtencatl-Cortes Juan
- Laboratorio de Bacteriología Intestinal, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (O.S.); (X.-C.J.)
| | - Cruz-Córdova Ariadnna
- Laboratorio de Inmunoquímica, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
| | | | - Arellano-Galindo José
- Laboratorio de Virología, Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (R.-H.P.); (M.-D.d.l.T.S.)
- Centro Interdisciplinario de Ciencias de la Salud Unidad Milpa Alta Instituto Politécnico Nacional, Mexico City 12000, Mexico
| |
Collapse
|
9
|
DiDonato M, Simpson CT, Vo T, Knuth M, Geierstanger B, Jamontt J, Jones DH, Fathman JW, DeLarosa D, Junt T, Picard D, Sommer U, Bagger M, Peters E, Meeusen S, Spraggon G. A novel interleukin-10 antibody graft to treat inflammatory bowel disease. Structure 2025; 33:475-488.e7. [PMID: 39798572 DOI: 10.1016/j.str.2024.12.010] [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: 07/19/2024] [Revised: 11/19/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025]
Abstract
Inflammatory bowel disease (IBD) consists of chronic conditions that severely impact a patient's health and quality of life. Interleukin-10 (IL-10), a potent anti-inflammatory cytokine has strong genetic links to IBD susceptibility and has shown strong efficacy in IBD rodent models, suggesting it has great therapeutic potential. However, when tested in clinical trials for IBD, recombinant human IL-10 (rhIL-10) showed weak and inconsistent efficacy due to its short half-life and pro-inflammatory properties that counteract the anti-inflammatory efficacy. Here we present an engineered, IL-10, antibody-graft therapeutic (GFT-IL10M) designed to rectify these issues. GFT-IL10M combines the half-life extension properties of a monoclonal IgG antibody with altered IL-10 cell-type selective signaling, retaining desirable signaling on monocytes while reducing unwanted signaling on T, natural killer (NK), and B cells. Our structural and biochemical results indicate that the altered IL-10 topology in GFT-IL10M leads to a predominantly anti-inflammatory profile, potentially altering cell-type specific signaling patterns and extending half-life.
Collapse
Affiliation(s)
- Michael DiDonato
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Carolina Turk Simpson
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Todd Vo
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Mark Knuth
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Bernhard Geierstanger
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | | | - David H Jones
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - John W Fathman
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Donnie DeLarosa
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Tobias Junt
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Damien Picard
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Ulrike Sommer
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Morten Bagger
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Eric Peters
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Shelly Meeusen
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Glen Spraggon
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA.
| |
Collapse
|
10
|
Xue P, Wang J, Fu Y, He H, Gan Q, Liu C. Material-Mediated Immunotherapy to Regulate Bone Aging and Promote Bone Repair. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409886. [PMID: 39981851 DOI: 10.1002/smll.202409886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/19/2025] [Indexed: 02/22/2025]
Abstract
As the global population ages, an increasing number of elderly people are experiencing weakened bone regenerative capabilities, resulting in slower bone repair processes and associated risks of various complications. This review outlines the research progress on biomaterials that promote bone repair through immunotherapy. This review examines how manufacturing technologies such as 3D printing, electrospinning, and microfluidic technology contribute to enhancing the therapeutic effects of these biomaterials. Following this, it provides detailed introductions to various anti-osteoporosis drug delivery systems, such as injectable hydrogels, nanoparticles, and engineered exosomes, as well as bone tissue engineering materials and coatings used in immunomodulation. Moreover, it critically analyzes the current limitations of biomaterial-mediated bone immunotherapy and explores future research directions for material-mediated bone immunotherapy. This review aims to inspire new approaches and broaden perspectives in addressing the challenges of bone repair and aging by exploring innovative biomaterial-mediated immunotherapy strategies.
Collapse
Affiliation(s)
- Pengfei Xue
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Jiayi Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Yu Fu
- School of Aerospace Engineering and Applied Mechanics, Tongji University, Zhangwu Road 100, Shanghai, 200092, China
| | - Hongyan He
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Qi Gan
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, P. R. China
| |
Collapse
|
11
|
Casey AM, Ryan DG, Prag HA, Chowdhury SR, Marques E, Turner K, Gruszczyk AV, Yang M, Wolf DM, Miljkovic JL, Valadares J, Chinnery PF, Hartley RC, Frezza C, Prudent J, Murphy MP. Pro-inflammatory macrophages produce mitochondria-derived superoxide by reverse electron transport at complex I that regulates IL-1β release during NLRP3 inflammasome activation. Nat Metab 2025; 7:493-507. [PMID: 39972217 PMCID: PMC11946910 DOI: 10.1038/s42255-025-01224-x] [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: 05/29/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025]
Abstract
Macrophages stimulated by lipopolysaccharide (LPS) generate mitochondria-derived reactive oxygen species (mtROS) that act as antimicrobial agents and redox signals; however, the mechanism of LPS-induced mitochondrial superoxide generation is unknown. Here we show that LPS-stimulated bone-marrow-derived macrophages produce superoxide by reverse electron transport (RET) at complex I of the electron transport chain. Using chemical biology and genetic approaches, we demonstrate that superoxide production is driven by LPS-induced metabolic reprogramming, which increases the proton motive force (∆p), primarily as elevated mitochondrial membrane potential (Δψm) and maintains a reduced CoQ pool. The key metabolic changes are repurposing of ATP production from oxidative phosphorylation to glycolysis, which reduces reliance on F1FO-ATP synthase activity resulting in a higher ∆p, while oxidation of succinate sustains a reduced CoQ pool. Furthermore, the production of mtROS by RET regulates IL-1β release during NLRP3 inflammasome activation. Thus, we demonstrate that ROS generated by RET is an important mitochondria-derived signal that regulates macrophage cytokine production.
Collapse
Affiliation(s)
- Alva M Casey
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Dylan G Ryan
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Hiran A Prag
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Suvagata Roy Chowdhury
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Eloïse Marques
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Keira Turner
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Anja V Gruszczyk
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK
- Department of Surgery and Cambridge NIHR Biomedical Research Centre, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Ming Yang
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute for Metabolomics in Ageing, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
- University of Cologne, Faculty of Mathematics and Natural Sciences, Institute of Genetics, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| | - Dane M Wolf
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Jan Lj Miljkovic
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Joyce Valadares
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Patrick F Chinnery
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK
| | | | - Christian Frezza
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute for Metabolomics in Ageing, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
- University of Cologne, Faculty of Mathematics and Natural Sciences, Institute of Genetics, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| | - Julien Prudent
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
12
|
Wang Z, Sun X, Lin Y, Fu Y, Yi Z. Stealth in non-tuberculous mycobacteria: clever challengers to the immune system. Microbiol Res 2025; 292:128039. [PMID: 39752805 DOI: 10.1016/j.micres.2024.128039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/19/2025]
Abstract
Non-tuberculous Mycobacteria (NTM) are found extensively in various environments, yet most are non-pathogenic. Only a limited number of these organisms can cause various infections, including those affecting the lungs, skin, and central nervous system, particularly when the host's autoimmune function is compromised. Among these, Non-tuberculous Mycobacteria Pulmonary Diseases (NTM-PD) are the most prevalent. Currently, there is a lack of effective treatments and preventive measures for NTM infections. This article aims to deepen the comprehension of the pathogenic mechanisms linked to NTM and to formulate new intervention strategies by synthesizing current research and detailing the different tactics used by NTM to avoid elimination by the host's immune response. These intricate mechanisms not only affect the innate immune response but also successfully oppose the adaptive immune response, establishing persistent infections within the host. This includes effects on the functions of macrophages, neutrophils, dendritic cells, and T lymphocytes, as well as modulation of cytokine production. The article particularly emphasizes the survival strategies of NTM within macrophages, such as inhibiting phagosome maturation and acidification, resisting intracellular killing mechanisms, and interfering with autophagy and cell death pathways. This review aims to deepen the understanding of NTM's immune evasion mechanisms, thereby facilitating efforts to inhibit its proliferation and spread within the host, ultimately providing new methods and strategies for NTM-related treatments.
Collapse
Affiliation(s)
- Zhenghao Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Xiurong Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Yuli Lin
- School of Medical Laboratory, Shandong Second Medical University, Weifang 261053, China
| | - Yurong Fu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China.
| | - Zhengjun Yi
- School of Medical Laboratory, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
13
|
Obare LM, Simmons J, Oakes J, Zhang X, Nochowicz C, Priest S, Bailin SS, Warren CM, Mashayekhi M, Beasley HK, Shao J, Meenderink LM, Sheng Q, Stolze J, Gangula R, Absi T, Ru Su Y, Neikirk K, Chopra A, Gabriel CL, Temu T, Pakala S, Wilfong EM, Gianella S, Phillips EJ, Harrison DG, Hinton A, Kalams SA, Kirabo A, Mallal SA, Koethe JR, Wanjalla CN. CD3+ T-cell: CD14+ monocyte complexes are dynamic and increased with HIV and glucose intolerance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae054. [PMID: 40073149 PMCID: PMC11952877 DOI: 10.1093/jimmun/vkae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/07/2024] [Indexed: 03/14/2025]
Abstract
Persistent systemic inflammation is associated with an elevated risk of cardiometabolic diseases. However, the characteristics of the innate and adaptive immune systems in individuals who develop these conditions remain poorly defined. Doublets, or cell-cell complexes, are routinely eliminated from flow cytometric and other immune phenotyping analyses, which limits our understanding of their relationship to disease states. Using well-characterized clinical cohorts, including participants with controlled human immunodeficiency virus (HIV) as a model for chronic inflammation and increased immune cell interactions, we show that circulating CD14+ monocytes complexed to CD3+ T cells are dynamic, biologically relevant, and increased in individuals with diabetes after adjusting for confounding factors. The complexes form functional immune synapses with increased expression of proinflammatory cytokines and greater glucose utilization. Furthermore, in persons with HIV, the CD3+ T cell: CD14+ monocyte complexes had more HIV copies compared to matched CD14+ monocytes or CD4+ T cells alone. Our results demonstrate that circulating CD3+ T-cell: CD14+ monocyte pairs represent dynamic cellular interactions that may contribute to inflammation and cardiometabolic disease pathogenesis. CD3+ T-cell: CD14+ monocyte complexes may originate or be maintained, in part, by chronic viral infections. These findings provide a foundation for future studies investigating mechanisms linking T cell-monocyte cell-cell complexes to developing immune-mediated diseases, including HIV and diabetes.
Collapse
Affiliation(s)
- Laventa M Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Joshua Simmons
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jared Oakes
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Xiuqi Zhang
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Cindy Nochowicz
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Stephen Priest
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Samuel S Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Christian M Warren
- Veterans Affairs Flow Cytometry Core, Veterans AffairsTennessee Valley Healthcare System, Nashville, TN, United States
| | - Mona Mashayekhi
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, United States
| | - Leslie M Meenderink
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, United States
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University, Nashville, TN, United States
| | - Joey Stolze
- Department of Biostatistics, Vanderbilt University, Nashville, TN, United States
| | - Rama Gangula
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tarek Absi
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Yan Ru Su
- Department of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - Curtis L Gabriel
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tecla Temu
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Suman Pakala
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Erin M Wilfong
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sara Gianella
- Division of Infectious Diseases, University of California, San Diego, CA, United States
| | - Elizabeth J Phillips
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - David G Harrison
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Spyros A Kalams
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Simon A Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, United States
| | - John R Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, United States
| | - Celestine N Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
14
|
Fey RM, Billo A, Clister T, Doan KL, Berry EG, Tibbitts DC, Kulkarni RP. Personalization of Cancer Treatment: Exploring the Role of Chronotherapy in Immune Checkpoint Inhibitor Efficacy. Cancers (Basel) 2025; 17:732. [PMID: 40075580 PMCID: PMC11899640 DOI: 10.3390/cancers17050732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/01/2025] [Accepted: 02/15/2025] [Indexed: 03/14/2025] Open
Abstract
In the era of precision medicine, mounting evidence suggests that the time of therapy administration, or chronotherapy, has a great impact on treatment outcomes. Chronotherapy involves planning treatment timing by considering circadian rhythms, which are 24 h oscillations in behavior and physiology driven by synchronized molecular clocks throughout the body. The value of chronotherapy in cancer treatment is currently under investigation, notably in the effects of treatment timing on efficacy and side effects. Immune checkpoint inhibitor (ICI) therapy is a promising cancer treatment. However, many patients still experience disease progression or need to stop the therapy early due to side effects. There is accumulating evidence that the time of day at which ICI therapy is administered can have a substantial effect on ICI efficacy. Thus, it is important to investigate the intersections of circadian rhythms, chronotherapy, and ICI efficacy. In this review, we provide a brief overview of circadian rhythms in the context of immunity and cancer. Additionally, we outline current applications of chronotherapy for cancer treatment. We synthesize the 29 studies conducted to date that examine the impact of time-of-day administration on the efficacy of ICI therapy, its associated side effects, and sex differences in both efficacy and side effects. We also discuss potential mechanisms underlying these observed results. Finally, we highlight the challenges in this area and future directions for research, including the potential for a chronotherapeutic personalized medicine approach that tailors the time of ICI administration to individual patients' circadian rhythms.
