1
|
Rosain J, Le Voyer T, Liu X, Gervais A, Polivka L, Cederholm A, Berteloot L, Parent AV, Pescatore A, Spinosa E, Minic S, Kiszewski AE, Tsumura M, Thibault C, Esnaola Azcoiti M, Martinovic J, Philippot Q, Khan T, Marchal A, Charmeteau-De Muylder B, Bizien L, Deswarte C, Hadjem L, Fauvarque MO, Dorgham K, Eriksson D, Falcone EL, Puel M, Ünal S, Geraldo A, Le Floc'h C, Li H, Rheault S, Muti C, Bobrie-Moyrand C, Welfringer-Morin A, Fuleihan RL, Lévy R, Roelens M, Gao L, Materna M, Pellegrini S, Piemonti L, Catherinot E, Goffard JC, Fekkar A, Sacko-Sow A, Soudée C, Boucherit S, Neehus AL, Has C, Hübner S, Blanchard-Rohner G, Amador-Borrero B, Utsumi T, Taniguchi M, Tani H, Izawa K, Yasumi T, Kanai S, Migaud M, Aubart M, Lambert N, Gorochov G, Picard C, Soudais C, L'Honneur AS, Rozenberg F, Milner JD, Zhang SY, Vabres P, Trpinac D, Marr N, Boddaert N, Desguerre I, Pasparakis M, Miller CN, Poziomczyk CS, Abel L, Okada S, Jouanguy E, Cheynier R, Zhang Q, Cobat A, Béziat V, Boisson B, Steffann J, Fusco F, Ursini MV, Hadj-Rabia S, Bodemer C, Bustamante J, Luche H, Puel A, Courtois G, Bastard P, Landegren N, Anderson MS, Casanova JL. Incontinentia pigmenti underlies thymic dysplasia, autoantibodies to type I IFNs, and viral diseases. J Exp Med 2024; 221:e20231152. [PMID: 39352576 PMCID: PMC11448874 DOI: 10.1084/jem.20231152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/17/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024] Open
Abstract
Human inborn errors of thymic T cell tolerance underlie the production of autoantibodies (auto-Abs) neutralizing type I IFNs, which predispose to severe viral diseases. We analyze 131 female patients with X-linked dominant incontinentia pigmenti (IP), heterozygous for loss-of-function (LOF) NEMO variants, from 99 kindreds in 10 countries. Forty-seven of these patients (36%) have auto-Abs neutralizing IFN-α and/or IFN-ω, a proportion 23 times higher than that for age-matched female controls. This proportion remains stable from the age of 6 years onward. On imaging, female patients with IP have a small, abnormally structured thymus. Auto-Abs against type I IFNs confer a predisposition to life-threatening viral diseases. By contrast, patients with IP lacking auto-Abs against type I IFNs are at no particular risk of viral disease. These results suggest that IP accelerates thymic involution, thereby underlying the production of auto-Abs neutralizing type I IFNs in at least a third of female patients with IP, predisposing them to life-threatening viral diseases.
Collapse
Affiliation(s)
- Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris (AP-HP) , Paris, France
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Clinical Immunology Department, AP-HP, Saint-Louis Hospital, Paris, France
| | - Xian Liu
- Diabetes Center, University of California San Francisco , San Francisco, CA, USA
| | - Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Laura Polivka
- Department of Dermatology, Reference Center for Genodermatosis and Rare Skin Diseases (MAGEC), University of Paris Cité, Necker Hospital for Sick Children, AP-HP, Paris, France
- Reference Center for Mastocytosis (CEREMAST), Necker Hospital for Sick Children, AP-HP , Paris, France
| | - Axel Cederholm
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Laureline Berteloot
- Pediatric Radiology Department, Necker Hospital for Sick Children, Imagine Inserm Institute, U1163, AP-HP, Paris, France
| | - Audrey V Parent
- Diabetes Center, University of California San Francisco , San Francisco, CA, USA
| | - Alessandra Pescatore
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso," IGB-CNR , Naples, Italy
| | - Ezia Spinosa
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso," IGB-CNR , Naples, Italy
| | - Snezana Minic
- Clinics of Dermatovenerology, Clinical Center of Serbia , Belgrade, Serbia
- School of Medicine, University of Belgrade , Belgrade, Serbia
| | - Ana Elisa Kiszewski
- Section of Dermatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
- Section of Pediatric Dermatology, Hospital da Criança Santo Antônio, Irmandade da Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, Brazil
| | - Miyuki Tsumura
- Hiroshima University Graduate School of Biomedical and Health Sciences , Hiroshima, Japan
| | - Chloé Thibault
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Maria Esnaola Azcoiti
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Jelena Martinovic
- Unit of Fetal Pathology, Hospital Antoine Béclère, Paris Saclay University , Paris, France
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Taushif Khan
- Department of Immunology, Sidra Medicine, Doha, Qatar
| | - Astrid Marchal
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | | | - Lucy Bizien
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Caroline Deswarte
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Lillia Hadjem
- Immunophenomics Center (CIPHE), Aix Marseille University, Inserm, CNRS , Marseille, France
| | | | - Karim Dorgham
- Sorbonne University, Inserm, Centre for Immunology and Microbial Infections, CIMI-Paris , Paris, France
| | - Daniel Eriksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Emilia Liana Falcone
- Center for Immunity, Inflammation and Infectious Diseases, Montréal Clinical Research Institute (IRCM) , Montréal, Canada
