1
|
Las Heras K, Garcia-Orue I, Rancan F, Igartua M, Santos-Vizcaino E, Hernandez RM. Modulating the immune system towards a functional chronic wound healing: A biomaterials and Nanomedicine perspective. Adv Drug Deliv Rev 2024; 210:115342. [PMID: 38797316 DOI: 10.1016/j.addr.2024.115342] [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: 01/26/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/29/2024]
Abstract
Chronic non-healing wounds persist as a substantial burden for healthcare systems, influenced by factors such as aging, diabetes, and obesity. In contrast to the traditionally pro-regenerative emphasis of therapies, the recognition of the immune system integral role in wound healing has significantly grown, instigating an approach shift towards immunological processes. Thus, this review explores the wound healing process, highlighting the engagement of the immune system, and delving into the behaviors of innate and adaptive immune cells in chronic wound scenarios. Moreover, the article investigates biomaterial-based strategies for the modulation of the immune system, elucidating how the adjustment of their physicochemical properties or their synergistic combination with other agents such as drugs, proteins or mesenchymal stromal cells can effectively modulate the behaviors of different immune cells. Finally this review explores various strategies based on synthetic and biological nanostructures, including extracellular vesicles, to finely tune the immune system as natural immunomodulators or therapeutic nanocarriers with promising biophysical properties.
Collapse
Affiliation(s)
- Kevin Las Heras
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Itxaso Garcia-Orue
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain
| | - Fiorenza Rancan
- Department of Dermatology, Venereology und Allergology,Clinical Research Center for Hair and Skin Science, Charité - Universitätsmedizin Berlin
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
2
|
Saadh MJ, Alfattah MA, Ismail AH, Saeed BA, Abbas HH, Elashmawy NF, Hashim GA, Ismail KS, Abo-Zaid MA, Waggiallah HA. The role of Interleukin-21 (IL-21) in allergic disorders: Biological insights and regulatory mechanisms. Int Immunopharmacol 2024; 134:111825. [PMID: 38723368 DOI: 10.1016/j.intimp.2024.111825] [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: 01/06/2024] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 06/03/2024]
Abstract
In recent decades, allergic diseases subsequent from an IgE-mediated response to specific allergens have become a progressively public chronic disease worldwide. They have shaped an important medical and socio-economic burden. A significant proportion of allergic disorders are branded via a form 2 immune response relating Th2 cells, type 2 natural lymphoid cells, mast cells and eosinophils. Interleukin-21 (IL-21) is a participant of the type-I cytokine family manufactured through numerous subsets of stimulated CD4+ T cells and uses controlling properties on a diversity of immune cells. Increasingly, experimental sign suggests a character for IL-21 in the pathogenesis of numerous allergic disorders. The purpose of this review is to discuss the biological properties of IL-21 and to summaries current developments in its role in the regulation of allergic disorders.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | - Mohammed A Alfattah
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Ahmed H Ismail
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Bashar Abdullah Saeed
- Department of Medical Laboratory Technics, Al-Noor University College, Nineveh, Iraq
| | | | - Nabila F Elashmawy
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Ghassan A Hashim
- Department of Nursing, Al-Zahrawi University College, Karbala, Iraq
| | - Khatib Sayeed Ismail
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia
| | - Mabrouk A Abo-Zaid
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan 45142, Saudi Arabia.
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| |
Collapse
|
3
|
Pan J, Yao WL, Liu LP, Wang BS, Chai WZ, Huang Z, Fan XP, He WH, Wang WH, Zhang WD. Moniezia benedeni infection increases IgE + cells in sheep (Ovis aries) small intestine. Vet Parasitol 2024; 328:110169. [PMID: 38520755 DOI: 10.1016/j.vetpar.2024.110169] [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: 10/17/2023] [Revised: 03/07/2024] [Accepted: 03/10/2024] [Indexed: 03/25/2024]
Abstract
The concentration of immunoglobulin (Ig) E is the lowest among serum Igs, but it can induces type I hypersensitivity and plays an important role in anti-parasitic infection. The present study aimed to explore the residence characteristics of IgE+ cells in the sheep small intestine and the impact of Moniezia benedeni infection on them. The recombinant plasmids pET-28a-IgE were constructed and induced and expressed in Escherichia coli. BL21 (DE3). The rabbit anti-sheep IgE polyclonal antibody was prepared using the obtained recombinant protein as antigen. Finally, the levels of IgE+ cells in the small intestine of healthy (Control group) and naturally M. benedeni-infected (Infected group) sheep were detected analyzed. The results showed that the rabbit anti-sheep IgE polyclonal antibody with good immunogenicity (titer = 1: 128000) could specifically bind to the heavy chain of natural sheep IgE. In the Control group, the IgE+ cells were mainly distributed in lamina propria of the small intestine, and the densities were significantly decreased from duodenum to ileum (P<0.05), with respective values of (4.28 cells / 104 μm2, 1.80 cells / 104 μm2, and 1.44 cells / 104 μm2 in duodenum, jejunum, and ileum. In the Infected group, IgE+ cells density were 6.26 cells / 104 μm2, 3.01 cells / 104 μm2, and 2.09 cells / 104 μm2 in duodenum, jejunum and ileum respectively, which were significantly higher in all segments compared to the Control group (P<0.05), increasing by 46.26%, 67.22% and 45.14%, respectively. In addition, compared with the Control group, the IgE protein levels were significantly increased in all intestinal segments of the Infected group (P<0.01), however, there was no significant differences among the different intestinal segments within the same group (P>0.05). The results demonstrated that M. benedeni infection could significantly increase the content of IgE and the distribution density of its secreting cells in sheep small intestine. The intestinal mucosal immune system of sheep presented obvious specificity against M. benedeni infection. This lays a good foundation for further exploring molecular mechanisms of the intestinal mucosal immune system monitoring and responding to M. benedeni infection.