Collapse
Affiliation(s)
- Rosalyn M. Fey
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Avery Billo
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Terri Clister
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Khanh L. Doan
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Elizabeth G. Berry
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Deanne C. Tibbitts
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rajan P. Kulkarni
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Portland, OR 97239, USA
- Operative Care Division, U.S. Department of Veterans Affairs Portland Health Care System, Portland, OR 97239, USA
| |
Collapse
|
15
|
Ruiz VY, Calderon TM, Leon-Rivera R, Chilunda V, Zhang J, Berman JW. Single-cell analysis of CD14 +CD16 + monocytes identifies a subpopulation with an enhanced migratory and inflammatory phenotype. Front Immunol 2025; 16:1475480. [PMID: 40051633 PMCID: PMC11883828 DOI: 10.3389/fimmu.2025.1475480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 01/28/2025] [Indexed: 03/09/2025] Open
Abstract
Monocytes in the central nervous system (CNS) play a pivotal role in surveillance and homeostasis, and can exacerbate pathogenic processes during injury, infection, or inflammation. CD14+CD16+ monocytes exhibit diverse functions and contribute to neuroinflammatory diseases, including HIV-associated neurocognitive impairment (HIV-NCI). Analysis of human CD14+CD16+ monocytes matured in vitro by single-cell RNA sequencing identified a heterogenous population of nine clusters. Ingenuity pathway analysis of differentially expressed genes in each cluster identified increased migratory and inflammatory pathways for a group of clusters, which we termed Group 1 monocytes. Group 1 monocytes, distinguished by increased ALCAM, CD52, CD63, and SDC2, exhibited gene expression signatures implicated in CNS inflammatory diseases, produced higher levels of CXCL12, IL-1Ra, IL-6, IL-10, TNFα, and ROS, and preferentially transmigrated across a human in vitro blood-brain barrier model. Thus, Group 1 cells within the CD14+CD16+ monocyte subset are likely to be major contributors to neuroinflammatory diseases.
Collapse
Affiliation(s)
- Vanessa Y. Ruiz
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, United States
| | - Tina M. Calderon
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, United States
| | - Rosiris Leon-Rivera
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, United States
| | - Vanessa Chilunda
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, United States
| | - Jinghang Zhang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, United States
| | - Joan W. Berman
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
16
|
Cocksedge SP, Mantecón L, Castaño E, Infante C, Bailey SJ. The Potential of Superoxide Dismutase-Rich Tetraselmis chuii as a Promoter of Cellular Health. Int J Mol Sci 2025; 26:1693. [PMID: 40004157 PMCID: PMC11855123 DOI: 10.3390/ijms26041693] [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: 01/19/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Tetraselmis chuii (T. chuii) is a green, marine, eukaryotic, microalgae that was authorized in the European Union (EU) as a novel food for human consumption in 2014, and as a food supplement in 2017. This narrative review will provide an overview of preclinical and clinical trials assessing the efficacy of a T. chuii-derived ingredient, characterized by a high superoxide dismutase (SOD) activity (SOD-rich T. chuii), to improve various aspects of cellular health. Collectively, results from in vitro, and more importantly in vivo research, support SOD-rich T. chuii as a potential promoter of cellular health. Principally, the ingredient appears to function as an indirect antioxidant by boosting intracellular antioxidant systems. Moreover, it can positively modulate inflammatory status by up-regulating anti-inflammatory and down-regulating pro-inflammatory cytokines and factors. In addition, SOD-rich T. chuii appears to promote cellular health though protecting from DNA damage, boosting immune function, strengthening cell structure and integrity, and positively modulating cell signaling pathways. There is also some evidence to suggest that SOD-rich T. chuii may improve aspects of mitochondrial function through the up-regulation of genes linked to mitochondrial biogenesis and ATP synthesis. From the trials conducted to date, transcriptional activation of nuclear factor erythroid 2-related factor 2 (NRF2) and sirtuin 1 (SIRT1) appear to be important in mediating the effects of SOD-rich T. chuii on cellular health. These exciting preliminary observations suggest that SOD-rich T. chuii may represent a natural blue food supplement with the potential to enhance various aspects of cellular health.
Collapse
Affiliation(s)
- Stuart P. Cocksedge
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK;
- Centre for Physical Activity, Sport and Exercise Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Lalia Mantecón
- Fitoplancton Marino, S.L., Dársena Comercial s/n, 11500 El Puerto de Santa María, Cádiz, Spain; (L.M.); (E.C.); (C.I.)
| | - Enrique Castaño
- Fitoplancton Marino, S.L., Dársena Comercial s/n, 11500 El Puerto de Santa María, Cádiz, Spain; (L.M.); (E.C.); (C.I.)
| | - Carlos Infante
- Fitoplancton Marino, S.L., Dársena Comercial s/n, 11500 El Puerto de Santa María, Cádiz, Spain; (L.M.); (E.C.); (C.I.)
| | - Stephen J. Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK;
| |
Collapse
|
17
|
Diao YF, Chen ZB, Gu JX, Xu XY, Lin WF, Yuan CZ, Xiong JQ, Li MH, Ni BQ, Zhao S, Shao YF, Zhang YY, Liu H. Incorporating Circulating Plasma Interleukin-10 Enhanced Risk Predictability of Mortality in Acute Type A Aortic Dissection Surgery. Rev Cardiovasc Med 2025; 26:26334. [PMID: 40026520 PMCID: PMC11868896 DOI: 10.31083/rcm26334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/13/2024] [Accepted: 10/30/2024] [Indexed: 03/05/2025] Open
Abstract
Background Acute type A aortic dissection (TAAD) is a life-threatening cardiovascular emergency with a high mortality rate. The peri-operative factors influencing in-hospital mortality among surgically treated TAAD patients remain unclear. This study aimed to identify key peri-operative risk factors associated with in-hospital mortality. Methods Peri-operative laboratory data, surgical strategies, and TAAD-related risk factors, associated with mortality, were collected. Machine learning techniques were applied to evaluate the impact of various parameters on in-hospital mortality. Based on the findings, a nomogram model was developed and validated using area under the receiver operating characteristic curve (AUC) analysis, calibration plots, and internal validation methods. Results A total of 199 patients with TAAD were included in the study cohort, which was divided into derivation and validation cohorts. Using the least absolute shrinkage and selection operator (LASSO) regression method, 66 features were narrowed down to six key predictors. These included age, lymphocyte count, use of continuous renal replacement therapy (CRRT), cardiopulmonary bypass (CPB) time, duration of mechanical ventilation, and postoperative interleukin-10 (IL-10) levels, all of which were identified as significant risk factors for in-hospital mortality following TAAD surgery. Conclusions We developed and validated a predictive model, presented as a nomogram, to estimate in-hospital survival in patients with TAAD. Post-operative IL-10 was identified as an independent prognostic factor for patients with TAAD. The combination of IL-10 with five additional indicators significantly improved the predictive accuracy, demonstrating superiority over the use of any single variable alone. Clinical Trial Registration This study protocol was registered at ClinicalTrials.gov (NCT04711889). https://clinicaltrials.gov/study/NCT04711889.
Collapse
Affiliation(s)
- Yi-fei Diao
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Zhi-bin Chen
- Department of Cardiovascular Surgery, First Affiliated Hospital of Guangzhou Medical University, 510120 Guangzhou, Guangdong, China
| | - Jia-xi Gu
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Xin-yang Xu
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Wen-feng Lin
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Chun-ze Yuan
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Jia-qi Xiong
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Ming-hui Li
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Bu-qing Ni
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Sheng Zhao
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Yong-feng Shao
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| | - Ying-yuan Zhang
- Department of Cardiovascular Surgery, First Affiliated Hospital of Guangzhou Medical University, 510120 Guangzhou, Guangdong, China
| | - Hong Liu
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, 210029 Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Liu L, Chen H, Zhao X, Han Q, Xu Y, Liu Y, Zhang A, Li Y, Zhang W, Chen B, Wang J. Advances in the application and research of biomaterials in promoting bone repair and regeneration through immune modulation. Mater Today Bio 2025; 30:101410. [PMID: 39811613 PMCID: PMC11731593 DOI: 10.1016/j.mtbio.2024.101410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/02/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
With the ongoing development of osteoimmunology, increasing evidence indicates that the local immune microenvironment plays a critical role in various stages of bone formation. Consequently, modulating the immune inflammatory response triggered by biomaterials to foster a more favorable immune microenvironment for bone regeneration has emerged as a novel strategy in bone tissue engineering. This review first examines the roles of various immune cells in bone tissue injury and repair. Then, the contributions of different biomaterials, including metals, bioceramics, and polymers, in promoting osteogenesis through immune regulation, as well as their future development directions, are discussed. Finally, various design strategies, such as modifying the physicochemical properties of biomaterials and integrating bioactive substances, to optimize material design and create an immune environment conducive to bone formation, are explored. In summary, this review comprehensively covers strategies and approaches for promoting bone tissue regeneration through immune modulation. It offers a thorough understanding of current research trends in biomaterial-based immune regulation, serving as a theoretical reference for the further development and clinical application of biomaterials in bone tissue engineering.
Collapse
Affiliation(s)
- Li Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Hao Chen
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Xue Zhao
- Department of Endocrinology, The First Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Qing Han
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Yongjun Xu
- Department of Orthopedics Surgery, Wangqing County People's Hospital, Yanbian, 133000, Jilin, China
| | - Yang Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Aobo Zhang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Yongyue Li
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Weilong Zhang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Bingpeng Chen
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Jincheng Wang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| |
Collapse
|
19
|
Zhao W, Lu J, Yan H, Zhu J, Liu Y, Song X, Suo T, Miao L. Treatment of acute pharyngitis in rats with season tea decoctions from traditional Chinese medicine through a synergistic and subtle regulation of ARNTL and BHLHE40. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118924. [PMID: 39389396 DOI: 10.1016/j.jep.2024.118924] [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: 06/03/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE While the seasonal variations in the human immune function and many infectious diseases are well-known, to develop therapeutic strategies regarding such seasonality is quite challenging. However, some traditional medical practices have already taken the seasonality into account, such as the "Season Tea" (ST) decoctions investigated in the present study. AIM OF THE STUDY We present a study of the ST decoctions from traditional Chinese medicine, which include four formulae designed for the four seasons, aiming to investigate their pharmacological commonality and distinction. MATERIALS AND METHODS A rat model of acute pharyngitis was utilized for the pharmacological study, and the effects of the ST decoctions were evaluated through histology, biomedical assays, microarray analysis, real-time quantitative PCR and Western blot. RESULTS The experimental data show that all of the four ST formulae display good pharmaceutical effects on acute pharyngitis, and circadian rhythm appears to be a significant pathway for investigating their pharmacological commonality and distinction. Specifically, while all of the four ST decoctions can regulate the circadian-rhythm-related genes ARNTL and BHLHE40, the regulation is along different directions with the modification of the supplements and the substrates in each ST formula. CONCLUSION These results indicate the correlation between the acute pharyngitis and circadian rhythm, and illustrate the possibility of synergistically and subtly regulating ARNTL and BHLHE40, which is significant for relevant drug development.
Collapse
Affiliation(s)
- Wei Zhao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China
| | - Jia Lu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Huimin Yan
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Junjie Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yang Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xinbo Song
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tongchuan Suo
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Chinese Medicine Modernization, Tianjin, 301617, China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Lin Miao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Chinese Medicine Modernization, Tianjin, 301617, China.
| |
Collapse
|
20
|
Robertson FP, Cuff AO, Male V, Wright GP, Pallett LJ, Fuller BJ, Davidson BR. Inflammatory Monocytes Are Rapidly Recruited to the Post-Ischaemic Liver in Patients Undergoing Liver Transplantation and Cytokines Associated with Their Activation Correlate with Graft Outcomes. Curr Issues Mol Biol 2025; 47:49. [PMID: 39852164 PMCID: PMC11763458 DOI: 10.3390/cimb47010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/31/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Liver ischaemia-reperfusion (IR) injury remains a major cause of morbidity and mortality following liver transplantation and resection. CD4+ T cells have been shown to play a key role in murine models; however, there is currently a lack of data that support their role in human patients. Methods: Data on clinical outcomes and complications were documented prospectively in 28 patients undergoing first elective liver transplant surgery. Peripheral blood samples were collected at baseline (pre-op), 2 h post graft reperfusion, immediately post-op, and 24 h post-op. A post-reperfusion biopsy was analysed in all patients, and in five patients, a donor liver biopsy was available pre-implantation. Circulating cytokines were measured, and T cells were analysed for activation markers and cytokine production. Results: Circulating levels of cytokines associated with innate immune cell recruitment and activation were significantly elevated in the peri-transplant period. High circulating IL-10 levels corresponded with the development of graft-specific complications. The proportion of CD4+ T cells in the peripheral circulation fell throughout the peri-operative period, suggesting CD4+ T cell recruitment to the graft. Although TNFα was the predominant cytokine produced by CD4+ T cells in the intrahepatic environment, the production of IFNγ was significantly upregulated by circulating CD4+ T cells. Furthermore, we demonstrated clear recruitment of inflammatory monocytes in the peri-operative period. In donor-and-recipient pairs with a mismatch at the HLA-A2 or A3 allele, we demonstrated that inflammatory monocytes in the liver are recipient-derived. Discussion: This is the first study to our knowledge that tracks early immune cell responses in humans undergoing liver transplantation. The recruitment of inflammatory monocytes from the recipient and their cytokine release is associated with liver-specific complications. Inflammatory monocytes would be an attractive target to ameliorate ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- Francis P. Robertson
- Division of Interventional and Surgical Science, Royal Free Campus, University College London, London NW3 2QG, UK; (B.J.F.); (B.R.D.)