- Department of Medicine, Montréal University, Montréal, Canada
| | - Mathilde Puel
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris (AP-HP) , Paris, France
| | - Sinem Ünal
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Amyrath Geraldo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Corentin Le Floc'h
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Hailun Li
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Sylvie Rheault
- Department of Medicine, Montréal University, Montréal, Canada
- Center of Research of the Geriatric University Institute of Montréal, University of Montréal , Montréal, Canada
| | - Christine Muti
- Department of Genetics, André Mignot Hospital, Versailles, France
| | | | - Anne Welfringer-Morin
- Department of Dermatology, Reference Center for Genodermatosis and Rare Skin Diseases (MAGEC), University of Paris Cité, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Ramsay L Fuleihan
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Romain Lévy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Marie Roelens
- Imagine Institute, University of Paris Cité , Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris (AP-HP) , Paris, France
| | - Liwei Gao
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Marie Materna
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Silvia Pellegrini
- Diabetes Research Institute, IRCCS Ospedale San Raffaele , Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele , Milan, Italy
| | | | - Jean-Christophe Goffard
- Internal Medicine, Brussels University Hospital, Free University of Brussels, Anderlecht, Belgium
| | - Arnaud Fekkar
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- Department of Parasitology Mycology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Aissata Sacko-Sow
- Department of Pediatrics, Jean Verdier Hospital, AP-HP, Bondy, France
| | - Camille Soudée
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Soraya Boucherit
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Cristina Has
- Department of Dermatology, Medical Center-University of Freiburg, Freiburg im Breisgau, Germany
- European Reference Network (ERN) for Rare and Undiagnosed Skin Disorders
| | - Stefanie Hübner
- Department of Dermatology, Medical Center-University of Freiburg, Freiburg im Breisgau, Germany
| | - Géraldine Blanchard-Rohner
- Unit of Immunology, Vaccinology, and Rheumatology, Division of General Pediatrics, Department of Woman, Child, and Adolescent Medicine, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Blanca Amador-Borrero
- Internal Medicine Department, Lariboisière Hospital, AP-HP, University of Paris Cité, Paris, France
| | - Takanori Utsumi
- Hiroshima University Graduate School of Biomedical and Health Sciences , Hiroshima, Japan
| | - Maki Taniguchi
- Hiroshima University Graduate School of Biomedical and Health Sciences , Hiroshima, Japan
| | - Hiroo Tani
- Department of Pediatrics, Hiroshima University Hospital, Hiroshima, Japan
- Department of Pediatrics, Hiroshima Prefectural Hospital, Hiroshima, Japan
| | - Kazushi Izawa
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahiro Yasumi
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sotaro Kanai
- Division of Child Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Mélanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
| | - Mélodie Aubart
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- Departments of Pediatric Neurology, Necker Hospital for Sick Children, AP-HP, University of Paris Cité, Paris, France
| | - Nathalie Lambert
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris (AP-HP) , Paris, France
| | - Guy Gorochov
- Sorbonne University, Inserm, Centre for Immunology and Microbial Infections, CIMI-Paris , Paris, France
- Department of Immunology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Capucine Picard
- Imagine Institute, University of Paris Cité , Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris (AP-HP) , Paris, France
- Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, Inserm U1163, Paris, France
| | - Claire Soudais
- Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, Inserm U1163, Paris, France
| | - Anne-Sophie L'Honneur
- Department of Virology, University of Paris Cité and Cochin Hospital, AP-HP, Paris, France
| | - Flore Rozenberg
- Department of Virology, University of Paris Cité and Cochin Hospital, AP-HP, Paris, France
| | - Joshua D Milner
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Pierre Vabres
- MAGEC Reference Center for Rare Skin Diseases, Dijon Bourgogne University Hospital, Dijon, France
| | - Dusan Trpinac
- Institute of Histology and Embryology, School of Medicine, University of Belgrade , Belgrade, Serbia
| | - Nico Marr
- Department of Immunology, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University , Doha, Qatar
| | - Nathalie Boddaert
- Pediatric Radiology Department, Necker Hospital for Sick Children, Imagine Inserm Institute, U1163, AP-HP, Paris, France
| | - Isabelle Desguerre
- Departments of Pediatric Neurology, Necker Hospital for Sick Children, AP-HP, University of Paris Cité, Paris, France
| | | | - Corey N Miller
- Diabetes Center, University of California San Francisco , San Francisco, CA, USA
| | | | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Satoshi Okada
- Hiroshima University Graduate School of Biomedical and Health Sciences , Hiroshima, Japan