Collapse
Affiliation(s)
- Jing Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Wan-Ling Yao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Li-Ping Liu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Bao-Shan Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Wen-Zhu Chai
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Zhen Huang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Xi-Ping Fan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Wan-Hong He
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Wen-Hui Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China
| | - Wang-Dong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu 730070, China.
| |
Collapse
|
4
|
Hu ST, Zhou G, Zhang J. Implications of innate lymphoid cells in oral diseases. Int Immunopharmacol 2024; 133:112122. [PMID: 38663313 DOI: 10.1016/j.intimp.2024.112122] [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: 02/26/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024]
Abstract
Innate lymphoid cells (ILCs), as newly discovered antigen-independent innate immune cells, respond promptly to stimuli by secreting effector cytokines to exert effector functions similar to those of T cells. ILCs predominantly reside at mucosal sites and play critical roles in defending against infections, maintaining mucosal homeostasis, regulating inflammatory and immune responses, and participating in tumorigenesis. Recently, there has been a growing interest in the role of ILCs in oral diseases. This review outlines the classifications and the major characteristics of ILCs, and then comprehensively expatiates the research on ILCs in oral cancer, primary Sjogren's syndrome, periodontal diseases, oral lichen planus, oral candidiasis, Behcet's disease, and pemphigus vulgaris, aiming at summarising the implications of ILCs in oral diseases and providing new ideas for further research.
Collapse
Affiliation(s)
- Si-Ting Hu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China
| | - Gang Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China; Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, China
| | - Jing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China; Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, China.
| |
Collapse
|
5
|
Ortega-Rodriguez AC, Guerra de Blas PDC, Ramírez-Torres R, Martínez-Shio EB, Monsiváis-Urenda AE. Quantitative Analysis of Innate Lymphoid Cells in Patients with ST-Segment Elevation Myocardial Infarction. Immunol Invest 2024; 53:586-603. [PMID: 38700235 DOI: 10.1080/08820139.2024.2316052] [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: 05/05/2024]
Abstract
BACKGROUND Acute myocardial infarction (AMI) is one of the principal causes of death in Mexico and worldwide. AMI triggers an acute inflammatory process that induces the activation of different populations of the innate immune system. Innate lymphoid cells (ILCs) are an innate immunity, highly pleiotropic population, which have been observed to participate in tissue repair and polarization of the adaptive immune response. OBJECTIVE We aimed to analyze the levels of subsets of ILCs in patients with ST-segment elevation myocardial infarction (STEMI), immediately 3 and 6 months post-AMI, and analyze their correlation with clinical parameters. RESULTS We evaluated 29 STEMI patients and 15 healthy controls and analyzed the different subsets of circulating ILCs, immediately 3 and 6 months post-AMI. We observed higher levels of circulating ILCs in STEMI patients compared to control subjects and a significant correlation between ILC levels and cardiac function. We also found increased production of the cytokines interleukin 5 (IL-5) and interleukin 17A (IL-17A), produced by ILC2 cells and by ILC3 cells, respectively, in the STEMI patients. CONCLUSION This study shows new evidence of the role of ILCs in the pathophysiology of AMI and their possible involvement in the maintenance of cardiac function.
Collapse
Affiliation(s)
- Alma Celeste Ortega-Rodriguez
- Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de la Salud y Biomedicina, Departamento de Inmunología, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Paola Del Carmen Guerra de Blas
- Coordinating Center, The Mexican Emerging Infectious Diseases Clinical Research Network (LaRed), Mexico City, Mexico
- Departamento de Infectología, Hospital Infantil de México Federico Gómez, Instituto Nacional de Salud, Mexico City, Mexico
| | - Ricardo Ramírez-Torres
- Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de la Salud y Biomedicina, Departamento de Inmunología, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Elena B Martínez-Shio
- Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de la Salud y Biomedicina, Departamento de Inmunología, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Adriana E Monsiváis-Urenda
- Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de la Salud y Biomedicina, Departamento de Inmunología, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| |
Collapse
|
6
|
Jahanbani F, Sing JC, Maynard RD, Jahanbani S, Dafoe J, Dafoe W, Jones N, Wallace KJ, Rastan A, Maecker HT, Röst HL, Snyder MP, Davis RW. Longitudinal cytokine and multi-modal health data of an extremely severe ME/CFS patient with HSD reveals insights into immunopathology, and disease severity. Front Immunol 2024; 15:1369295. [PMID: 38650940 PMCID: PMC11033372 DOI: 10.3389/fimmu.2024.1369295] [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: 01/12/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) presents substantial challenges in patient care due to its intricate multisystem nature, comorbidities, and global prevalence. The heterogeneity among patient populations, coupled with the absence of FDA-approved diagnostics and therapeutics, further complicates research into disease etiology and patient managment. Integrating longitudinal multi-omics data with clinical, health,textual, pharmaceutical, and nutraceutical data offers a promising avenue to address these complexities, aiding in the identification of underlying causes and providing insights into effective therapeutics and diagnostic strategies. Methods This study focused on an exceptionally severe ME/CFS patient with hypermobility spectrum disorder (HSD) during a period of marginal symptom improvements. Longitudinal cytokine profiling was conducted alongside the collection of extensive multi-modal health data to explore the dynamic nature of symptoms, severity, triggers, and modifying factors. Additionally, an updated severity assessment platform and two applications, ME-CFSTrackerApp and LexiTime, were introduced to facilitate real-time symptom tracking and enhance patient-physician/researcher communication, and evaluate response to medical intervention. Results Longitudinal cytokine profiling revealed the significance of Th2-type cytokines and highlighted synergistic activities between mast cells and eosinophils, skewing Th1 toward Th2 immune responses in ME/CFS pathogenesis, particularly in cognitive impairment and sensorial intolerance. This suggests a potentially shared underlying mechanism with major ME/CFS comorbidities such as HSD, Mast cell activation syndrome, postural orthostatic tachycardia syndrome (POTS), and small fiber neuropathy. Additionally, the data identified potential roles of BCL6 and TP53 pathways in ME/CFS etiology and emphasized the importance of investigating adverse reactions to medication and supplements and drug interactions in ME/CFS severity and progression. Discussion Our study advocates for the integration of longitudinal multi-omics with multi-modal health data and artificial intelligence (AI) techniques to better understand ME/CFS and its major comorbidities. These findings highlight the significance of dysregulated Th2-type cytokines in patient stratification and precision medicine strategies. Additionally, our results suggest exploring the use of low-dose drugs with partial agonist activity as a potential avenue for ME/CFS treatment. This comprehensive approach emphasizes the importance of adopting a patient-centered care approach to improve ME/CFS healthcare management, disease severity assessment, and personalized medicine. Overall, these findings contribute to our understanding of ME/CFS and offer avenues for future research and clinical practice.