- Department of Surgery, School of Medicine, Gilmorehill Campus, University of Glasgow Medical School, Glasgow G12 8QQ, UK
| | - Antonia O. Cuff
- Division of Biomedical Sciences, Warwick Medical School, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK;
| | - Victoria Male
- Department of Metabolism, Digestion and Reproduction, Chelsea and Westminster Hospital Campus, Imperial College London, London W12 0NN, UK;
| | - Graham P. Wright
- School of Applied Science, Edinburgh Napier University, Edinburgh EH11 4BN, UK;
| | - Laura J. Pallett
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK;
| | - Barry J. Fuller
- Division of Interventional and Surgical Science, Royal Free Campus, University College London, London NW3 2QG, UK; (B.J.F.); (B.R.D.)
| | - Brian R. Davidson
- Division of Interventional and Surgical Science, Royal Free Campus, University College London, London NW3 2QG, UK; (B.J.F.); (B.R.D.)
- Department of HPB and Liver Transplant Surgery, Royal Free Hospital, London NW3 2QG, UK
| |
Collapse
|
21
|
Mishra V, Baranwal V, Mugale MN, Sharma S, Mishra RK. Stat3 Induces IL-10 and SR-A/CD204 Expression in Silica Nanoparticle-Triggered Pulmonary Fibrosis through Transactivation. ACS Biomater Sci Eng 2025; 11:609-622. [PMID: 39643585 DOI: 10.1021/acsbiomaterials.4c01473] [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] [Indexed: 12/09/2024]
Abstract
Inhalation of silica dust in the workplace has been addressed as a serious occupational pulmonary disease subsequently leading to inflammation and fibrosis. Enhanced expression of IL-10 significantly contributes to the disease etiology, along with an elevated Th2-type paradigm. Previously, we showed that the exaggerated Th2-type response was also associated with consistent upregulation of Stat3 in mouse airways stimulated with silica microparticles. However, a precise understanding of silicosis in light of the IL-10/Stat3 immune axis is required. We, therefore, aimed to determine the regulatory role of IL-10 in nanosized silica (nSiO2)-induced pulmonary fibrosis in association with Stat3. Herein, we report that amorphous nSiO2 could induce pulmonary fibrosis with consistent and concomitant upregulation of IL-10, Stat3, and SR-A/CD204. Following exogenous administration of siStat3 and rIL-10, the study further confirmed that Stat3 mediates the regulation of IL-10 and SR-A/CD204 and that IL-10 could regulate its own expression in an autoregulatory loop. The ChIP assay highlighted the localization of Stat3 over two putative binding sites in the IL-10 promoter region, which subsequently resulted in the overexpression of SR-A/CD204. Conclusively, Stat3-mediated transregulation of IL-10 through an autoregulatory loop in silicosis could offer novel molecular targets for therapeutic interventions.
Collapse
Affiliation(s)
- Vani Mishra
- Department of Biotechnology, Motilal Nehru National Institute of Technology (MNNIT), Prayagraj 211004, India
| | - Vikas Baranwal
- Toshniwal Brothers (SR) Pvt. Ltd., 11, AECS Layout, Sanjay Nagar, Bengaluru, Karnataka 560094, India
| | - Madhav Nilakanth Mugale
- Department of Toxicology and Experimental Medicine, CSIR─Central Drug Research Institute (CDRI), Lucknow 226031, India
| | - Shivesh Sharma
- Department of Biotechnology, Motilal Nehru National Institute of Technology (MNNIT), Prayagraj 211004, India
| | - Rohit Kumar Mishra
- Centre of Science and Society, Institute of Interdisciplinary Sciences (IIDS), University of Allahabad, Prayagraj 211002, India
| |
Collapse
|
22
|
Chegini Z, Khoshbayan A, Kashi M, Zare Shahraki R, Didehdar M, Shariati A. The possible pathogenic mechanisms of microorganisms in infertility: a narrative review. Arch Microbiol 2025; 207:27. [PMID: 39777552 DOI: 10.1007/s00203-024-04231-w] [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: 11/12/2024] [Revised: 12/10/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025]
Abstract
Infertility can harm a patient in physical, psychological, spiritual, and medical ways. This illness is unusual because it affects the patient's companion and the patient individually. Infertility is a multifactorial disease, and various etiological factors like infection are known to develop this disorder. Recently published studies reported that different bacteria, such as Chlamydia trachomatis, Mycoplasma spp., Ureaplasma urealyticum, Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa, can lead to infertility by immunopathological effects, oxidative stress, and adverse effects on sperm concentration, motility, morphology, and DNA condensation. Among viruses, Human papillomavirus and Herpes simplex virus reduce sperm progressive motility and sperm concentration. The viruses can lead to the atrophy of the germinal epithelium and degenerative changes in the testes. Candida albicans also harm sperm quality, motility, and chromatin integrity and induce apoptosis in sperm cells. Finally, Trichomonas vaginalis leads to distorted heads, broken necks, and acrosomes exocytosis in sperms. This parasite decreases sperm viability and functional integrity. Noteworthy, oxidative stress could have a role in many pathological changes in the reproductive system. Recent findings show that microorganisms can increase reactive oxygen species concentration inside the host cells, leading to oxidative stress and sperm distress and dysfunction. Therefore, this article explores the potential significance of critical bacteria linked to infertility and their pathogenic mechanisms that can affect sperm function and the female reproductive system.
Collapse
Affiliation(s)
- Zahra Chegini
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amin Khoshbayan
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Milad Kashi
- Student Research Committee, Arak University of Medical Sciences, Arak, Iran
| | - Raha Zare Shahraki
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mojtaba Didehdar
- Department of Medical Parasitology and Mycology, Arak University of Medical Sciences, Arak, Iran
| | - Aref Shariati
- Infectious Diseases Research Center (IDRC), Arak University of Medical Sciences, Arak, Iran.
| |
Collapse
|
23
|
Erlandson G, Magzamen S, Sharp JL, Seidel J, Poole JA, Bradford M, Schaeffer JW. Hypertonic Saline Nasal Rinse Intervention: Immunomodulatory Effects in Dairy Workers. J Agromedicine 2025; 30:27-37. [PMID: 39440411 DOI: 10.1080/1059924x.2024.2416425] [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] [Indexed: 10/25/2024]
Abstract
OBJECTIVE Increased risk of occupational exposure to bioaerosols has long been recognized in livestock operations including dairy facilities. Spanning the inhalable fraction (0-100 μm), dairy bioaerosols comprise a wide variety of inflammatory components that deposit in the nasopharyngeal region. The resultant inflammatory response from bioaerosol exposure is likely driving the increased prevalence of respiratory disease observed in dairy workers. It is also thought the microbiome of the upper respiratory system may help mediate this inflammation. We investigated the viability of a low-cost hypertonic saline nasal rinse intervention in modulating inflammatory responses in bioaerosol exposed dairy workers and its impact on microbial diversity. METHODS Pre- and post-shift nasal rinses were administered and collected alongside full shift inhalable personal breathing zone (PBZ) samples for each participant for up to 5 consecutive days. Treatment group participants (n = 23) received hypertonic saline rinses while control group participants (n = 22) received normotonic saline rinses. Particulate matter (PM) and endotoxin concentrations were quantified from PBZ samples using gravimetric and enzymatic analytical methods, respectively. Pre- and post-shift rinses were analyzed for pro- and anti-inflammatory markers and microbial diversity using a multiplex assay and 16S rRNA sequencing, respectively. RESULTS PM and endotoxin concentrations were comparable between groups indicating similar exposures. Post-shift pro-inflammatory markers were significantly higher than pre-shift for IL-13 (p = .047), IL-1β (p < .001), IL-6 (p < .001), IL-8 (p < .001), and TNF-α (p = .024). There was no evidence of a difference in log concentrations between intervention group or day among any of the measured inflammatory markers. Anti-inflammatory IL-10 concentrations increased across the 5 sample days, independent of treatment group suggesting tonicity may not be driving the change. However, this result was not significant (p = .217). Nasal microbiome alpha (within sample) and beta (between sample) diversity metrics did not differ significantly between group or day demonstrating no adverse washout intervention effects. CONCLUSION This study provided encouraging results that warrant future research to further evaluate saline nasal rinses as a workplace intervention.
Collapse
Affiliation(s)
- Grant Erlandson
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Sheryl Magzamen
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Julia L Sharp
- Department of Human Development and Family Studies, Colorado State University, Fort Collins, CO, USA
- Sharp Analytics LLC, Fort Collins, CO, USA
| | - James Seidel
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Jill A Poole
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mary Bradford
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Joshua W Schaeffer
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Environmental and Occupational Health, Colorado School of Public Health, Denver, CO, USA
| |
Collapse
|
24
|
Wang M, Zhou S, Hu Y, Tong W, Zhou H, Ma M, Cai X, Zhang Z, Zhang L, Chen Y. Macrophages overexpressing interleukin-10 target and prevent atherosclerosis: Regression of plaque formation and reduction in necrotic core. Bioeng Transl Med 2025; 10:e10717. [PMID: 39801756 PMCID: PMC11711221 DOI: 10.1002/btm2.10717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/27/2024] [Accepted: 08/08/2024] [Indexed: 01/16/2025] Open
Abstract
Atherosclerosis, a slowly progressing inflammatory disease, is characterized by the presence of monocyte-derived macrophages. Interventions targeting the inflammatory characteristics of atherosclerosis hold promising potential. Although interleukin (IL)-10 is widely acknowledged for its anti-inflammatory effects, systemic administration of IL-10 has limitations due to its short half-life and significant systemic side effects. In this study, we aimed to investigate the effectiveness of an approach designed to overexpress IL-10 in macrophages and subsequently introduce these genetically modified cells into ApoE-/- mice to promote atherosclerosis regression. We engineered RAW264.7 cells to overexpress IL-10 (referred to as IL-10M) using lentivirus vectors. The IL-10M exhibited robust IL-10 secretion, maintained phagocytic function, improved mitochondrial membrane potentials, reduced superoxide production and showed a tendency toward the M2 phenotype when exposed to inflammatory stimuli. IL-10M can selectively target plaques in ApoE-/- mice and has the potential to reduce plaque area and necrotic core at both early and late stages of plaque progression. Moreover, there was a significant reduction in MMP9, a biomarker associated with plaque rupture, in IL-10M-treated plaques from both the early and late intervention groups. Additionally, the administration of IL-10M showed no obvious side effects. This study serves as proof that cell therapy based on anti-inflammatory macrophages might be a promising strategy for the intervention of atherosclerosis.