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Rémi Cheynier
- University of Paris Cité, CNRS, Inserm, Institut Cochin , Paris, France
| | - Qian Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Bertrand Boisson
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Julie Steffann
- Department of Genomic Medicine, Necker Hospital for Sick Children, AP-HP, University of Paris Cité, Paris, France
| | - Francesca Fusco
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso," IGB-CNR , Naples, Italy
| | - Matilde Valeria Ursini
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso," IGB-CNR , Naples, Italy
| | - Smail Hadj-Rabia
- Department of Dermatology, Reference Center for Genodermatosis and Rare Skin Diseases (MAGEC), University of Paris Cité, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Christine Bodemer
- Department of Dermatology, Reference Center for Genodermatosis and Rare Skin Diseases (MAGEC), University of Paris Cité, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris (AP-HP) , Paris, France
| | - Hervé Luche
- Immunophenomics Center (CIPHE), Aix Marseille University, Inserm, CNRS , Marseille, France
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Gilles Courtois
- University Grenoble Alpes, CEA, Inserm , BGE UA13, Grenoble, France
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Center for Molecular Medicine, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Mark S Anderson
- Diabetes Center, University of California San Francisco , San Francisco, CA, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris Cité , Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, Paris, France
- Howard Hughes Medical Institute , New York, NY, USA
| |
Collapse
|
2
|
Suster D, Suster S. On the Histologic Classification of Thymoma. Adv Anat Pathol 2024; 31:22-33. [PMID: 37702296 DOI: 10.1097/pap.0000000000000412] [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: 09/14/2023]
Abstract
The classification of thymoma continues to be a source of controversy in pathology. The difficulties in histologic classification are evident from the number of proposals that have been offered over the years, as well as for the continuous changes and modifications introduced by the World Health Organization to their classification system over the past 20 years. We analyze here some of the issues involved in the classification of these tumors and the difficulties encountered for practicing pathologists in deciphering the "letters and numbers" system devised by the World Health Organization. We would like to propose an alternate approach to thymoma histologic classification that capitalizes on the basic observation of their cytologic features and incorporates the pattern of growth resulting from the interplay of the tumor cells with other cellular constituents as a secondary characteristic. The proposed histologic classification provides a simplified, reproducible means of histologically categorizing these tumors and can be easily understood by most practicing pathologists in simple and clear morphologic terms.
Collapse
Affiliation(s)
- David Suster
- Department of Pathology, Rutgers University Hospital, Newark, NJ
| | - Saul Suster
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
3
|
Alamdaran SA, Mahdavi Rashed M, Yekta M, Teimouri Sani F. Changes in the thymus gland with age: A sonographic evaluation. ULTRASOUND (LEEDS, ENGLAND) 2023; 31:204-211. [PMID: 37538966 PMCID: PMC10395379 DOI: 10.1177/1742271x221124484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 07/28/2022] [Indexed: 08/05/2023]
Abstract
Background Ultrasound evaluation of normal, ectopic, asymmetric, and hyperplastic thymus and also its differentiation from abnormalities are challenging in children, and few studies have addressed this issue. This study aimed to investigate the thymus sonographic changes with age. Methods In this cross-sectional study, 118 healthy children were categorised into six age groups. Sonographic features of the thymus, including volume, anatomical position, symmetry, and echo-texture, were recorded. Results The thymus was visible at all ages from the suprasternal view. In 77.5% of participants, the thymus gland volume in lobes was symmetrical; however, left (21.2%) and right (1.3%) predominance were also found. The most common position of the thymus was in front of the great vessels (100%) with suprasternal extension (97.5%). The mean volume of thymus was 21.3 ± 10.5 (mm). There was no significant difference in the volumes of the thymus between different age groups. The predominant echo-texture of the thymus in different age groups was hypoechoic with thin echogenic septa (liver-like) in below 2-3 years of age, the appearance of echogenic foci and hyperechoic echo-texture (liver-like with starry sky) in 2-14 years, and uniform hyperechoic echo-texture (fatty liver-like) or geographic echo-texture with coarse reticular pattern in above 14 years. Conclusion In children, the thymus gland is visible in ultrasound examination in all age groups from the suprasternal view; however, the echo-texture of the normal thymus changes with age. There was no significant correlation between age and sex with total thymic volume. The specificity of these appearances has made ultrasound a problem-solving modality in children.