Collapse
Affiliation(s)
- Fereshteh Jahanbani
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Justin Cyril Sing
- Department of Molecular Genetics, Donnelly Center, University of Toronto, Toronto, ON, Canada
| | - Rajan Douglas Maynard
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Shaghayegh Jahanbani
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Veterans Affairs (VA) Palo Alto Health Care System, Palo Alto, CA, United States
| | - Janet Dafoe
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Whitney Dafoe
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Nathan Jones
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Kelvin J. Wallace
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Azuravesta Rastan
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Holden T. Maecker
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Pulmonary and Critical Care Medicine, Institute of Immunity, Transplantation, and Infectious Diseases, Stanford University, Palo Alto, CA, United States
| | - Hannes L. Röst
- Department of Molecular Genetics, Donnelly Center, University of Toronto, Toronto, ON, Canada
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
| | - Ronald W. Davis
- ME/CFS Collaborative Research Center at Stanford, Stanford Genome Technology Center, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
7
|
CHEN JK, WU Y, LI WZ, HUANG MF, HUANG B, XIE CC, LI JQ, LU Y, CHEN Y. The regulatory effects of electroacupuncture on type 2 innate lymphoid cell (ILC2) function and browning of white adipose tissue in obese rats. WORLD JOURNAL OF ACUPUNCTURE-MOXIBUSTION 2024; 34:138-146. [DOI: 10.1016/j.wjam.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
8
|
Coulis G, Villalta SA. Muscle immune cells protect mitochondrial organelles during exercise. Nature 2024; 625:35-36. [PMID: 38110631 PMCID: PMC11414740 DOI: 10.1038/d41586-023-03972-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
|
9
|
Pokhreal D, Crestani B, Helou DG. Macrophage Implication in IPF: Updates on Immune, Epigenetic, and Metabolic Pathways. Cells 2023; 12:2193. [PMID: 37681924 PMCID: PMC10486697 DOI: 10.3390/cells12172193] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal interstitial lung disease of unknown etiology with a poor prognosis. It is a chronic and progressive disease that has a distinct radiological and pathological pattern from common interstitial pneumonia. The use of immunosuppressive medication was shown to be completely ineffective in clinical trials, resulting in years of neglect of the immune component. However, recent developments in fundamental and translational science demonstrate that immune cells play a significant regulatory role in IPF, and macrophages appear to be among the most crucial. These highly plastic cells generate multiple growth factors and mediators that highly affect the initiation and progression of IPF. In this review, we will provide an update on the role of macrophages in IPF through a systemic discussion of various regulatory mechanisms involving immune receptors, cytokines, metabolism, and epigenetics.
Collapse
Affiliation(s)
- Deepak Pokhreal
- Physiopathologie et Epidémiologie des Maladies Respiratoires, Inserm U1152, UFR de Médecine, Université Paris Cité, 75018 Paris, France
| | - Bruno Crestani
- Physiopathologie et Epidémiologie des Maladies Respiratoires, Inserm U1152, UFR de Médecine, Université Paris Cité, 75018 Paris, France
- FHU APOLLO, Service de Pneumologie A, Hôpital Bichat, Assistance Publique des Hôpitaux de Paris, 75877 Paris, France
| | - Doumet Georges Helou
- Physiopathologie et Epidémiologie des Maladies Respiratoires, Inserm U1152, UFR de Médecine, Université Paris Cité, 75018 Paris, France
| |
Collapse
|
10
|
Jan-Abu SC, Kabil A, McNagny KM. Parallel origins and functions of T cells and ILCs. Clin Exp Immunol 2023; 213:76-86. [PMID: 37235977 PMCID: PMC10324547 DOI: 10.1093/cei/uxad056] [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: 03/01/2023] [Revised: 04/19/2023] [Accepted: 05/26/2023] [Indexed: 05/28/2023] Open
Abstract
Innate lymphoid cells (ILCs) are tissue resident cells that are triggered through a relatively broad spectrum of alarmins, inflammatory cues, neuropeptides, and hormones. Functionally, ILCs are akin to subsets of helper T cells and are characterized by a similar effector cytokine profile. They also share a dependency on many of the same essential transcription factors identified for the maintenance and survival of T cells. The key distinguishing factor between the ILC family and T cells is the lack of antigen-specific T cell receptor (TCR) on ILCs and, thus, they can be considered the "ultimate invariant T cells". ILCs, like T cells, orchestrate downstream effector inflammatory responses by adjusting the cytokine microenvironment in a fashion that promotes protection, health, and homeostasis at mucosal barrier sites. But also, like T cells, ILCs have recently been implicated in several pathological inflammatory disease states. This review focuses on the selective role of ILCs in the development of allergic airway inflammation (AAI) and fibrosis in the gut where a complex ILC interplay has been shown to either attenuate or worsen disease. Finally, we discuss new data on TCR gene rearrangements in subsets of ILCs that challenge the current dogma linking their origin to committed bone marrow progenitors and instead propose a thymic origin for at least some ILCs. In addition, we highlight how naturally occurring TCR rearrangements and the expression of major histocompatibility (MHC) molecules in ILCs provide a useful natural barcode for these cells and may prove instrumental in studying their origins and plasticity.