Collapse
Affiliation(s)
- Mingyi Wang
- Medical School of Chinese PLABeijingChina
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
| | - Shanshan Zhou
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- Department of CardiologyThe First Medical Center of PLA General HospitalBeijingChina
| | - Yingyun Hu
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- The Medical School of Nankai UniversityTianjinChina
| | - Wei Tong
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- Department of CardiologyThe First Medical Center of PLA General HospitalBeijingChina
| | - Hao Zhou
- Department of CardiologyNo. 966 Hospital of Joint Logisties ForceDandongChina
| | - Mingrui Ma
- Medical School of Chinese PLABeijingChina
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
| | - Xingxuan Cai
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- The Second Medical School of Southern Medical UniversityGuangzhouChina
| | - Zhengbin Zhang
- Medical School of Chinese PLABeijingChina
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
| | - Luo Zhang
- Medical School of Chinese PLABeijingChina
- Research Center of BioengineeringThe Medical Innovation Research Division of PLA General HospitalBeijingChina
| | - Yundai Chen
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- Department of CardiologyThe First Medical Center of PLA General HospitalBeijingChina
| |
Collapse
|
25
|
Avecilla V, Doke M, Appunni S, Rubens M, Ramamoorthy V, Das JK. Pathophysiological Features of Remodeling in Vascular Diseases: Impact of Inhibitor of DNA-Binding/Differentiation-3 and Estrogenic Endocrine Disruptors. Med Sci (Basel) 2024; 13:2. [PMID: 39846697 PMCID: PMC11755649 DOI: 10.3390/medsci13010002] [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: 09/12/2024] [Revised: 12/12/2024] [Accepted: 12/22/2024] [Indexed: 01/30/2025] Open
Abstract
Vascular diseases, such as hypertension, atherosclerosis, cerebrovascular, and peripheral arterial diseases, present major clinical and public health challenges, largely due to their common underlying process: vascular remodeling. This process involves structural alterations in blood vessels, driven by a variety of molecular mechanisms. The inhibitor of DNA-binding/differentiation-3 (ID3), a crucial member of ID family of transcriptional regulators, has been identified as a key player in vascular biology, significantly impacting the progression of these diseases. This review explores the role of ID3 in vascular remodeling, emphasizing its involvement in processes such as apoptosis, cell proliferation, and extracellular matrix regulation. Furthermore, we examine how oxidative stress, intensified by exposure to estrogenic endocrine disruptors (EEDs) like polychlorinated biphenyls (PCBs) and bisphenol A (BPA), affects ID3 activity and contributes to vascular disease. Understanding the interaction between ID3 signaling and EED exposure provides critical insights into the molecular mechanisms underlying vascular remodeling and its role in the development and progression of vascular diseases.
Collapse
Affiliation(s)
- Vincent Avecilla
- Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA
- Avecilla Consulting LLC, Miami, FL 33131, USA
| | - Mayur Doke
- Diabetes Research Institute, University of Miami, Miami, FL 33136, USA
| | - Sandeep Appunni
- Department of Biochemistry, Government Medical College, Kozhikode 673008, Kerala, India
| | - Muni Rubens
- Baptist Health South Florida, Miami Gardens, FL 33176, USA
| | | | - Jayanta Kumar Das
- Department of Health and Natural Sciences, Florida Memorial University, Miami Gardens, FL 33054, USA
| |
Collapse
|
26
|
Jin X, Li H, Pan S, Song B, Jiang Y, Shi H, Zhang J, Chu B, Wang H, He Y. DNA Nanopatch-Specific Modification of Probiotics for Ultrasound-Triggered Inflammatory Bowel Disease Therapy. J Am Chem Soc 2024; 146:33817-33831. [PMID: 39508560 DOI: 10.1021/jacs.4c12139] [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: 11/15/2024]
Abstract
Probiotics offer promising results for treating inflammatory bowel disease, yet precision therapy remains challenging, particularly in manipulating probiotics spatially and temporally and shielding them from oxidative stress. To address these limitations, herein we synthesized bacteria-specific DNA nanopatches to modify ultrasound-triggered engineered Escherichia coli Nissle 1917. These probiotics produced the anti-inflammatory cytokine interleukin-10 when stimulated by ultrasound and were fortified with DNPs for oxidative stress resistance. The DNPs were composed of rectangular DNA origami nanosheets with reactive oxygen species' scavenging ability and bacterial targeting ligands of maltodextrin molecules. We systematically demonstrated that the DNPs could selectively attach to bacterial surface but not mammalian cell surface via the maltodextrin transporter pathway. To further enhance the bioavailability of engineered probiotics in the gastrointestinal tract, we employed a self-assembly strategy to encapsulate them using chitosan and sodium alginate. In a murine model of ulcerative colitis, this system significantly improved the gut barrier integrity and reduced inflammation. Our results indicate that this DNA nanopatch-bacteria system holds substantial promise for mitigating oxidative stress, correcting microbiota dysbiosis, and enhancing the intestinal barrier function in colitis.
Collapse
Affiliation(s)
- Xiangbowen Jin
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Hongyang Li
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Sheng Pan
- Department of Orthopaedics, Second Affiliated Hospital of Soochow University, Osteoporosis Research Institute of Soochow University, Suzhou 215000, China
| | - Bin Song
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Yanping Jiang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Haoliang Shi
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Jiawei Zhang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Binbin Chu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Houyu Wang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
| | - Yao He
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123, China
- Macao Translational Medicine Center, Macau University of Science and Technology, Taipa 999078, Macau SAR, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa 999078, Macau SAR, China
| |
Collapse
|
27
|
Relouw FJA, Kox M, Taal HR, Koch BCP, Prins MWJ, van Riel NAW. Mathematical model of the inflammatory response to acute and prolonged lipopolysaccharide exposure in humans. NPJ Syst Biol Appl 2024; 10:146. [PMID: 39638779 PMCID: PMC11621538 DOI: 10.1038/s41540-024-00473-y] [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: 01/15/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
One in five deaths worldwide is associated with sepsis, which is defined as organ dysfunction caused by a dysregulated host response to infection. An increased understanding of the pathophysiology of sepsis could provide improved approaches for early detection and treatment. Here we describe the development and validation of a mechanistic mathematical model of the inflammatory response, making use of a combination of in vitro and human in vivo data obtained from experiments where bacterial lipopolysaccharide (LPS) was used to induce an inflammatory response. The new model can simulate the responses to both acute and prolonged inflammatory stimuli in an experimental setting, as well as the response to infection in the clinical setting. This model serves as a foundation for a sepsis simulation model with a potentially wide range of applications in different disciplines involved with sepsis research.
Collapse
Affiliation(s)
- Freek J A Relouw
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands.
- Department of Neonatal and Paediatric Intensive Care, Division of Neonatology, Erasmus University Medical Center, Rotterdam, The Netherlands.
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands.
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
- Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - H Rob Taal
- Department of Neonatal and Paediatric Intensive Care, Division of Neonatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Menno W J Prins
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
- Department of Applied Physics, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Natal A W van Riel
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
- Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
28
|
Jha J, Singh MK, Singh L, Pushker N, Kakkar A, Meel R, Lomi N, Bakhshi S, Nag TC, Panwar C, Sen S, Kashyap S. Deciphering the Intricate Relationship Between Macrophages, Pigmentation, and Prognosis in Uveal Melanoma. J Transl Med 2024; 104:102167. [PMID: 39491651 DOI: 10.1016/j.labinv.2024.102167] [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: 06/19/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024] Open
Abstract
High pigmentation and the abundance of M2 macrophages have been identified as negative predictors in uveal melanoma (UM). Risk factors associated with UM that are prevalent in high-risk White populations are still present, although less common, in relatively low-risk Asian populations. Research indicates that proangiogenic M2 macrophages and monosomy 3 play significant roles in UM progression. Our aim was to investigate the impact of tumor-associated macrophages in UM and examine their correlation with monosomy 3 and pigmentation. Transmission electron microscopy was used to analyze the morphology of macrophages in UM. Forty UM samples underwent fluorescent in situ hybridization for monosomy 3 identification. Immunohistochemistry was done to assess M2/M1 macrophages on 82 UM tissue samples. IL-10 and IL-12 expressions were quantified in UM serum samples by enzyme-linked immunosorbent assay. The expression of all markers was correlated with pigmentation markers (tyrosinase-related protein 1, tyrosinase-related protein 2, silver protein, and microphthalmia-associated transcription factor). Prognostic outcomes were determined using the Cox proportional hazard model and log-rank tests. Increased expression of M2/M1 macrophages was observed in 31 UM cases, which correlated with the high expression of pigmentation markers. IL-10 concentration was high in UM cases. Monosomy 3 was evident in 50% of UM cases and significantly associated with increased immunoexpression of M2/M1 macrophages and pigmentation markers. Reduced metastasis-free survival was observed in patients with UM with high M2/M1 macrophage expression (P = .001). High pigmentation and increased M2 macrophage density could impact the tumor microenvironment in UM. This could contribute to ineffective antitumor immune responses in patients with UM. Our findings suggest avenues for developing novel therapeutic approaches to counteract these immunosuppressive effects in UM.
Collapse
Affiliation(s)
- Jayanti Jha
- Department of Ocular Pathology, Dr R.P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Delhi, India
| | | | - Lata Singh
- Department of Paediatrics, All India Institute of Medical Sciences, Delhi, India
| | - Neelam Pushker
- Department of Ophthalmology, Dr R.P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Delhi, India
| | - Aanchal Kakkar
- Department of Pathology, All India Institute of Medical Sciences, Delhi, India
| | - Rachna Meel
- Department of Ophthalmology, Dr R.P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Delhi, India
| | - Neiwete Lomi
- Department of Ophthalmology, Dr R.P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, All India Institute of Medical Sciences, Delhi, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, Delhi, India
| | - Chanda Panwar
- Department of Anatomy, All India Institute of Medical Sciences, Delhi, India
| | - Seema Sen
- Department of Ocular Pathology, Dr R.P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Delhi, India
| | - Seema Kashyap
- Department of Ocular Pathology, Dr R.P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Delhi, India.
| |
Collapse
|
29
|
Arreza L, Thanasupawat T, Krishnan SN, Kraljevic M, Klonisch T, Hombach-Klonisch S. C1QTNF Related protein 8 (CTRP8) is a marker of myeloid derived innate immune cell populations in the human breast cancer microenvironment. Biochem Pharmacol 2024; 230:116624. [PMID: 39542181 DOI: 10.1016/j.bcp.2024.116624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Innate immune cells in the tumor microenvironment (TME) play an important role in breast cancer (BC) metastatic spread and influence patient survival. Macrophages differentiate along a proinflammatory M1 to protumorigenic M2 phenotype spectrum which affects distinct functions, like angiogenesis and cytokine production, and modulates BC aggressiveness and affects patient survival. Mast cells (MCs) are myeloid derived cells that serve as the first line of innate immune defense but their role in the TME of BC is not well understood. In this study, we have identified a subpopulation of innate immune cells that shows strong immunopositivity for the least studied adipokine CTRP8. Using a new and highly specific polyclonal antiserum on patient BC tissues, we identify a subset of tryptase + MCs and CD68 + macrophages co-expressing immunoreactive CTRP8. In M1 polarized THP-1 myeloid cells, this adipokine stimulated increased secretion of pro-inflammatory cytokines and elevated expression of the relaxin/ CTRP8 receptor RXFP1. Comparative analysis of secreted cytokine profiles in THP-1 M1 macrophages exposed to either CTRP8, relaxin-2 (RLN2), or the small molecule RXFP1 agonist ML-290 revealed ligand-specific cytokine signatures. Our study identified novel subsets of CTRP8 + myeloid derived innate immune cells and links this adipokine to pro-inflammatory events in the TME of BC.
Collapse
Affiliation(s)
- Leanne Arreza
- Departments of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Thatchawan Thanasupawat
- Departments of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sai Nivedita Krishnan
- Departments of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Matthew Kraljevic
- Children's Hospital Research Institute of Manitoba (CHRIM), Research Institute CancerCare Manitoba, Winnipeg, Canada
| | - Thomas Klonisch
- Departments of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Departments of Pathology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Departments of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Children's Hospital Research Institute of Manitoba (CHRIM), Research Institute CancerCare Manitoba, Winnipeg, Canada; Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, Canada
| | - Sabine Hombach-Klonisch
- Departments of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Departments of Pathology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Children's Hospital Research Institute of Manitoba (CHRIM), Research Institute CancerCare Manitoba, Winnipeg, Canada.
| |
Collapse
|
30
|
Zeylabi F, Jalali MT, Kaydani GA, Jaseb K, Saki N. rs1800890 Polymorphism of IL-10 and Susceptibility to Idiopathic Thrombocytopenic Purpura. J Pediatr Genet 2024; 13:263-271. [PMID: 39502848 PMCID: PMC11534459 DOI: 10.1055/s-0043-1775558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/26/2023] [Indexed: 11/08/2024]
Abstract
Immune thrombocytopenic purpura (ITP) is an immune bleeding disorder that is reported in approximately 2 out of every 100,000 adults with a mean age of 50 years. Several factors such as various genetic backgrounds are associated with the pathogenesis of ITP. Interleukin (IL)-10 is a complicated cytokine that has a role in tumor progression, antitumor immunity, and immune system regulation. rs1800890 is an IL-10 single nucleotide polymorphism linked to lower levels of IL-10. A total of 67 patients with ITP and 70 healthy individuals (controls) were considered in this study. The IL-10 polymorphism was detected by the amplification refractory mutation system-polymerase chain reaction technique. According to our analysis, individual carriers of the AA genotype were less likely to develop ITP. The AT genotype was more common in patients with ITP in comparison to the control group. However, there was no significant association between rs1800890 genotypes ( p = 0.775, odds ratio =1.517, 95%) in the acute and chronic groups. We observed that women had a higher mean frequency of this polymorphism ( p = 0.0012). The rs1800890 AA genotype was associated with the highest platelet counts. However, the mean platelet volume and platelet distribution width values among alleles of the polymorphisms did not vary significantly. The IL-10 rs1800890 polymorphism may have a role in idiopathic thrombocytopenic purpura etiology. As a result, more research with a larger number of sample sizes is suggested.