Collapse
Affiliation(s)
- Seyed Ali Alamdaran
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoud Mahdavi Rashed
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Yekta
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Teimouri Sani
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Suster D, Ronen N, Pierce DC, Suster S. Thymic Parenchymal Hyperplasia. Mod Pathol 2023; 36:100207. [PMID: 37149223 DOI: 10.1016/j.modpat.2023.100207] [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: 03/22/2023] [Revised: 04/17/2023] [Accepted: 04/25/2023] [Indexed: 05/08/2023]
Abstract
Thymic hyperplasia is a rare condition generally caused by lymphoid follicular hyperplasia associated with autoimmune disorders. True thymic parenchymal hyperplasia unassociated with lymphoid follicular hyperplasia is extremely rare and may give rise to difficulties in diagnosis. We have studied 44 patients with true thymic hyperplasia (38 females and 6 males) aged 7 months to 64 years (mean, 36 years). Eighteen patients presented with symptoms of chest discomfort or shortness of breath; in 20 patients, the lesions were discovered incidentally. Imaging studies demonstrated enlargement of the mediastinum by a mass lesion suspicious for malignancy. All patients were treated with complete surgical excision. The tumors measured from 3.5 to 24 cm (median, 10 cm; mean, 10.46 cm). Histologic examination showed lobules of thymic tissue displaying well-developed corticomedullary architecture, with scattered Hassall corpuscles separated by mature adipose tissue and bounded by a thin fibrous capsule. No cases showed evidence of lymphoid follicular hyperplasia, cytologic atypia, or confluence of the lobules. Immunohistochemical studies showed a normal pattern of distribution for keratin-positive thymic epithelial cells against a background rich in CD3/TdT/CD1a+ lymphocytes. Twenty-nine cases had an initial clinical or pathological diagnosis of thymoma or thymoma vs thymic hyperplasia. Clinical follow-up in 26 cases showed that all patients were alive and well between 5 and 15 years after diagnosis (mean, 9 years). Thymic parenchymal hyperplasia causing significant enlargement of the normal thymus that is sufficient to cause symptoms or worrisome imaging findings should be considered in the differential diagnosis of anterior mediastinal masses. The criteria for distinguishing such lesions from lymphocyte-rich thymoma are presented.
Collapse
Affiliation(s)
- David Suster
- Department of Pathology, Rutgers New Jersey Medical School, Newark, New Jersey.
| | - Natali Ronen
- Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Douglas C Pierce
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Saul Suster
- Department of Pathology, Rutgers New Jersey Medical School, Newark, New Jersey; Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
5
|
Baliu-Piqué M, Tesselaar K, Borghans JAM. Are homeostatic mechanisms aiding the reconstitution of the T-cell pool during lymphopenia in humans? Front Immunol 2022; 13:1059481. [PMID: 36483556 PMCID: PMC9723355 DOI: 10.3389/fimmu.2022.1059481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/02/2022] [Indexed: 11/23/2022] Open
Abstract
A timely recovery of T-cell numbers following haematopoietic stem-cell transplantation (HSCT) is essential for preventing complications, such as increased risk of infection and disease relapse. In analogy to the occurrence of lymphopenia-induced proliferation in mice, T-cell dynamics in humans are thought to be homeostatically regulated in a cell density-dependent manner. The idea is that T cells divide faster and/or live longer when T-cell numbers are low, thereby helping the reconstitution of the T-cell pool. T-cell reconstitution after HSCT is, however, known to occur notoriously slowly. In fact, the evidence for the existence of homeostatic mechanisms in humans is quite ambiguous, since lymphopenia is often associated with infectious complications and immune activation, which confound the study of homeostatic regulation. This calls into question whether homeostatic mechanisms aid the reconstitution of the T-cell pool during lymphopenia in humans. Here we review the changes in T-cell dynamics in different situations of T-cell deficiency in humans, including the early development of the immune system after birth, healthy ageing, HIV infection, thymectomy and hematopoietic stem cell transplantation (HSCT). We discuss to what extent these changes in T-cell dynamics are a side-effect of increased immune activation during lymphopenia, and to what extent they truly reflect homeostatic mechanisms.
Collapse
Affiliation(s)
| | | | - José A. M. Borghans
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
6
|
Kinetics of anti-nucleocapsid IgG response in COVID-19 immunocompetent convalescent patients. Sci Rep 2022; 12:12403. [PMID: 35859108 PMCID: PMC9297274 DOI: 10.1038/s41598-022-16402-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 07/11/2022] [Indexed: 01/02/2023] Open
Abstract
The comprehension of a long-term humoral immune response against SARS-CoV-2 can shed light on the treatment and vaccination strategies of COVID-19 disease, improving the knowledge about this virus infection and/or re-infection. We assessed the IgG antibodies against SARS-CoV-2 nucleocapsid (N) protein (anti-SARS-CoV-2 (N) IgG) in 1441 COVID-19 convalescent patients within 15 months longitudinal study from middle-developed country. The main inclusion criteria was positive RT– PCR result on nasopharyngeal swab samples at least one month before antibody testing and absence of any induced or inherited immunodeficiency. 92.7% of convalescent patients’ serum contained anti-SARS-CoV-2 (N) IgG and only 1.3% of patients had a delayed antibody response. In the majority of convalescent patients’ the durability of antibodies lasted more than one year. The kinetics of anti-SARS-CoV-2 (N) IgG took a bell-shaped character—increased first 25–30 weeks, then started to decrease, but were still detectable for more than 15 months. We found that on the one hand anti-SARS-CoV-2 humoral response level correlates with disease severity, on the other, in particular, the level of peak antibodies correlates with age—older patients develop more robust humoral response regardless of sex, disease severity and BMI.