Collapse
Affiliation(s)
- Sia C Jan-Abu
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Ahmed Kabil
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Kelly M McNagny
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Centre for Heart and Lung Innovation (HLI), St Paul’s Hospital, Vancouver, BC, Canada
| |
Collapse
|
11
|
Monitoring of the Forgotten Immune System during Critical Illness-A Narrative Review. Medicina (B Aires) 2022; 59:medicina59010061. [PMID: 36676685 PMCID: PMC9866378 DOI: 10.3390/medicina59010061] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/24/2022] [Accepted: 12/25/2022] [Indexed: 12/29/2022] Open
Abstract
Immune organ failure is frequent in critical illness independent of its cause and has been acknowledged for a long time. Most patients admitted to the ICU, whether featuring infection, trauma, or other tissue injury, have high levels of alarmins expression in tissues or systemically which then activate innate and adaptive responses. Although necessary, this response is frequently maladaptive and leads to organ dysfunction. In addition, the counter-response aiming to restore homeostasis and repair injury can also be detrimental and contribute to persistent chronic illness. Despite intensive research on this topic in the last 40 years, the immune system is not routinely monitored in critical care units. In this narrative review we will first discuss the inflammatory response after acute illness and the players of maladaptive response, focusing on neutrophils, monocytes, and T cells. We will then go through commonly used biomarkers, like C-reactive protein, procalcitonin and pancreatic stone protein (PSP) and what they monitor. Next, we will discuss the strengths and limitations of flow cytometry and related techniques as an essential tool for more in-depth immune monitoring and end with a presentation of the most promising cell associated markers, namely HLA-DR expression on monocytes, neutrophil expression of CD64 and PD-1 expression on T cells. In sum, immune monitoring critically ill patients is a forgotten and missing piece in the monitoring capacity of intensive care units. New technology, including bed-side equipment and in deep cell phenotyping using emerging multiplexing techniques will likely allow the definition of endotypes and a more personalized care in the future.
Collapse
|
12
|
Ding T, Ge S. Metabolic regulation of type 2 immune response during tissue repair and regeneration. J Leukoc Biol 2022; 112:1013-1023. [PMID: 35603496 DOI: 10.1002/jlb.3mr0422-665r] [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: 02/17/2022] [Revised: 04/26/2022] [Indexed: 12/24/2022] Open
Abstract
Type 2 immune responses are mediated by the cytokines interleukin (IL)-4, IL-5, IL-10, and IL-13 and associated cell types, including T helper (Th)2 cells, group 2 innate lymphoid cells (ILC2s), basophils, mast cells, eosinophils, and IL-4- and IL-13-activated macrophages. It can suppress type 1-driven autoimmune diseases, promote antihelminth immunity, maintain cellular metabolic homeostasis, and modulate tissue repair pathways following injury. However, when type 2 immune responses become dysregulated, they can be a significant pathogenesis of many allergic and fibrotic diseases. As such, there is an intense interest in studying the pathways that modulate type 2 immune response so as to identify strategies of targeting and controlling these responses for tissue healing. Herein, we review recent literature on the metabolic regulation of immune cells initiating type 2 immunity and immune cells involved in the effector phase, and talk about how metabolic regulation of immune cell subsets contribute to tissue repair. At last, we discuss whether these findings can provide a novel prospect for regenerative medicine.
Collapse
Affiliation(s)
- Tian Ding
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Shaohua Ge
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
13
|
Shichkin VP, Antica M. Key Factors for Thymic Function and Development. Front Immunol 2022; 13:926516. [PMID: 35844535 PMCID: PMC9280625 DOI: 10.3389/fimmu.2022.926516] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022] Open
Abstract
The thymus is the organ responsible for T cell development and the formation of the adaptive immunity function. Its multicellular environment consists mainly of the different stromal cells and maturing T lymphocytes. Thymus-specific progenitors of epithelial, mesenchymal, and lymphoid cells with stem cell properties represent only minor populations. The thymic stromal structure predominantly determines the function of the thymus. The stromal components, mostly epithelial and mesenchymal cells, form this specialized area. They support the consistent developmental program of functionally distinct conventional T cell subpopulations. These include the MHC restricted single positive CD4+ CD8- and CD4- CD8+ cells, regulatory T lymphocytes (Foxp3+), innate natural killer T cells (iNKT), and γδT cells. Several physiological causes comprising stress and aging and medical treatments such as thymectomy and chemo/radiotherapy can harm the thymus function. The present review summarizes our knowledge of the development and function of the thymus with a focus on thymic epithelial cells as well as other stromal components and the signaling and transcriptional pathways underlying the thymic cell interaction. These critical thymus components are significant for T cell differentiation and restoring the thymic function after damage to reach the therapeutic benefits.