Collapse
Affiliation(s)
- Fatemeh Zeylabi
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Taha Jalali
- Department of Laboratory Sciences, Hyperlipidemia Research Center, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Gholam-Abbas Kaydani
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kaveh Jaseb
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
31
|
Somé GF, Séré M, Somda BM, Dayo GK, Ouédraogo GA, Boulangé A, Maarifi G, Chantal I, Berthier-Teyssedre D, Thévenon S. Immune Response in Cattle Trypanosomosis and Trypanotolerance: Main Findings and Gaps. Parasite Immunol 2024; 46:e13075. [PMID: 39508487 DOI: 10.1111/pim.13075] [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: 05/29/2024] [Revised: 09/30/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024]
Abstract
Trypanosome parasites of the genus Trypanosoma cause African animal trypanosomosis, a devastating livestock disease plaguing sub-Saharan Africa. Unlike many protozoan parasites, these extracellular blood-borne pathogens directly engage the host's immune system. While the mouse model has provided valuable insights, a comprehensive understanding of the bovine immune response to trypanosomes remains elusive. Addressing the immune response in cattle, the most relevant host species, and how it takes part in mitigating the negative impact of the disease could contribute to setting up sustainable control strategies. This review summarises the current knowledge of the immune response in cattle during trypanosomosis. Following a brief overview of infection processes and bovine trypanotolerance, we present advances in the regulation of host innate, inflammatory and adaptive responses and delve into the key immunological players involved in immunoactivities and immunosuppression. We discuss how these mechanisms contribute to tolerance or susceptibility to infection, highlighting critical gaps in knowledge that require further investigation.
Collapse
Affiliation(s)
- Gnohion Fabrice Somé
- Centre International de Recherche-Développement Sur l'Elevage en Zone Subhumide (CIRDES), Bobo-Dioulasso, Burkina Faso
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| | - Modou Séré
- Centre International de Recherche-Développement Sur l'Elevage en Zone Subhumide (CIRDES), Bobo-Dioulasso, Burkina Faso
- Université Daniel-Ouezzin-COULIBALLY, Dédougou, Burkina Faso
| | - Bienvenu Martin Somda
- Centre International de Recherche-Développement Sur l'Elevage en Zone Subhumide (CIRDES), Bobo-Dioulasso, Burkina Faso
- Université Nazi BONI, Bobo-Dioulasso, Burkina Faso
| | - Guiguigbaza-Kossigan Dayo
- Centre International de Recherche-Développement Sur l'Elevage en Zone Subhumide (CIRDES), Bobo-Dioulasso, Burkina Faso
| | | | - Alain Boulangé
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| | - Ghizlane Maarifi
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| | - Isabelle Chantal
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| | - David Berthier-Teyssedre
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| | - Sophie Thévenon
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| |
Collapse
|
32
|
Niu K, Zhang C, Liu C, Wu W, Yan Y, Zheng A, Liu S, Shi Z, Yang M, Wang W, Xiao Q. An unexpected role of IL10 in mesoderm induction and differentiation from pluripotent stem cells: Implications in zebrafish angiogenic sprouting, vascular organoid development, and therapeutic angiogenesis. Eur J Cell Biol 2024; 103:151465. [PMID: 39471724 DOI: 10.1016/j.ejcb.2024.151465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/21/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024] Open
Abstract
Mesoderm induction is a crucial step for vascular cell specification, vascular development and vasculogenesis. However, the cellular and molecular mechanisms underlying mesoderm induction remain elusive. In the present study, a chemically-defined differentiation protocol was used to induce mesoderm formation and generate functional vascular cells including smooth muscle cells (SMCs) and endothelial cells (ECs) from human induced pluripotent stem cells (hiPSCs). Zebrafish larvae were used to detect an in vivo function of interleukin 10 (IL10) in mesoderm formation and vascular development. A three dimensional approach was used to create hiPSC-derived blood vessel organoid (BVO) and explore a potential impact of IL10 on BVO formation. A murine model hind limb ischemia was applied to investigate a therapeutic potential of hiPSC-derived cells treated with or without IL10 during differentiation. We found that IL10 was significantly and specifically up-regulated during mesoderm stage of vascular differentiation. IL10 addition in mesoderm induction media dramatically increased mesoderm induction and vascular cell generation from hiPSCs, whereas an opposite effect was observed with IL10 inhibition. Mechanistic studies revealed that IL10 promotes mesoderm formation and vascular cell differentiation by activating signal transducer and activator of transcription 3 signal pathway. Functional studies with an in vivo model system confirmed that knockdown of IL10 using morpholino antisense oligonucleotides in zebrafish larvae caused defective mesoderm formation, angiogenic sprouting and vascular development. Additionally, our data also show IL10 promotes blood vessel organoid development and enhances vasculogenesis and angiogenesis. Importantly, we demonstrate that IL10 treatment during mesoderm induction stage enhances blood flow perfusion recovery and increases vasculogenesis and therapeutic angiogenesis after hind limb ischemia. Our data, therefore, demonstrate a regulatory role for IL10 in mesoderm formation from hiPSCs and during zebrafish vascular development, providing novel insights into mesoderm induction and vascular cell specifications.
Collapse
Affiliation(s)
- Kaiyuan Niu
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London EC1M 6BQ, UK; Department of Otolaryngology, Head & Neck Surgery, First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, Anhui 230022, PR China
| | - Chengxin Zhang
- Cardiovascular Surgery, First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, Anhui 230022, PR China
| | - Chenxin Liu
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Wei Wu
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Yi Yan
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, PR China
| | - Ancheng Zheng
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Silin Liu
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Zhenning Shi
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Mei Yang
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Wen Wang
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Qingzhong Xiao
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| |
Collapse
|
33
|
Lara-Guzmán OJ, Arango-González Á, Rivera DA, Muñoz-Durango K, Sierra JA. The colonic polyphenol catabolite dihydroferulic acid (DHFA) regulates macrophages activated by oxidized LDL, 7-ketocholesterol, and LPS switching from pro- to anti-inflammatory mediators. Food Funct 2024; 15:10399-10413. [PMID: 39320081 DOI: 10.1039/d4fo02114b] [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: 09/26/2024]
Abstract
Macrophage activation plays a central role in the development of atherosclerotic plaques. Interaction with oxidized low-density lipoprotein (oxLDL) leads to macrophage differentiation into foam cells and oxylipin production, contributing to plaque formation. 7-Ketocholesterol (7KC) is an oxidative byproduct of cholesterol found in oxLDL particles and is considered a factor contributing to plaque progression. During atherosclerotic lesion regression or stabilization, macrophages undergo a transformation from a pro-inflammatory phenotype to a reparative anti-inflammatory state. Interleukin-10 (IL-10) and PGE1 appear to be crucial in resolving both acute and chronic inflammatory processes. After coffee consumption, the gut microbiota processes non-absorbed chlorogenic acids producing various lower size phenolic acids. These colonic catabolites, including dihydroferulic acid (DHFA), may exert various local and systemic effects. We focused on DHFA's impact on inflammation and oxidative stress in THP-1 macrophages exposed to oxLDL, 7KC, and lipopolysaccharides (LPS). Our findings reveal that DHFA inhibits the release of several pro-inflammatory mediators induced by LPS in macrophages, such as CCL-2, CCL-3, CCL-5, TNF-α, IL-6, and IL-17. Furthermore, DHFA reduces IL-18 and IL-1β secretion in an inflammasome-like model. DHFA demonstrated additional benefits: it decreased oxLDL uptake and CD36 expression induced by oxLDL, regulated reactive oxygen species (ROS) and 8-isoprostane secretion (indicating oxidative stress modulation), and selectively increased IL-10 and PGE1 levels in the presence of inflammatory stimuli (LPS and 7KC). Finally, our study highlights the pivotal role of PGE1 in foam cell inhibition and inflammation regulation within activated macrophages. This study highlights DHFA's potential as an antioxidant and anti-inflammatory agent, particularly due to its ability to induce PGE1 and IL-10.
Collapse
Affiliation(s)
- Oscar J Lara-Guzmán
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Ángela Arango-González
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Diego A Rivera
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Katalina Muñoz-Durango
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Jelver A Sierra
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| |
Collapse
|
34
|
Dani C, Tarchi L, Cassioli E, Rossi E, Merola GP, Ficola A, Cordasco VZ, Ricca V, Castellini G. A transdiagnostic and diagnostic-specific approach on inflammatory biomarkers in eating disorders: A meta-analysis and systematic review. Psychiatry Res 2024; 340:116115. [PMID: 39128168 DOI: 10.1016/j.psychres.2024.116115] [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: 04/16/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 08/13/2024]
Abstract
Eating disorders (EDs) are severe mental illnesses with a multifactorial etiology and a chronic course. Among the biological factors related to pathogenesis and maintenance of EDs, inflammation acquired growing scientific interest. This study aimed to assess the inflammatory profile of EDs, focusing on anorexia nervosa, bulimia nervosa, and including for the first time binge eating disorder. A comprehensive research of existing literature identified 51 eligible studies for meta-analysis, comparing levels of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), C-reactive protein (CRP), osteoprotegerin (OPG), soluble receptor activator of nuclear factor kappa-B ligand (sRANKL), interleukin-1β (IL-1β), and interleukin-10 (IL-10) between patients with EDs and healthy controls (HCs). The systematic review explored other inflammatory biomarkers of interest, which did not meet the meta-analysis criteria. Results revealed significantly elevated levels of TNF-α, OPG, sRANKL, and IL-1β in patients with EDs compared to HCs. Additionally, the results highlighted the heterogeneity of inflammatory state among patients with EDs, emphasizing the need for further research into the association between inflammatory biomarkers and psychopathological correlates. This approach should transcend categorical diagnoses, enabling more precise subcategorizations of patients. Overall, this study contributed to the understanding of the inflammatory pathways involved in EDs, emphasizing potential implications for diagnosis, staging, and targeted interventions.
Collapse
Affiliation(s)
- Cristiano Dani
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Livio Tarchi
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Emanuele Cassioli
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Eleonora Rossi
- Department of Health Sciences, University of Florence, Florence, Italy
| | | | - Arianna Ficola
- Department of Health Sciences, University of Florence, Florence, Italy
| | | | - Valdo Ricca
- Department of Health Sciences, University of Florence, Florence, Italy
| | | |
Collapse
|
35
|
Grunwell JR, Huang M, Stephenson ST, Tidwell M, Ripple MJ, Fitzpatrick AM, Kamaleswaran R. RNA Sequencing Analysis of Monocytes Exposed to Airway Fluid From Children With Pediatric Acute Respiratory Distress Syndrome. Crit Care Explor 2024; 6:e1125. [PMID: 39365167 PMCID: PMC11458172 DOI: 10.1097/cce.0000000000001125] [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] [Indexed: 10/05/2024] Open
Abstract
OBJECTIVES Monocytes are plastic cells that assume different polarization states that can either promote inflammation or tissue repair and inflammation resolution. Polarized monocytes are partially defined by their transcriptional profiles that are influenced by environmental stimuli. The airway monocyte response in pediatric acute respiratory distress syndrome (PARDS) is undefined. To identify differentially expressed genes and networks using a novel transcriptomic reporter assay with donor monocytes exposed to the airway fluid of intubated children with and at-risk for PARDS. To determine differences in gene expression at two time points using the donor monocyte assay exposed to airway fluid from intubated children with PARDS obtained 48-96 hours following initial tracheal aspirate sampling. DESIGN In vitro pilot study carried out using airway fluid supernatant. SETTING Academic 40-bed PICU. PARTICIPANTS Fifty-seven children: 44 children with PARDS and 13 children at-risk for PARDS. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We performed bulk RNA sequencing using a transcriptomic reporter assay of monocytes exposed to airway fluid from intubated children to discover gene networks differentiating PARDS from at-risk for PARDS and those differentiating mild/moderate from severe PARDS. We also report differences in gene expression in children with PARDS 48-96 hours following initial tracheal aspirate sampling. We found that interleukin (IL)-10, IL-4, and IL-13, cytokine/chemokine signaling, and the senescence-associated secretory phenotype are upregulated in monocytes exposed to airway fluid from intubated children with PARDS compared with those at-risk for PARDS. Signaling by NOTCH, histone deacetylation/acetylation, DNA methylation, chromatin modifications (B-WICH complex), and RNA polymerase I transcription and its associated regulatory apparatus were upregulated in children with PARDS 48-96 hours following initial tracheal aspirate sampling. CONCLUSIONS We identified gene networks important to the PARDS airway immune response using bulk RNA sequencing from a monocyte reporter assay that exposed monocytes to airway fluid from intubated children with and at-risk for PARDS. Mechanistic investigations are needed to validate our findings.