Collapse
|
7
|
Andreu-Sánchez S, Aubert G, Ripoll-Cladellas A, Henkelman S, Zhernakova DV, Sinha T, Kurilshikov A, Cenit MC, Jan Bonder M, Franke L, Wijmenga C, Fu J, van der Wijst MGP, Melé M, Lansdorp P, Zhernakova A. Genetic, parental and lifestyle factors influence telomere length. Commun Biol 2022; 5:565. [PMID: 35681050 PMCID: PMC9184499 DOI: 10.1038/s42003-022-03521-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/22/2022] [Indexed: 11/09/2022] Open
Abstract
The average length of telomere repeats (TL) declines with age and is considered to be a marker of biological ageing. Here, we measured TL in six blood cell types from 1046 individuals using the clinically validated Flow-FISH method. We identified remarkable cell-type-specific variations in TL. Host genetics, environmental, parental and intrinsic factors such as sex, parental age, and smoking are associated to variations in TL. By analysing the genome-wide methylation patterns, we identified that the association of maternal, but not paternal, age to TL is mediated by epigenetics. Single-cell RNA-sequencing data for 62 participants revealed differential gene expression in T-cells. Genes negatively associated with TL were enriched for pathways related to translation and nonsense-mediated decay. Altogether, this study addresses cell-type-specific differences in telomere biology and its relation to cell-type-specific gene expression and highlights how perinatal factors play a role in determining TL, on top of genetics and lifestyle.
Collapse
Affiliation(s)
- Sergio Andreu-Sánchez
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Geraldine Aubert
- Terry Fox Laboratory, British Columbia Cancer Research Center, Vancouver, BC, Canada
- Repeat Diagnostics Inc, Vancouver, BC, Canada
| | - Aida Ripoll-Cladellas
- Life Sciences Department, Barcelona Supercomputing Center, 08034, Barcelona, Catalonia, Spain
| | - Sandra Henkelman
- European Research Institute for the Biology of Ageing, University of Groningen, Groningen, the Netherlands
| | - Daria V Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Laboratory of Genomic Diversity, Center for Computer Technologies, ITMO University, St. Petersburg, 197101, Russia
| | - Trishla Sinha
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Maria Carmen Cenit
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Microbial Ecology, Nutrition, and Health Research Unit, Institute of Agrochemistry and Food Technology (IATA-CSIC), 46980, Paterna-Valencia, Spain
| | - Marc Jan Bonder
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, 69117, Heidelberg, Germany
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Monique G P van der Wijst
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marta Melé
- Life Sciences Department, Barcelona Supercomputing Center, 08034, Barcelona, Catalonia, Spain
| | - Peter Lansdorp
- Terry Fox Laboratory, British Columbia Cancer Research Center, Vancouver, BC, Canada.
- European Research Institute for the Biology of Ageing, University of Groningen, Groningen, the Netherlands.
- Departments of Hematology and Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
8
|
Crews DW, Dombroski JA, King MR. Prophylactic Cancer Vaccines Engineered to Elicit Specific Adaptive Immune Response. Front Oncol 2021; 11:626463. [PMID: 33869008 PMCID: PMC8044825 DOI: 10.3389/fonc.2021.626463] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Vaccines have been used to prevent and eradicate different diseases for over 200 years, and new vaccine technologies have the potential to prevent many common illnesses. Cancer, despite many advances in therapeutics, is still the second leading causes of death in the United States. Prophylactic, or preventative, cancer vaccines have the potential to reduce cancer prevalence by initiating a specific immune response that will target cancer before it can develop. Cancer vaccines can include many different components, such as peptides and carbohydrates, and be fabricated for delivery using a variety of means including through incorporation of stabilizing chemicals like polyethylene glycol (PEG) and pan-DR helper T-lymphocyte epitope (PADRE), fusion with antigen-presenting cells (APCs), microneedle patches, and liposomal encapsulation. There are currently five cancer vaccines used in the clinic, protecting against either human papillomavirus (HPV) or hepatitis B virus (HBV), and preventing several different types of cancer including cervical and oral cancer. Prophylactic cancer vaccines can promote three different types of adaptive responses: humoral (B cell, or antibody-mediated), cellular (T cell) or a combination of the two types. Each vaccine has its advantages and challenges at eliciting an adaptive immune response, but these prophylactic cancer vaccines in development have the potential to prevent or delay tumor development, and reduce the incidence of many common cancers.