Collapse
|
14
|
Liu GH, Zhuo XC, Huang YH, Liu HM, Wu RC, Kuo CJ, Chen NH, Chuang LP, Lin SW, Chen YL, Yang HY, Lee TY. Alterations in Gut Microbiota and Upregulations of VPAC2 and Intestinal Tight Junctions Correlate with Anti-Inflammatory Effects of Electroacupuncture in Colitis Mice with Sleep Fragmentation. BIOLOGY 2022; 11:biology11070962. [PMID: 36101343 PMCID: PMC9311573 DOI: 10.3390/biology11070962] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/14/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Along with the modernization of society and people getting older, sleep disturbances and gut health have been identified as two key factors influencing aging, with dramatic effects on immunity and metabolism. Sleep is closely related to the gut, reflects the degree of chronic inflammation, and is associated with many degenerative diseases, hence the term “inflammaging”. This article addresses how sleep fragmentation affects the inflammatory state of the gut and elucidates the effects of restorative sleep and acupuncture on inflammatory gut remodeling and gut microbial recovery. In summary, fragmented sleep disrupted intestinal repair in mice with colitis, while electroacupuncture demonstrated likely results in alleviating colon inflammation, including maintaining colon length and daily body weight changes. In addition, the structure of the microbiota changed with decreasing gut inflammatory status. The intestinal tight junction proteins may be the key mechanism of electroacupuncture in treating sleep-fragmented ulcerative colitis mice. Electroacupuncture affects VIP through VPAC2 and further regulates intestinal mucosal immunity. This experiment demonstrates how physical stimulation stabilizes the intestinal epithelium and exerts an important anti-inflammatory effect. Abstract The relationship between inflammatory bowel disease and sleep disturbances is complicated and of increasing interest. We investigated the inflammatory and immunological consequences of EA in sleep-deprived colitis and found that dextran sulfate sodium (DSS)-induced colitis in sleep-fragmented (SF) mice was more severe than that in mice with normal sleep. This increase in the severity of colitis was accompanied by reduced body weight, shortened colon length, and deteriorated disease activity index. DSS with SF mice presented obvious diminished intestinal tight junction proteins (claudin-1 and occludin), elevated proinflammatory cytokines (CRP, IFN-γ, IL-6), lowered melatonin and adiponectin levels, downregulated vasoactive intestinal peptide (VIP) type 1 and 2 receptor (VPAC1, VPAC2) expression, and decreased diversity of gut bacteria. EA ameliorated colitis severity and preserved the performance of the epithelial tight junction proteins and VIP receptors, especially VPAC2. Meanwhile, the innate lymphoid cells-derived cytokines in both group 2 (IL-4, IL5, IL-9, IL-13) and group 3 (IL-22, GM-CSF) were elevated in mice colon tissue. Furthermore, dysbiosis was confirmed in the DSS group with and without SF, and EA could maintain the species diversity. Firmicutes could be restored, such as Lachnospiraceae, and Proteobacteria become rebalanced, mainly Enterobacteriaceae, after EA intervention. On the other hand, SF plays different roles in physiological and pathological conditions. In normal mice, interrupted sleep did not affect the expression of claudin-1 and occludin. But VPAC1, VPAC2, and gut microbiota diversity, including Burkholderiaceae and Rhodococcus, were opposite to mice in an inflamed state.
Collapse
Affiliation(s)
- Geng-Hao Liu
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan; (G.-H.L.); (R.-C.W.); (N.-H.C.)
- Graduate Institute of Traditional Chinese Medicine, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan;
- Division of Acupuncture and Moxibustion, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan;
- Sleep Center, Chang Gung Memorial Hospital, Taoyuan 333008, Taiwan; (L.-P.C.); (S.-W.L.)
| | - Xin-Cheng Zhuo
- Department of General Medicine, Taipei Medical University Hospital, Taipei 110301, Taiwan;
| | - Yueh-Hsiang Huang
- Division of Chinese Internal Medicine, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taipei 105406, Taiwan;
| | - Hsuan-Miao Liu
- Graduate Institute of Traditional Chinese Medicine, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan;
| | - Ren-Chin Wu
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan; (G.-H.L.); (R.-C.W.); (N.-H.C.)
- Department of Anatomic Pathology, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
| | - Chia-Jung Kuo
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan;
| | - Ning-Hung Chen
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan; (G.-H.L.); (R.-C.W.); (N.-H.C.)
- Sleep Center, Chang Gung Memorial Hospital, Taoyuan 333008, Taiwan; (L.-P.C.); (S.-W.L.)
- Department of Pulmonary and Critical Care Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
| | - Li-Pang Chuang
- Sleep Center, Chang Gung Memorial Hospital, Taoyuan 333008, Taiwan; (L.-P.C.); (S.-W.L.)
- Department of Pulmonary and Critical Care Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
| | - Shih-Wei Lin
- Sleep Center, Chang Gung Memorial Hospital, Taoyuan 333008, Taiwan; (L.-P.C.); (S.-W.L.)