Collapse
Affiliation(s)
- Jocelyn R. Grunwell
- Department of Pediatrics/Division of Critical Care Medicine, Egleston Hospital, Children’s Healthcare of Atlanta, Atlanta, GA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Min Huang
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA
| | | | - Mallory Tidwell
- Department of Pediatrics/Division of Critical Care Medicine, Egleston Hospital, Children’s Healthcare of Atlanta, Atlanta, GA
| | - Michael J. Ripple
- Department of Pediatrics/Division of Critical Care Medicine, Egleston Hospital, Children’s Healthcare of Atlanta, Atlanta, GA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Anne M. Fitzpatrick
- Department of Pediatrics/Division of Critical Care Medicine, Egleston Hospital, Children’s Healthcare of Atlanta, Atlanta, GA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Rishikesan Kamaleswaran
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
36
|
Fogang B, Schoenhals M, Maloba FM, Biabi MF, Essangui E, Donkeu C, Cheteug G, Kapen M, Keumoe R, Kemleu S, Nsango S, Cornwall DH, Eboumbou C, Perraut R, Megnekou R, Lamb TJ, Ayong LS. Asymptomatic carriage of Plasmodium falciparum in children living in a hyperendemic area occurs independently of IgG responses but is associated with a balanced inflammatory cytokine ratio. Malar J 2024; 23:268. [PMID: 39232787 PMCID: PMC11375831 DOI: 10.1186/s12936-024-05086-8] [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: 06/06/2024] [Accepted: 08/17/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Asymptomatic carriage of infected red blood cells (iRBCs) can be prevalent in communities regardless of transmission patterns and can occur with infection of different Plasmodium species. Clinical immunity dampens the inflammatory responses leading to disease symptoms in malaria. The aim of this study was to define the immunological correlates of asymptomatic carriage of Plasmodium falciparum in a highly exposed population. METHODS 142 asymptomatic Plasmodium-infected individuals greater than 2 years of age without fever (body temperature <37.5 ℃) were followed weekly for 10 weeks before being treated with artemisinin-based combination therapy (ACT). Plasma levels of 38 cytokines were measured at baseline by Luminex and the quantity and growth inhibitory activities of circulating parasite-reactive antibodies measured. The Plasmodium antigen tested included P. falciparum merozoite extract (ME) and schizont extract (SE), and the recombinant proteins erythrocyte binding antigen 175 (EBA-175) and merozoite surface protein 1 (MSP-119). RESULTS Median levels of IgG against P. falciparum EBA-175 and MSP-119 at baseline were significantly higher in those older than 20 years of age compared with the younger age group and appeared to correlate with better parasite control. Amongst all participants there were no discernible changes in IgG levels over time. Parasite density was higher in the younger age group and associated with IL-10, TNF and MCP-1 levels. A balanced IL-10:TNF ratio was associated with asymptomatic malaria regardless of age, and balanced ratios of IL-10/TNF and IL-10/IFN-γ were the only significant correlate of maintenance of asymptomatic malaria over the course of the study in individuals 20 years of age and younger. CONCLUSION The above findings indicate that asymptomatic carriage of P. falciparum in children living in a hyperendemic area occurs independently of IgG but is associated with a balanced inflammatory cytokine ratio.
Collapse
Affiliation(s)
- Balotin Fogang
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
- Department of Animal Biology and Physiology of the University of Yaoundé I, BP 812, Yaounde, Cameroon
| | - Matthieu Schoenhals
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
| | - Franklin M Maloba
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
| | - Marie Florence Biabi
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
- Department of Biochemistry, University of Douala, BP 24157, Douala, Cameroon
| | - Estelle Essangui
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
| | - Christiane Donkeu
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
- Department of Animal Biology and Physiology of the University of Yaoundé I, BP 812, Yaounde, Cameroon
| | - Glwadys Cheteug
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
- Department of Medical Laboratory Sciences, University of Buea, BP 63, Buea, Cameroon
| | - Marie Kapen
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
| | - Rodrigue Keumoe
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
| | - Sylvie Kemleu
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
| | - Sandrine Nsango
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
- Faculty of Medicine and Pharmaceutical Sciences, University of Douala, BP 2701, Douala, Cameroon
| | - Douglas H Cornwall
- Department of Pathology, University of Utah, 15 N Medical Drive, Salt Lake City, 84112, USA
| | - Carole Eboumbou
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
- Faculty of Medicine and Pharmaceutical Sciences, University of Douala, BP 2701, Douala, Cameroon
| | - Ronald Perraut
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon
| | - Rosette Megnekou
- Department of Animal Biology and Physiology of the University of Yaoundé I, BP 812, Yaounde, Cameroon
| | - Tracey J Lamb
- Department of Pathology, University of Utah, 15 N Medical Drive, Salt Lake City, 84112, USA.
| | - Lawrence S Ayong
- Molecular Parasitology Laboratory, Centre Pasteur du Cameroun, BP 1274, Yaounde, Cameroon.
| |
Collapse
|
37
|
Qian F, He R, Du X, Wei Y, Zhou Z, Fan J, He Y. Microglia and Astrocytes Responses Contribute to Alleviating Inflammatory Damage by Repetitive Transcranial Magnetic Stimulation in Rats with Traumatic Brain Injury. Neurochem Res 2024; 49:2636-2651. [PMID: 38909329 DOI: 10.1007/s11064-024-04197-7] [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: 03/12/2024] [Revised: 05/30/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a therapeutic strategy that shows promise in ameliorating the clinical sequelae following traumatic brain injury (TBI). These improvements are associated with neuroplastic changes in neurons and their synaptic connections. However, it has been hypothesized that rTMS may also modulate microglia and astrocytes, potentially potentiating their neuroprotective capabilities. This study aims to investigate the effects of high-frequency rTMS on microglia and astrocytes that may contribute to its neuroprotective effects. Feeney's weight-dropping method was used to establish rat models of moderate TBI. To evaluate the neuroprotective effect of high frequency rTMS on rats by observing the synaptic ultrastructure and the level of neuron apoptosis. The levels of several important inflammation-related proteins within microglia and astrocytes were assessed through immunofluorescence staining and western blot. Our findings demonstrate that injured neurons can be rescued through the modulation of microglia and astrocytes by rTMS. This modulation plays a key role in preserving the synaptic ultrastructure and inhibiting neuronal apoptosis. Among microglia, we observed that rTMS inhibited the levels of proinflammatory factors (CD16, IL-6 and TNF-α) and promoted the levels of anti-inflammatory factors (CD206, IL-10 and TNF-β). rTMS also reduced the levels of pyroptosis within microglia and pyroptosis-related proteins (NLRP3, Caspase-1, GSDMD, IL-1β and IL-18). Moreover, rTMS downregulated P75NTR expression and up-regulated IL33 expression in astrocytes. These findings suggest that regulation of microglia and astrocytes is the mechanism through which rTMS attenuates neuronal inflammatory damage after moderate TBI.
Collapse
Affiliation(s)
- FangFang Qian
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - RenHong He
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - XiaoHui Du
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Yi Wei
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Zhou Zhou
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - JianZhong Fan
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
| | - YouHua He
- Department of Comprehensive Medical Treatment Ward, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
| |
Collapse
|
38
|
Khalili L, Park G, Nagpal R, Salazar G. The Role of Akkermansia muciniphila on Improving Gut and Metabolic Health Modulation: A Meta-Analysis of Preclinical Mouse Model Studies. Microorganisms 2024; 12:1627. [PMID: 39203469 PMCID: PMC11356609 DOI: 10.3390/microorganisms12081627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Akkermansia muciniphila (A. muciniphila) and its derivatives, including extracellular vesicles (EVs) and outer membrane proteins, are recognized for enhancing intestinal balance and metabolic health. However, the mechanisms of Akkermansia muciniphila's action and its effects on the microbiome are not well understood. In this study, we examined the influence of A. muciniphila and its derivatives on gastrointestinal (GI) and metabolic disorders through a meta-analysis of studies conducted on mouse models. A total of 39 eligible studies were identified through targeted searches on PubMed, Web of Science, Science Direct, and Embase until May 2024. A. muciniphila (alive or heat-killed) and its derivatives positively affected systemic and gut inflammation, liver enzyme level, glycemic response, and lipid profiles. The intervention increased the expression of tight-junction proteins in the gut, improving gut permeability in mouse models of GI and metabolic disorders. Regarding body weight, A. muciniphila and its derivatives prevented weight loss in animals with GI disorders while reducing body weight in mice with metabolic disorders. Sub-group analysis indicated that live bacteria had a more substantial effect on most analyzed biomarkers. Gut microbiome analysis using live A. muciniphila identified a co-occurrence cluster, including Desulfovibrio, Family XIII AD3011 group, and Candidatus Saccharimonas. Thus, enhancing the intestinal abundance of A. muciniphila and its gut microbial clusters may provide more robust health benefits for cardiometabolic, and age-related diseases compared with A. muciniphila alone. The mechanistic insight elucidated here will pave the way for further exploration and potential translational applications in human health.
Collapse
Affiliation(s)
- Leila Khalili
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Gwoncheol Park
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Ravinder Nagpal
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Gloria Salazar
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
- Center for Advancing Exercise and Nutrition Research on Aging (CAENRA), Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
39
|
Aladag T, Acar G, Mogulkoc R, Baltaci AK. Improvement of neuronal and cognitive functions following treatment with 3',4' dihydroxyflavonol in experimental focal cerebral ischemia-reperfusion injury in rats. Eur J Pharmacol 2024; 976:176670. [PMID: 38795755 DOI: 10.1016/j.ejphar.2024.176670] [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: 03/06/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/28/2024]
Abstract
INTRODUCTION Ischemia/reperfusion is a pathological condition by the restoration of perfusion and oxygenation following a period of restricted blood flow to an organ. To address existing uncertainty in the literature regarding the effects of 3', 4'-dihydroxy flavonol (DiOHF) on cerebral ischemia/reperfusion injury, our study aims to investigate the impact of DiOHF on neurological parameters, apoptosis (Caspase-3), aquaporin 4 (AQP4), and interleukin-10 (IL-10) levels in an experimental rat model of brain ischemia-reperfusion injury. MATERIALS/METHODS A total of 28 Wistar-albino male rats were used in this study. Experimental groups were formed as 1-Control, 2-Sham, 3-Ischemia-reperfusion, 4-Ischemia-reperfusion + DiOHF (10 mg/kg). The animals were anaesthetized, and the carotid arteries were ligated (ischemia) for 30 min, followed by reperfusion for 30 min. Following reperfusion, DiOHF was administered intraperitoneally to the animals at a dose of 10 mg/kg for 1 week. During the one-week period neurological scores and new object recognition tests were performed. Then, caspase 3 and AQP4 levels were determined by PCR method and IL-10 by ELISA method in hippocampus tissue samples taken from animals sacrificed under anaesthesia. RESULTS Brain ischemia reperfusion significantly increased both caspase 3 and AQP4 values in the hippocampus tissue, while decreasing IL-10 levels. However, 1-week DiOHF supplementation significantly suppressed increased caspase 3 and AQP4 levels and increased IL-10 values. While I/R also increased neurological score values, it suppressed the ability to recognize new objects, and the administered treatment effectively ameliorated the adverse effects observed, resulting in a positive outcome. CONCLUSIONS The results of the study show that brain ischemia caused by bilateral carotid occlusion in rats and subsequent reperfusion causes tissue damage, but 1-week DiOHF application has a healing effect on both hippocampus tissue and neurological parameters.
Collapse
Affiliation(s)
- Tugce Aladag
- Selcuk University, Medical Faculty, Department of Physiology, Konya, Turkey
| | - Gozde Acar
- Selcuk University, Medical Faculty, Department of Physiology, Konya, Turkey
| | - Rasim Mogulkoc
- Selcuk University, Medical Faculty, Department of Physiology, Konya, Turkey.
| | | |
Collapse
|
40
|
Li R, Ye JJ, Gan L, Zhang M, Sun D, Li Y, Wang T, Chang P. Traumatic inflammatory response: pathophysiological role and clinical value of cytokines. Eur J Trauma Emerg Surg 2024; 50:1313-1330. [PMID: 38151578 PMCID: PMC11458723 DOI: 10.1007/s00068-023-02388-5] [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: 06/05/2023] [Accepted: 10/23/2023] [Indexed: 12/29/2023]
Abstract
Severe trauma is an intractable problem in healthcare. Patients have a widespread immune system response that is complex and vital to survival. Excessive inflammatory response is the main cause of poor prognosis and poor therapeutic effect of medications in trauma patients. Cytokines are signaling proteins that play critical roles in the body's response to injuries, which could amplify or suppress immune responses. Studies have demonstrated that cytokines are closely related to the severity of injuries and prognosis of trauma patients and help present cytokine-based diagnosis and treatment plans for trauma patients. In this review, we introduce the pathophysiological mechanisms of a traumatic inflammatory response and the role of cytokines in trauma patients. Furthermore, we discuss the potential of cytokine-based diagnosis and therapy for post-traumatic inflammatory response, although further clarification to elucidate the underlying mechanisms of cytokines following trauma is warranted.