Collapse
Affiliation(s)
- Davis W Crews
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | - Jenna A Dombroski
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
9
|
Saghafian-Hedengren S, Sverremark-Ekström E, Nilsson A. T Cell Subsets During Early Life and Their Implication in the Treatment of Childhood Acute Lymphoblastic Leukemia. Front Immunol 2021; 12:582539. [PMID: 33763058 PMCID: PMC7982872 DOI: 10.3389/fimmu.2021.582539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 02/01/2021] [Indexed: 11/13/2022] Open
Abstract
The immune system plays a major role in recognizing and eliminating malignant cells, and this has been exploited in the development of immunotherapies aimed at either activating or reactivating the anti-tumor activity of a patient's immune system. A wide range of therapeutic approaches involving T lymphocytes, such as programmed cell death protein ligand-1 (PDL-1) inhibitors, cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) blockers, and CD19-targeted T-cell therapy through chimeric antigen receptor (CAR)-T cells or CD19/CD3 bi-specific T-cell engagers, have been introduced to the field of oncology, leading to significant improvements in overall survival of adult cancer patients. During the past few years, the availability and approval of T-cell based immunotherapies have become a reality also for the treatment of childhood cancers. However, the distribution, ratio of regulatory to effector cells and the quality of T-cell responses early in life are distinct from those during adolescence and adulthood, raising the possibility that these differences impact the efficacy of immunotherapy. Herein we provide a brief overview of the properties of conventional T cell subsets during early life. Focusing on the most common cancer type during childhood, acute lymphoblastic leukemia (ALL), we describe how current conventional therapies used against ALL influence the T-cell compartment of small children. We describe early life T-cell responses in relation to immunotherapies engaging T-cell anticancer reactivity and present our opinion that it is not only immaturity of the adaptive immune system, but also the impact of an immunosuppressive environment that may prove disadvantageous in the setting of immunotherapies targeting pediatric cancer cells.
Collapse
Affiliation(s)
- Shanie Saghafian-Hedengren
- Division of Paediatric Oncology and Paediatric Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Anna Nilsson
- Division of Paediatric Oncology and Paediatric Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Albarrán-Tamayo F, Murillo-Ortiz B, González Amaro R, López Briones S. Both in vitro T cell proliferation and telomere length are decreased, but CD25 expression and IL-2 production are not affected in aged men. Arch Med Sci 2021; 17:775-784. [PMID: 34025848 PMCID: PMC8130486 DOI: 10.5114/aoms.2019.87593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 06/03/2018] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Aging is a natural process involving dysfunction of multiple organs and is characterized by increased susceptibility to infections, cancer and autoimmune diseases. The functionality of the immune system depends on the capacity of lymphocytes to proliferate in response to antigenic challenges, and telomere length has an important role regulating the number of cell divisions. The aim of this study was to determine the possible relationship between telomere length, interleukin 2 (IL-2) production, CD25 expression and proliferation of peripheral blood mononuclear cells (PBMCs) in aged men. MATERIAL AND METHODS Telomere length was measured by RT-PCR in PBMCs from young and aged men. IL-2 production and CD25 expression were determined by ELISA and flow cytometry, respectively. Cell proliferation was measured by CFSE dilution assays upon in vitro stimulation with concanavalin A (Con A). RESULTS PBMCs from aged men showed a shorter telomere length and a reduced capacity to proliferate in vitro, compared to young men. In contrast, no significant differences in the level of CD25 expression on T lymphocytes, and in vitro production of IL-2 were detected in both groups. In addition, no significant correlation was detected between levels of CD25 expression, IL-2 production, cell proliferation, and telomere length in aged men. CONCLUSIONS In aged men the telomere length shortening and the reduced T cell proliferation are not related to the capacity of IL-2 production and CD25 expression on T lymphocytes.