- Department of Pulmonary and Critical Care Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
| | - Yen-Lung Chen
- Division of Acupuncture and Moxibustion, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan;
- Sleep Center, Chang Gung Memorial Hospital, Taoyuan 333008, Taiwan; (L.-P.C.); (S.-W.L.)
| | - Huang-Yu Yang
- Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21287, USA
- Correspondence: (H.-Y.Y.); (T.-Y.L.); Tel.: +886-03-328-1200 (ext. 8181) (H.-Y.Y.); +886-03-211-8800 (ext. 3537) (T.-Y.L.)
| | - Tzung-Yan Lee
- Graduate Institute of Traditional Chinese Medicine, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan;
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung 204201, Taiwan
- Correspondence: (H.-Y.Y.); (T.-Y.L.); Tel.: +886-03-328-1200 (ext. 8181) (H.-Y.Y.); +886-03-211-8800 (ext. 3537) (T.-Y.L.)
| |
Collapse
|
15
|
Crosstalk between ILC2s and Th2 CD4+ T Cells in Lung Disease. J Immunol Res 2022; 2022:8871037. [PMID: 35592688 PMCID: PMC9113865 DOI: 10.1155/2022/8871037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/30/2022] [Accepted: 04/18/2022] [Indexed: 12/03/2022] Open
Abstract
Cytokine secretion, such as interleukin-4 (IL-4), IL-5, IL-9, IL-13, and amphiregulin (Areg), by type 2 innate lymphoid cells (ILC2s) is indispensable for homeostasis, remodeling/repairing tissue structure, inflammation, and tumor immunity. Often viewed as the innate cell surrogate of T helper type 2 (Th2) cells, ILC2s not only secrete the same type 2 cytokines, but are also inextricably related to CD4+T cells in terms of cell origin and regulatory factors, bridging between innate and adaptive immunity. ILC2s interact with CD4+T cells to play a leading role in a variety of diseases through secretory factors. Here, we review the latest progress on ILC2s and CD4+T cells in the lung, the close relationship between the two, and their relevance in the lung disease and immunity. This literature review aids future research in pulmonary type 2 immune diseases and guides innovative treatment approaches for these diseases.
Collapse
|
16
|
Newnes HV, Armitage JD, Audsley KM, Bosco A, Waithman J. Directing the Future Breakthroughs in Immunotherapy: The Importance of a Holistic Approach to the Tumour Microenvironment. Cancers (Basel) 2021; 13:cancers13235911. [PMID: 34885021 PMCID: PMC8656826 DOI: 10.3390/cancers13235911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Immunotherapies have changed the way we treat cancer and, while some patients have benefitted greatly, there are still those that do not respond to therapy. Understanding why some patients respond to therapy and others do not is critical in developing new immunotherapeutic strategies. The increasing awareness of the importance of investigating the tumour in its entirety, including the surrounding tissue and role of various immune cells is helping to differentiate responders and non-responders. In addition, the resolution gained by the development of sophisticated bioinformatic technologies allows for a deeper understanding of the complex roles of individual cells in the tumour. This advancement will be critical for the development of novel therapies to treat cancer. Abstract Immunotherapy has revolutionised the treatment of cancers by exploiting the immune system to eliminate tumour cells. Despite the impressive response in a proportion of patients, clinical benefit has been limited thus far. A significant focus to date has been the identification of specific markers associated with response to immunotherapy. Unfortunately, the heterogeneity between patients and cancer types means identifying markers of response to therapy is inherently complex. There is a growing appreciation for the role of the tumour microenvironment (TME) in directing response to immunotherapy. The TME is highly heterogeneous and contains immune, stromal, vascular and tumour cells that all communicate and interact with one another to form solid tumours. This review analyses major cell populations present within the TME with a focus on their diverse and often contradictory roles in cancer and how this informs our understanding of immunotherapy. Furthermore, we discuss the role of integrated omics in providing a comprehensive view of the TME and demonstrate the potential of leveraging multi-omics to decipher the underlying mechanisms of anti-tumour immunity for the development of novel immunotherapeutic strategies.
Collapse
|
17
|
Abaricia JO, Farzad N, Heath TJ, Simmons J, Morandini L, Olivares-Navarrete R. Control of innate immune response by biomaterial surface topography, energy, and stiffness. Acta Biomater 2021; 133:58-73. [PMID: 33882355 DOI: 10.1016/j.actbio.2021.04.021] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/27/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022]
Abstract
As the focus of implantable biomaterials has shifted from bioinert implants to bioactive designs, recent research has highlighted the complex interactions between cell physiologic systems and material properties, particularly physical cues. From the cells known to interact with implanted biomaterials, the response of the immune system has been a critical target of study recently. Here, we review studies characterizing the response of innate immune cells to various material cues, particularly of those at the surface of implanted materials.The innate immune system consists of cell types with various roles in inflammation. Neutrophils and macrophages serve both phagocytic and signaling roles, especially early in the inflammatory phase of biomaterial implantation. These cell types ultimately dictate the outcome of implants as chronic inflammation, fibrosis, or integration. Other cell types like dendritic cells, mast cells, natural killer cells, and innate lymphoid cells may also serve an immunomodulatory role in the biomaterial context. This review highlights recent advances in our understanding of the role of innate immunity in the response to implantable biomaterials as well as key mechanobiological findings in innate immune cells underpinning these advances. STATEMENT OF SIGNIFICANCE: This review highlights recent advances in the understanding of the role of innate immunity in the response to implantable biomaterials, especially in neutrophils and macrophages, as well as key mechanobiological findings in innate immune cells underpinning these advances. Here we discuss how physicochemical properties of biomaterials control innate immune cell behavior.