Collapse
Affiliation(s)
- Rui Li
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, People's Republic of China
- Key Laboratory of Trauma and Neural Regeneration (Peking University) Ministry of Education, Beijing, 100044, People's Republic of China
- National Center for Trauma Medicine of China, Beijing, 100044, People's Republic of China
| | - Jing Jing Ye
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, People's Republic of China
- Key Laboratory of Trauma and Neural Regeneration (Peking University) Ministry of Education, Beijing, 100044, People's Republic of China
- National Center for Trauma Medicine of China, Beijing, 100044, People's Republic of China
| | - Lebin Gan
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, People's Republic of China
- Key Laboratory of Trauma and Neural Regeneration (Peking University) Ministry of Education, Beijing, 100044, People's Republic of China
- National Center for Trauma Medicine of China, Beijing, 100044, People's Republic of China
| | - Mengwei Zhang
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, People's Republic of China
- Key Laboratory of Trauma and Neural Regeneration (Peking University) Ministry of Education, Beijing, 100044, People's Republic of China
- National Center for Trauma Medicine of China, Beijing, 100044, People's Republic of China
| | - Diya Sun
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, People's Republic of China
- Key Laboratory of Trauma and Neural Regeneration (Peking University) Ministry of Education, Beijing, 100044, People's Republic of China
- National Center for Trauma Medicine of China, Beijing, 100044, People's Republic of China
| | - Yongzheng Li
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, People's Republic of China.
| | - Tianbing Wang
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, People's Republic of China.
- Key Laboratory of Trauma and Neural Regeneration (Peking University) Ministry of Education, Beijing, 100044, People's Republic of China.
- National Center for Trauma Medicine of China, Beijing, 100044, People's Republic of China.
| | - Panpan Chang
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, People's Republic of China.
- Key Laboratory of Trauma and Neural Regeneration (Peking University) Ministry of Education, Beijing, 100044, People's Republic of China.
- National Center for Trauma Medicine of China, Beijing, 100044, People's Republic of China.
| |
Collapse
|
41
|
Griffin H, Ceron-Gutierrez L, Gharahdaghi N, Ebrahimi S, Davies S, Loo PS, Szabo A, Williams E, Mukhopadhyay A, McLoughlin L, Irwin S, Travis S, Klenerman P, Bunn S, Cant AJ, Hambleton S, Uhlig HH, Doffinger R. Neutralizing Autoantibodies against Interleukin-10 in Inflammatory Bowel Disease. N Engl J Med 2024; 391:434-441. [PMID: 39083772 PMCID: PMC7616361 DOI: 10.1056/nejmoa2312302] [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] [Indexed: 08/02/2024]
Abstract
We discovered high-titer neutralizing autoantibodies against interleukin-10 in a child with infantile-onset inflammatory bowel disease (IBD), a phenocopy of inborn errors of interleukin-10 signaling. After B-cell-depletion therapy and an associated decrease in the anti-interleukin-10 titer, conventional IBD therapy could be withdrawn. A second child with neutralizing anti-interleukin-10 autoantibodies had a milder course of IBD and has been treated without B-cell depletion. We conclude that neutralizing anti-interleukin-10 autoantibodies may be a causative or modifying factor in IBD, with potential implications for therapy. (Funded by the National Institute for Health and Care Research and others.).
Collapse
Affiliation(s)
- Helen Griffin
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Lourdes Ceron-Gutierrez
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Nima Gharahdaghi
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Soraya Ebrahimi
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Sophie Davies
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Peh Sun Loo
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Andras Szabo
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Eleri Williams
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Anirban Mukhopadhyay
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Louise McLoughlin
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Steven Irwin
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Simon Travis
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Paul Klenerman
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Su Bunn
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Andrew J Cant
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Sophie Hambleton
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Holm H Uhlig
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| | - Rainer Doffinger
- From the Immunity and Inflammation Theme, Newcastle University Translational and Clinical Research Institute (H.G., S.H.), and the Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust (P.S.L., E.W., A.M., A.J.C., S.H.), Newcastle upon Tyne, the Department of Clinical Biochemistry and Immunology, Cambridge University Hospital (L.C.-G., S.E., S.D., R.D.), and the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (R.D.), Cambridge, the Translational Gastroenterology Unit (N.G., S.T., P.K., H.H.U.), the Kennedy Institute of Rheumatology (S.T.), the NIHR Oxford Biomedical Research Centre (S.T., P.K., H.H.U.), and the Department of Pediatrics (H.H.U.), University of Oxford, Oxford, the Department of Pediatric Gastroenterology, Royal Belfast Hospital for Sick Children (A.S., L.M.), and the Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust (S.I.), Belfast, and the Department of Pediatric Gastroenterology, Royal Aberdeen Children's Hospital, Aberdeen (S.B.) - all in the United Kingdom; and the Pediatric Gastroenterology Department, Pál Heim National Pediatric Institute, Budapest, Hungary (A.S.)
| |
Collapse
|
42
|
Liu Y, Wenren M, Cheng W, Zhou X, Xu D, Chi C, Lü Z, Liu H. Identification, functional characterization and immune response profiles of interleukin-10 in Nibea albiflora. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109654. [PMID: 38810711 DOI: 10.1016/j.fsi.2024.109654] [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: 04/10/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024]
Abstract
Interleukin-10 (IL-10) is an immunosuppressive cytokine, which plays a vital role in regulating inflammation for inhibiting the generation and function of pro-inflammatory cytokines in vivo or in vitro. In the present study, the full length cDNA of IL-10 was characterized from Nibea albiflora (named as NaIL-10) of 1238 base pairs (bp), containing a 5'-UTR (untranslated region) of 350 bp, a 3'-UTR of 333 bp and an open reading frame (ORF) of 555 bp (Fig. 1A) to encode 184 amino acid residues with a signal peptide at the N-terminus. The sequence analysis showed that NaIL-10 possessed the typical IL-10 family symbolic motif and conversed cysteine residues, similar to its teleost orthologues. Real-time PCR indicated that NaIL-10 had wide distribution in different healthy tissues, with a relatively high expression in immune-related tissues (head kidney, spleen, kidney, liver and gill). Significantly, up-regulations of NaIL-10 after infection against Vibrio parahaemolyticus, Vibrio alginolyticus and Poly I:C were also observed. Subcellular localization manifested that NaIL-10 mainly distributed in the cytoplasm unevenly and aggregately, and there was also a small amount on the cell membrane, indicating that NaIL-10 was secreted to the extracellular space as the known IL-10 homologous molecules. It could co-locate with IL-10 Rα on the membrane of HEK293T cells for their potential interaction, and GST pull-down and Co-IP studies certified the specific and direct interaction between NaIL-10 and NaIL-10 Rα, confirming that an IL-10 ligand-receptor system existed in N.albiflora. The expression of pro-inflammatory cytokines, including TNF-α, IL-6, IL-1β, were dramatically inhibited in LPS-stimulated RAW264.7 macrophages pre-incubated with recombinant NaIL-10 protein, demonstrating its anti-inflammatory roles. Taken together, the results demonstrated the existence of IL-10 ligand-receptor system in N.albiflora for the first time, and indicated the suppressive function of NaIL-10 on pro-inflammatory cytokine expression in inflammatory response, which would be conducive to better comprehending the role of IL-10 in the immunomodulatory mechanisms of teleost.
Collapse
Affiliation(s)
- Yue Liu
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Mingming Wenren
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Wei Cheng
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Xu Zhou
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Dongdong Xu
- Zhejiang Marine Fisheries Research Institute, Key Lab of Mariculture and Enhancement of Zhejiang province, Zhoushan, 316100, China
| | - Changfeng Chi
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Zhenming Lü
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Huihui Liu
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China.
| |
Collapse
|
43
|
Cebesoy EI, Altaca M, Kocak-Oztug NA, Bingül I, Cifcibasi E. Associations between interleukin-10, -12, and - 18 and periodontal health and disease: a cross-sectional study. Clin Oral Investig 2024; 28:458. [PMID: 39080003 PMCID: PMC11289060 DOI: 10.1007/s00784-024-05843-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVE We assessed the levels of Interleukin-10 (IL-10), Interleukin-12 (IL-12), and Interleukin-18 (IL-18) in the gingival crevicular fluid (GCF) of subjects with advanced periodontitis (SIII-SIV) compared to healthy controls and evaluated their correlations with clinical measurements. METHODS This cross-sectional study involved subjects (n = 60) diagnosed with stage III grade B-C (n = 13) to stage IV grade C (n = 17) periodontitis, and periodontally healthy controls (n = 30). Clinical periodontal measurements involved full-mouth. The concentrations of IL-10, IL-12, and IL-18 were determined using enzyme-linked immunosorbent assay (ELISA). RESULTS There were no significant differences in IL-12 level and IL-18/IL-10 ratio between the healthy and periodontitis groups (p = 0.413, p = 0.636, respectively). The IL-10 and IL-18 levels were significantly higher in the periodontitis group than in controls (p < 0.001, p < 0.001, respectively). Significant associations were observed between the periodontitis and IL-10 and IL-18 levels (OR = 1.46, %95 CI 1.19-1.795; OR = 1.13, %95 CI 1.059-1.207, respectively) (p < 0.001, p < 0.001, respectively). CONCLUSIONS There was a correlation between pocket depth and the presence of IL-18 and a strong association between periodontitis and a high level of IL-18. However, there were no direct correlations among the three biomarkers and IL-18/IL-10 ratio, indicating that their roles in periodontal health are complex and multidimensional. CLINICAL RELEVANCE Understanding the cytokine dynamics in GCF provides valuable insights into their potential clinical implications for periodontal disease diagnosis, risk assessment, and tailored therapeutic interventions.
Collapse
Affiliation(s)
- Elif Ilke Cebesoy
- Faculty of Dentistry, Department of Periodontology, Istanbul University, Istanbul, 34116, Turkey
- Institute of Graduate Studies in Health Sciences, Department of Periodontology, Istanbul University, Istanbul, 34126, Turkey
| | - Müge Altaca
- Faculty of Dentistry, Department of Periodontology, Istanbul University, Istanbul, 34116, Turkey
- Institute of Graduate Studies in Health Sciences, Department of Periodontology, Istanbul University, Istanbul, 34126, Turkey
| | - Necla Asli Kocak-Oztug
- Faculty of Dentistry, Department of Periodontology, Istanbul University, Istanbul, 34116, Turkey
- School of Dentistry, Faculty of Health and Behavioural Sciences, The University of Queensland, Brisbane, QLD, 4006, Australia
| | - Ilknur Bingül
- Department of Medical Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Emine Cifcibasi
- Faculty of Dentistry, Department of Periodontology, Istanbul University, Istanbul, 34116, Turkey.
| |
Collapse
|
44
|
Möttönen M, Teräsjärvi J, Rahikkala H, Kvist S, Mertsola J, He Q. Association of IL-17A and IL-10 Polymorphisms with Juvenile Idiopathic Arthritis in Finnish Children. Int J Mol Sci 2024; 25:8323. [PMID: 39125893 PMCID: PMC11311899 DOI: 10.3390/ijms25158323] [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: 05/16/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
To analyze the role of interleukin IL-17A and IL-10 polymorphisms in susceptibility to juvenile idiopathic arthritis (JIA), 98 Finnish children and adolescents with JIA were studied. Data from the 1000 Genomes Project, consisting of 99 healthy Finns, served as the controls. The patients were analyzed for four IL-17A and three IL-10 gene-promoter polymorphisms, and the serum IL-17A, IL-17F, IL-10, and IL-6 levels were determined. The IL-17A rs8193036 variant genotypes (CT/CC) were more common among the patients than controls, especially in those with polyarthritis (OR 1.93, 95% CI 1.11-3.36; p = 0.020). IL-17A rs2275913 minor allele A was more common in patients (OR 1.45, 95% Cl 1.08-1.94; p = 0.014) and especially among patients with oligoarthritis and polyarthritis than the controls (OR 1.61, 95%CI 1.06-2.43; p = 0.024). Carriers of the IL-17A rs4711998 variant genotype (AG/AA) had higher serum IL-17A levels than those with genotype GG. However, carriers of the variant genotypes of IL-17A rs9395767 and rs4711998 appeared to have higher IL-17F levels than those carrying wildtype. IL-10 rs1800896 variant genotypes (TC/CC) were more abundant in patients than in the controls (OR 1.97, 95%CI 1.06-3.70; p = 0.042). Carriers of the IL-10 rs1800896 variant genotypes had lower serum levels of IL-17F than those with wildtype. These data provide preliminary evidence of the roles of IL-17 and IL-10 in the pathogenesis of JIA and its subtypes in the Finnish population. However, the results should be interpreted with caution, as the number of subjects included in this study was limited.