Collapse
Affiliation(s)
| | - Blanca Murillo-Ortiz
- Unidad de Investigación en Epidemiología Clínica, Unidad Médica de Alta Especialidad (UMAE) No. 1 Bajío, Instituto Mexicano del Seguro Social (IMSS), León, Guanajuato, México
| | - Roberto González Amaro
- Departamento de Inmunología, Escuela de Medicina, Universidad Autónoma de San Luís Potosí, San Luís Potosí, México
| | - Sergio López Briones
- Departamento de Medicina y Nutrición, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato, México
| |
Collapse
|
11
|
Boda F, Banfai K, Garai K, Kovacs B, Almasi A, Scheffer D, Sinkler RL, Csonka R, Czompoly T, Kvell K. Effect of Bitis gabonica and Dendroaspis angusticeps snake venoms on apoptosis-related genes in human thymic epithelial cells. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20200057. [PMID: 33402885 PMCID: PMC7745260 DOI: 10.1590/1678-9199-jvatitd-2020-0057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background: Certain environmental toxins permanently damage the thymic epithelium, accelerate immune senescence and trigger secondary immune pathologies. However, the exact underlying cellular mechanisms and pathways of permanent immune intoxication remain unknown. The aim of the present study was to demonstrate gene expressional changes of apoptosis-related cellular pathways in human thymic epithelial cells following exposure to snake venom from Bitis gabonica and Dendroaspis angusticeps. Methods: Snake venoms were characterized by analytical methods including reversed phase high-performance liquid chromatography and sodium dodecyl sulphate-polyacrylamide gel electrophoresis, then applied on human thymic epithelial cells (1889c) for 24 h at 10 μg/mL (as used in previous TaqMan Array study). Gene expressional changes restricted to apoptosis were assayed by TaqMan Array (Human Apoptosis Plate). Results: The most prominent gene expressional changes were shown by CASP5 (≈ 2.5 million-fold, confirmed by dedicated quantitative polymerase chain reaction) and CARD9 (0.016-fold) for B. gabonica, and BIRC7 (6.46-fold) and CASP1 (0.30-fold) for D. angusticeps. Conclusion: The observed apoptotic environment suggests that pyroptosis may be the dominant pathway through which B. gabonica and D. angusticeps snake venoms trigger thymic epithelial apoptosis following envenomation.
Collapse
Affiliation(s)
- Francisc Boda
- Department F1, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania
| | - Krisztina Banfai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, Pecs, Hungary.,Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary
| | - Kitti Garai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, Pecs, Hungary.,Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary
| | - Bela Kovacs
- Department F1, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania
| | - Attila Almasi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Pecs, Pecs, Hungary
| | - Dalma Scheffer
- Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary.,Soft Flow Ltd., Pecs, Hungary
| | - Reka Lambertne Sinkler
- Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary.,Soft Flow Ltd., Pecs, Hungary
| | - Robert Csonka
- Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary.,Soft Flow Ltd., Pecs, Hungary
| | - Tamas Czompoly
- Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary.,Soft Flow Ltd., Pecs, Hungary
| | - Krisztian Kvell
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, Pecs, Hungary.,Food Biotechnology Research Group, Szentagothai Research Center, University of Pecs, Pecs, Hungary
| |
Collapse
|
12
|
de Barros SC, Suterwala BT, He C, Ge S, Chick B, Blumberg GK, Kim K, Klein S, Zhu Y, Wang X, Casero D, Crooks GM. Pleiotropic Roles of VEGF in the Microenvironment of the Developing Thymus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:2423-2436. [PMID: 32989093 PMCID: PMC7679052 DOI: 10.4049/jimmunol.1901519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 08/27/2020] [Indexed: 01/04/2023]
Abstract
Neonatal life marks the apogee of murine thymic growth. Over the first few days after birth, growth slows and the murine thymus switches from fetal to adult morphology and function; little is known about the cues driving this dramatic transition. In this study, we show for the first time (to our knowledge) the critical role of vascular endothelial growth factor (VEGF) on thymic morphogenesis beyond its well-known role in angiogenesis. During a brief window a few days after birth, VEGF inhibition induced rapid and profound remodeling of the endothelial, mesenchymal and epithelial thymic stromal compartments, mimicking changes seen during early adult maturation. Rapid transcriptional changes were seen in each compartment after VEGF inhibition, including genes involved in migration, chemotaxis, and cell adhesion as well as induction of a proinflammatory and proadipogenic signature in endothelium, pericytes, and mesenchyme. Thymocyte numbers fell subsequent to the stromal changes. Expression patterns and functional blockade of the receptors VEGFR2 and NRP1 demonstrated that VEGF mediates its pleiotropic effects through distinct receptors on each microenvironmental compartment of the developing mouse thymus.