Collapse
|
18
|
BCG for the prevention and treatment of allergic asthma. Vaccine 2021; 39:7341-7352. [PMID: 34417052 DOI: 10.1016/j.vaccine.2021.07.092] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/20/2021] [Accepted: 07/30/2021] [Indexed: 12/30/2022]
Abstract
Allergic diseases, in particular atopic asthma, have been on the rise in most industrialized countries for several decades now. Allergic asthma is characterized by airway narrowing, bronchial hyperresponsiveness, excessive airway mucus production, eosinophil influx in the lungs and an imbalance of the Th1/Th2 responses, including elevated IgE levels. Most available interventions provide only short-term relief from disease symptoms and do not alter the underlying immune imbalance. A number of studies, mostly in mouse models, have shown that Mycobacterium bovis bacillus Calmette-Guérin (BCG) treatment is capable of preventing or reducing an established allergen-driven inflammatory response, by redirecting pathogenic Th2 towards protective Th1 and/or regulatory T cell responses. Dendritic cells stimulated by BCG appear to be a crucial first step in the immunomodulatory effects of BCG. While the protective and therapeutic effects of BCG against allergy and asthma are well documented in animal models, they are less clear in humans, both in observational studies and in randomized controlled trials. The purpose of this article is to provide an up-to-date overview of the available evidence on the anti-allergy, in particular anti-asthma effects of BCG in mice, rats and humans.
Collapse
|
19
|
Atkinson SP. A preview of selected articles. Stem Cells Transl Med 2021. [PMCID: PMC8133349 DOI: 10.1002/sctm.21-0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
20
|
Shin SB, McNagny KM. ILC-You in the Thymus: A Fresh Look at Innate Lymphoid Cell Development. Front Immunol 2021; 12:681110. [PMID: 34025680 PMCID: PMC8136430 DOI: 10.3389/fimmu.2021.681110] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/20/2021] [Indexed: 01/20/2023] Open
Abstract
The discovery of innate lymphoid cells (ILCs) has revolutionized our understanding of innate immunity and immune cell interactions at epithelial barrier sites. Their presence and maintenance are critical for modulating immune homeostasis, responding to injury or infection, and repairing damaged tissues. To date, ILCs have been defined by a set of transcription factors, surface antigens and cytokines, and their functions resemble those of three major classes of helper T cell subsets, Th1, Th2 and Th17. Despite this, the lack of antigen-specific surface receptors and the notion that ILCs can develop in the absence of the thymic niche have clearly set them apart from the T-cell lineage and promulgated a dogma that ILCs develop directly from progenitors in the bone marrow. Interestingly however, emerging studies have challenged the BM-centric view of adult ILC development and suggest that ILCs could arise neonatally from developing T cell progenitors. In this review, we discuss ILC development in parallel to T-cell development and summarize key findings that support a T-cell-centric view of ILC ontogeny.
Collapse
Affiliation(s)
- Samuel B Shin
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Kelly M McNagny
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Theret M, Rossi FMV, Contreras O. Evolving Roles of Muscle-Resident Fibro-Adipogenic Progenitors in Health, Regeneration, Neuromuscular Disorders, and Aging. Front Physiol 2021; 12:673404. [PMID: 33959042 PMCID: PMC8093402 DOI: 10.3389/fphys.2021.673404] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Normal skeletal muscle functions are affected following trauma, chronic diseases, inherited neuromuscular disorders, aging, and cachexia, hampering the daily activities and quality of life of the affected patients. The maladaptive accumulation of fibrous intramuscular connective tissue and fat are hallmarks of multiple pathologies where chronic damage and inflammation are not resolved, leading to progressive muscle replacement and tissue degeneration. Muscle-resident fibro-adipogenic progenitors are adaptable stromal cells with multilineage potential. They are required for muscle homeostasis, neuromuscular integrity, and tissue regeneration. Fibro-adipogenic progenitors actively regulate and shape the extracellular matrix and exert immunomodulatory functions via cross-talk with multiple other residents and non-resident muscle cells. Remarkably, cumulative evidence shows that a significant proportion of activated fibroblasts, adipocytes, and bone-cartilage cells, found after muscle trauma and disease, descend from these enigmatic interstitial progenitors. Despite the profound impact of muscle disease on human health, the fibrous, fatty, and ectopic bone tissues' origins are poorly understood. Here, we review the current knowledge of fibro-adipogenic progenitor function on muscle homeostatic integrity, regeneration, repair, and aging. We also discuss how scar-forming pathologies and disorders lead to dysregulations in their behavior and plasticity and how these stromal cells can control the onset and severity of muscle loss in disease. We finally explore the rationale of improving muscle regeneration by understanding and modulating fibro-adipogenic progenitors' fate and behavior.
Collapse
Affiliation(s)
- Marine Theret
- Biomedical Research Centre, Department of Medical Genetics, School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Fabio M. V. Rossi
- Biomedical Research Centre, Department of Medical Genetics, School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Osvaldo Contreras
- Departamento de Biología Celular y Molecular, Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, Australia
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| |
Collapse
|
22
|
Garn H, Potaczek DP, Pfefferle PI. The Hygiene Hypothesis and New Perspectives-Current Challenges Meeting an Old Postulate. Front Immunol 2021; 12:637087. [PMID: 33815389 PMCID: PMC8012489 DOI: 10.3389/fimmu.2021.637087] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/04/2021] [Indexed: 01/11/2023] Open
Abstract
During its 30 years history, the Hygiene Hypothesis has shown itself to be adaptable whenever it has been challenged by new scientific developments and this is a still a continuously ongoing process. In this regard, the mini review aims to discuss some selected new developments in relation to their impact on further fine-tuning and expansion of the Hygiene Hypothesis. This will include the role of recently discovered classes of innate and adaptive immune cells that challenges the old Th1/Th2 paradigm, the applicability of the Hygiene Hypothesis to newly identified allergy/asthma phenotypes with diverse underlying pathomechanistic endotypes, and the increasing knowledge derived from epigenetic studies that leads to better understanding of mechanisms involved in the translation of environmental impacts on biological systems. Further, we discuss in brief the expansion of the Hygiene Hypothesis to other disease areas like psychiatric disorders and cancer and conclude that the continuously developing Hygiene Hypothesis may provide a more generalized explanation for health burden in highly industrialized countries also relation to global changes.