Collapse
Affiliation(s)
- Milja Möttönen
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, 20520 Turku, Finland; (M.M.); (H.R.); (J.M.)
| | - Johanna Teräsjärvi
- Institute of Biomedicine, Research Centre for Infections and Immunity, University of Turku, 20520 Turku, Finland; (J.T.); (S.K.)
| | - Heidi Rahikkala
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, 20520 Turku, Finland; (M.M.); (H.R.); (J.M.)
- Research Unit of Clinical Medicine, University of Oulu, 90014 Oulu, Finland
| | - Sonja Kvist
- Institute of Biomedicine, Research Centre for Infections and Immunity, University of Turku, 20520 Turku, Finland; (J.T.); (S.K.)
| | - Jussi Mertsola
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, 20520 Turku, Finland; (M.M.); (H.R.); (J.M.)
| | - Qiushui He
- Institute of Biomedicine, Research Centre for Infections and Immunity, University of Turku, 20520 Turku, Finland; (J.T.); (S.K.)
- InFLAMES Research Flagship Centre, University of Turku, 20520 Turku, Finland
| |
Collapse
|
45
|
Banerjee D, Feng J, Sellke FW. Strategies to attenuate maladaptive inflammatory response associated with cardiopulmonary bypass. Front Surg 2024; 11:1224068. [PMID: 39022594 PMCID: PMC11251955 DOI: 10.3389/fsurg.2024.1224068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/07/2024] [Indexed: 07/20/2024] Open
Abstract
Cardiopulmonary bypass (CPB) initiates an intense inflammatory response due to various factors: conversion from pulsatile to laminar flow, cold cardioplegia, surgical trauma, endotoxemia, ischemia-reperfusion injury, oxidative stress, hypothermia, and contact activation of cells by the extracorporeal circuit. Redundant and overlapping inflammatory cascades amplify the initial response to produce a systemic inflammatory response, heightened by coincident activation of coagulation and fibrinolytic pathways. When unchecked, this inflammatory response can become maladaptive and lead to serious postoperative complications. Concerted research efforts have been made to identify technical refinements and pharmacologic interventions that appropriately attenuate the inflammatory response and ultimately translate to improved clinical outcomes. Surface modification of the extracorporeal circuit to increase biocompatibility, miniaturized circuits with sheer resistance, filtration techniques, and minimally invasive approaches have improved clinical outcomes in specific populations. Pharmacologic adjuncts, including aprotinin, steroids, monoclonal antibodies, and free radical scavengers, show real promise. A multimodal approach incorporating technical, circuit-specific, and pharmacologic strategies will likely yield maximal clinical benefit.
Collapse
Affiliation(s)
| | | | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Brown University/Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
46
|
Gęgotek A, Moniuszko-Malinowska A, Groth M, Skrzydlewska E. Changes in cerebrospinal fluid proteome of patients with tick-borne encephalitis. J Med Virol 2024; 96:e29763. [PMID: 38949193 DOI: 10.1002/jmv.29763] [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: 03/13/2024] [Revised: 05/11/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
Tick-borne encephalitis (TBE) is one of the main diseases transmitted by ticks, the incidence of which is increasing. Moreover, its diagnosis and therapy are often long and difficult according to nonspecific symptoms and complex etiology. This study aimed to observe changes in the proteome of cerebrospinal fluid from TBE patients. Cerebrospinal fluid (CSF) of TBE patients (n = 20) and healthy individuals (n = 10) was analyzed using a proteomic approach (QExactiveHF-Orbitrap mass spectrometer) and zymography. Obtained results show that in CSF of TBE patients, the top-upregulated proteins are involved in pro-inflammatory reaction (interleukins), as well as antioxidant/protective response (peroxiredoxins, heat shock proteins). Moreover, changes in the proteome of CSF are not only the result of this disease development, but they can also be an indicator of its course. This mainly applies to proteins involved in proteolysis including serpins and metalloproteinases, whose activity is proportional to the length of patients' convalescence. The obtained proteomic data strongly direct attention to the changes caused by the development of TBE to antioxidant, pro-inflammatory, and proteolytic proteins, knowledge about which can significantly contribute to faster and more accurate diagnosis of various clinical forms of TBE.
Collapse
Affiliation(s)
- Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Monika Groth
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
47
|
Luo W, Zhang H, Wan R, Cai Y, Liu Y, Wu Y, Yang Y, Chen J, Zhang D, Luo Z, Shang X. Biomaterials-Based Technologies in Skeletal Muscle Tissue Engineering. Adv Healthc Mater 2024; 13:e2304196. [PMID: 38712598 DOI: 10.1002/adhm.202304196] [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: 11/28/2023] [Revised: 04/26/2024] [Indexed: 05/08/2024]
Abstract
For many clinically prevalent severe injuries, the inherent regenerative capacity of skeletal muscle remains inadequate. Skeletal muscle tissue engineering (SMTE) seeks to meet this clinical demand. With continuous progress in biomedicine and related technologies including micro/nanotechnology and 3D printing, numerous studies have uncovered various intrinsic mechanisms regulating skeletal muscle regeneration and developed tailored biomaterial systems based on these understandings. Here, the skeletal muscle structure and regeneration process are discussed and the diverse biomaterial systems derived from various technologies are explored in detail. Biomaterials serve not merely as local niches for cell growth, but also as scaffolds endowed with structural or physicochemical properties that provide tissue regenerative cues such as topographical, electrical, and mechanical signals. They can also act as delivery systems for stem cells and bioactive molecules that have been shown as key participants in endogenous repair cascades. To achieve bench-to-bedside translation, the typical effect enabled by biomaterial systems and the potential underlying molecular mechanisms are also summarized. Insights into the roles of biomaterials in SMTE from cellular and molecular perspectives are provided. Finally, perspectives on the advancement of SMTE are provided, for which gene therapy, exosomes, and hybrid biomaterials may hold promise to make important contributions.
Collapse
Affiliation(s)
- Wei Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Hanli Zhang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Renwen Wan
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yuxi Cai
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yinuo Liu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yang Wu
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yimeng Yang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Jiani Chen
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, Hong Kong
| | - Zhiwen Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Xiliang Shang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| |
Collapse
|
48
|
Obare LM, Simmons J, Oakes J, Zhang X, Nochowicz C, Priest S, Bailin SS, Warren CM, Mashayekhi M, Beasley HK, Shao J, Meenderink LM, Sheng Q, Stolze J, Gangula R, Absi T, Su YR, Neikirk K, Chopra A, Gabriel CL, Temu T, Pakala S, Wilfong EM, Gianella S, Phillips EJ, Harrison DG, Hinton A, Kalams SA, Kirabo A, Mallal SA, Koethe JR, Wanjalla CN. CD3 + T-cell: CD14 +monocyte complexes are dynamic and increased with HIV and glucose intolerance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.24.538020. [PMID: 37162990 PMCID: PMC10168203 DOI: 10.1101/2023.04.24.538020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
An increased risk of cardiometabolic disease accompanies persistent systemic inflammation. Yet, the innate and adaptive immune system features in persons who develop these conditions remain poorly defined. Doublets, or cell-cell complexes, are routinely eliminated from flow cytometric and other immune phenotyping analyses, which limits our understanding of their relationship to disease states. Using well-characterized clinical cohorts, including participants with controlled HIV as a model for chronic inflammation and increased immune cell interactions, we show that circulating CD14+ monocytes complexed to CD3+ T cells are dynamic, biologically relevant, and increased in individuals with diabetes after adjusting for confounding factors. The complexes form functional immune synapses with increased expression of proinflammatory cytokines and greater glucose utilization. Furthermore, in persons with HIV, the CD3+T-cell: CD14+monocyte complexes had more HIV copies compared to matched CD14+ monocytes or CD4+ T cells alone. Our results demonstrate that circulating CD3+T-cell:CD14+monocyte pairs represent dynamic cellular interactions that may contribute to inflammation and cardiometabolic disease pathogenesis and may originate or be maintained, in part, by chronic viral infections. These findings provide a foundation for future studies investigating mechanisms linking T cellmonocyte cell-cell complexes to developing immune-mediated diseases, including HIV and diabetes.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua Simmons
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jared Oakes
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xiuqi Zhang
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cindy Nochowicz
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephen Priest
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Samuel S. Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Mona Mashayekhi
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, USA
| | - Leslie M. Meenderink
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - Joey Stolze
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - Rama Gangula
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tarek Absi
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yan Ru Su
- Department of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - Curtis L. Gabriel
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tecla Temu
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Suman Pakala
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erin M. Wilfong
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara Gianella
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth J. Phillips
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David G. Harrison
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Spyros A. Kalams
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - John R. Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
49
|
Jo HS, Kim HJ. Stabilin-2 mediated apoptotic cell phagocytosis induces interleukin-10 expression by p38 and Pbx1 signaling. Cell Biochem Biophys 2024; 82:919-925. [PMID: 38480573 PMCID: PMC11344723 DOI: 10.1007/s12013-024-01243-7] [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] [Accepted: 02/24/2024] [Indexed: 08/25/2024]
Abstract
Apoptotic cell death occurs under normal physiological conditions, such as development, tissue remodeling, and inflammation. Appropriate removal of apoptotic cells by phagocytes and the secretion of anti-inflammatory cytokines such as IL-10 are important mechanisms for maintaining tissue homeostasis. Apoptotic cell phagocytosis is mediated by several phosphatidylserine recognition receptors on non-professional or professional phagocytes, such as neighboring epithelial cells or macrophages. Stabilin-2 is reported as a phosphatidylserine recognition receptor for apoptotic cell phagocytosis, and its downstream signaling pathway for cytoskeletal rearrangement for phagocytosis is well known. However, the mechanisms for stabilin-2-mediated IL-10 production has not yet been reported. In this study, we aimed to investigate stabilin-2 receptor-mediated IL-10 transcription regulation signaling pathway.
Collapse
Affiliation(s)
- Han-Seul Jo
- Department of Physiology, Cell and Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, South Korea.
| | - Ha-Jeong Kim
- Department of Physiology, Cell and Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, South Korea.
| |
Collapse
|
50
|
Iraji Asiabadi A, Esmaeil N, Zargar Kharazi A, Dabiri A, Varshosaz J. Harnessing IL-10 induced anti-inflammatory response in maturing macrophages in presence of electrospun dexamethasone-loaded PLLA scaffold. J Biomed Mater Res B Appl Biomater 2024; 112:e35411. [PMID: 38773758 DOI: 10.1002/jbm.b.35411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/19/2024] [Accepted: 04/13/2024] [Indexed: 05/24/2024]
Abstract
The ultimate goal of tissue engineering is to repair and regenerate damaged tissue or organ. Achieving this goal requires blood vessel networks to supply oxygen and nutrients to new forming tissues. Macrophages are part of the immune system whose behavior plays a significant role in angiogenesis and blood vessel formation. On the other hand, macrophages are versatile cells that change their behavior in response to environmental stimuli. Given that implantation of a biomaterial is followed by inflammation; therefore, we reasoned that this inflammatory condition in tissue spaces modulates the final phenotype of macrophages. Also, we hypothesized that anti-inflammatory glucocorticoid dexamethasone improves modulating macrophages behavior. To check these concepts, we investigated the macrophages that had matured in an inflammatory media. Furthermore, we examined macrophages' behavior after maturation on a dexamethasone-containing scaffold and analyzed how the behavioral change of maturing macrophages stimulates other macrophages in the same environment. In this study, the expression of pro-inflammatory markers TNFa and NFκB1 along with pro-healing markers IL-10 and CD163 were investigated to study the behavior of macrophages. Our results showed that macrophages that were matured in the inflammatory media in vitro increase expression of IL-10, which in turn decreased the expression of pro-inflammatory markers TNFa and NFκB in maturing macrophages. Also, macrophages that were matured on dexamethasone-containing scaffolds decreased the expression of IL-10, TNFa, and NFκB and increase the expression of CD163 compared to the control group. Moreover, the modulation of anti-inflammatory response in maturing macrophages on dexamethasone-containing scaffold resulted in increased expression of TNFa and CD163 by other macrophages in the same media. The results obtained in this study, proposing strategies to improve healing through controlling the behavior of maturing macrophages and present a promising perspective for inflammation control using tissue engineering scaffolds.
Collapse
Affiliation(s)
- Arash Iraji Asiabadi
- Tissue Engineering and Nanotechnology, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anousheh Zargar Kharazi
- Tissue Engineering and Nanotechnology, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arezou Dabiri
- Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|