Collapse
Affiliation(s)
- Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Biochemical Pharmacological Center (BPC), Philipps University of Marburg, Marburg, Germany.,German Center for Lung Research (DZL), Marburg, Germany
| | - Daniel Piotr Potaczek
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Biochemical Pharmacological Center (BPC), Philipps University of Marburg, Marburg, Germany
| | - Petra Ina Pfefferle
- German Center for Lung Research (DZL), Marburg, Germany.,Comprehensive Biobank Marburg (CBBMR), Medical Faculty, Philipps University of Marburg, Marburg, Germany.,German Biobank Alliance (GBA), Marburg, Germany
| |
Collapse
|
23
|
Yip W, Hughes MR, Li Y, Cait A, Hirst M, Mohn WW, McNagny KM. Butyrate Shapes Immune Cell Fate and Function in Allergic Asthma. Front Immunol 2021; 12:628453. [PMID: 33659009 PMCID: PMC7917140 DOI: 10.3389/fimmu.2021.628453] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/25/2021] [Indexed: 12/19/2022] Open
Abstract
The microbiome plays a fundamental role in how the immune system develops and how inflammatory responses are shaped and regulated. The “gut-lung axis” is a relatively new term that highlights a crucial biological crosstalk between the intestinal microbiome and lung. A growing body of literature suggests that dysbiosis, perturbation of the gut microbiome, is a driving force behind the development, and severity of allergic asthma. Animal models have given researchers new insights into how gut microbe-derived components and metabolites, such as short-chain fatty acids (SCFAs), influence the development of asthma. While the full understanding of how SCFAs influence allergic airway disease remains obscure, a recurring theme of epigenetic regulation of gene expression in several immune cell compartments is emerging. This review will address our current understanding of how SCFAs, and specifically butyrate, orchestrates cell behavior, and epigenetic changes and will provide a detailed overview of the effects of these modifications on immune cells in the context of allergic airway disease.
Collapse
Affiliation(s)
- William Yip
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada.,The Biomedical Research Centre, The University of British Columbia, Vancouver, BC, Canada
| | - Michael R Hughes
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada.,The Biomedical Research Centre, The University of British Columbia, Vancouver, BC, Canada
| | - Yicong Li
- The Biomedical Research Centre, The University of British Columbia, Vancouver, BC, Canada
| | - Alissa Cait
- Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada
| | - Martin Hirst
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada.,Michael Smith Laboratories, The University of British Columbia, Vancouver, BC, Canada
| | - William W Mohn
- Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada
| | - Kelly M McNagny
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada.,The Biomedical Research Centre, The University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
Bruton K, Koenig JFE, Phelps A, Jordana M. Perturbations to Homeostasis in Experimental Models Revealed Innate Pathways Driving Food Allergy. Front Immunol 2020; 11:603272. [PMID: 33362786 PMCID: PMC7758527 DOI: 10.3389/fimmu.2020.603272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
While type 2 immunity has been conventionally viewed as beneficial against helminths, venoms, and poisons, and harmful in allergy, contemporary research has uncovered its critical role in the maintenance of homeostasis. The initiation of a type 2 immune response involves an intricate crosstalk between structural and immune cells. Structural cells react to physical and chemical tissue perturbations by secreting alarmins, which signal the innate immune system to restore homeostasis. This pathway acts autonomously in the context of sterile injury and in the presence of foreign antigen initiates an adaptive Th2 response that is beneficial in the context of venoms, toxins, and helminths, but not food allergens. The investigation of the triggers and mechanisms underlying food allergic sensitization in humans is elusive because sensitization is a silent process. Therefore, the central construct driving food allergy modeling is based on introducing perturbations of tissue homeostasis along with an allergen which will result in an immunological and clinical phenotype that is consistent with that observed in humans. The collective evidence from multiple models has revealed the pre-eminent role of innate cells and molecules in the elicitation of allergic sensitization. We posit that, with the expanding use of technologies capable of producing formidable datasets, models of food allergy will continue to have an indispensable role to delineate mechanisms and establish causal relationships.
Collapse
Affiliation(s)
| | | | | | - Manel Jordana
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton ON, Canada
| |
Collapse
|
25
|
Lei A, He Y, Yang Q, Li X, Li R. Role of myeloid cells in the regulation of group 2 innate lymphoid cell-mediated allergic inflammation. Immunology 2020; 161:18-24. [PMID: 32609880 DOI: 10.1111/imm.13232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/15/2020] [Accepted: 06/20/2020] [Indexed: 12/11/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are an important component of the innate immune system that execute important effector functions at barrier surfaces, such as lung and skin. Like T helper type 2 cells, ILC2s are able to release high amounts of type 2 cytokines that are essential in inducing allergic inflammation and eliminating helminth infections. The past few years have contributed to our better understanding of the interactions between ILC2s and other cells of the immune system via soluble factors or in a cell-cell contact manner. Myeloid cells, including mononuclear leukocytes and polymorphonuclear leukocytes, are excellent sensors of tissue damage and infection and can influence ILC2 responses in the process of allergic inflammation. In this review, we summarize recent insights on how myeloid cell subsets regulate ILC2 activation with focus on soluble factors in the context of allergic inflammation.
Collapse
Affiliation(s)
- Aihua Lei
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Yumei He
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qiong Yang
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaofang Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Ranhui Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
26
|
Affiliation(s)
- Jorge C Correia
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jorge L Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|