Collapse
Affiliation(s)
- Stephanie C de Barros
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Batul T Suterwala
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Chongbin He
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Shundi Ge
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Brent Chick
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Garrett K Blumberg
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Kenneth Kim
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Sam Klein
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Yuhua Zhu
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Xiaoyan Wang
- Department of General Internal Medicine and Health Services Research, University of California Los Angeles, Los Angeles, CA 90095
| | - David Casero
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Gay M Crooks
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095;
- Department of Pediatrics, University of California Los Angeles, Los Angeles, CA 90095; and
- Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
13
|
Yu Y, Singh H, Kwon K, Tsitrin T, Petrini J, Nelson KE, Pieper R. Protein signatures from blood plasma and urine suggest changes in vascular function and IL-12 signaling in elderly with a history of chronic diseases compared with an age-matched healthy cohort. GeroScience 2020; 43:593-606. [PMID: 32974878 PMCID: PMC8110643 DOI: 10.1007/s11357-020-00269-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/14/2020] [Indexed: 01/02/2023] Open
Abstract
Key processes characterizing human aging are immunosenescence and inflammaging. The capacity of the immune system to adequately respond to external perturbations (e.g., pathogens, injuries, and biochemical irritants) and to repair somatic mutations that may cause cancers or cellular senescence declines. An important goal remains to identify genetic or biochemical, predictive biomarkers for healthy aging. We recruited two cohorts in the age range 70 to 82, one afflicted by chronic illnesses (non-healthy aging, NHA) and the other in good health (healthy aging, HA). NHA criteria included major cardiovascular, neurodegenerative, and chronic pulmonary diseases, diabetes, and cancers. Quantitative analysis of forty proinflammatory cytokines in blood plasma and more than 500 proteins in urine was performed to identify candidate biomarkers for and biological pathway implications of healthy aging. Nine cytokines revealed lower quantities in blood plasma for the NHA compared with the HA groups (fold change > 1.5; p value < 0.025) including IL-12p40 and IL-12p70. We note that, sampling at two timepoints, intra-individual cytokine abundance patterns clustered in 86% of all 60 cases, indicative of person-specific, highly controlled multi-cytokine signatures in blood plasma. Twenty-three urinary proteins were differentially abundant (HA versus NHA; fold change > 1.5; p value < 0.01). Among the proteins increased in abundance in the HA cohort were glycoprotein MUC18, ephrin type-B receptor 4, matrix remodeling-associated protein 8, angiopoietin-related protein 2, K-cadherin, and plasma protease C1 inhibitor. These proteins have been linked to the extracellular matrix, cell adhesion, and vascular remodeling and repair processes. In silico network analysis identified the regulation of coagulation, antimicrobial humoral immune responses, and the IL-12 signaling pathway as enriched GO terms. To validate links of these preliminary biomarkers and IL-12 signaling with healthy aging, clinical studies using larger cohorts and functional characterization of the genes/proteins in cellular models of aging need to be conducted.
Collapse
Affiliation(s)
- Yanbao Yu
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Harinder Singh
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Keehwan Kwon
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Tamara Tsitrin
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA
| | - Joann Petrini
- Western Connecticut Health Network, 24 Hospital Avenue, Danbury, CT, 06810, USA
| | - Karen E Nelson
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA.,J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA
| | - Rembert Pieper
- J. Craig Venter Institute, 9605 Medical Center Drive, Rockville, MD, 20850, USA.
| |
Collapse
|
14
|
Age-related transcriptional modules and TF-miRNA-mRNA interactions in neonatal and infant human thymus. PLoS One 2020; 15:e0227547. [PMID: 32294112 PMCID: PMC7159188 DOI: 10.1371/journal.pone.0227547] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/18/2019] [Indexed: 12/15/2022] Open
Abstract
The human thymus suffers a transient neonatal involution, recovers and then starts a process of decline between the 1st and 2nd years of life. Age-related morphological changes in thymus were extensively investigated, but the genomic mechanisms underlying this process remain largely unknown. Through Weighted Gene Co-expression Network Analysis (WGCNA) and TF-miRNA-mRNA integrative analysis we studied the transcriptome of neonate and infant thymic tissues grouped by age: 0–30 days (A); 31days-6 months (B); 7–12 months (C); 13–18 months (D); 19-31months (E). Age-related transcriptional modules, hubs and high gene significance (HGS) genes were identified, as well as TF-miRNA-hub/HGS co-expression correlations. Three transcriptional modules were correlated with A and/or E groups. Hubs were mostly related to cellular/metabolic processes; few were differentially expressed (DE) or related to T-cell development. Inversely, HGS genes in groups A and E were mostly DE. In A (neonate) one third of the hyper-expressed HGS genes were related to T-cell development, against one-twentieth in E, what may correlate with the early neonatal depletion and recovery of thymic T-cell populations. This genomic mechanism is tightly regulated by TF-miRNA-hub/HGS interactions that differentially govern cellular and molecular processes involved in the functioning of the neonate thymus and in the beginning of thymic decline.
Collapse
|
15
|
Han J, Mu W, Zhao H, Hao Y, Song C, Zhou H, Sun X, Li G, Dai G, Zhang Y, Zhang F, Zeng H. HIV-1 low-level viremia affects T cell activation rather than T cell development in school-age children, adolescents, and young adults during antiretroviral therapy. Int J Infect Dis 2019; 91:210-217. [PMID: 31821891 DOI: 10.1016/j.ijid.2019.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/18/2019] [Accepted: 12/01/2019] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES Given the improvements in antiretroviral therapy (ART) in recent years, more pediatric HIV patients receiving ART are reaching adolescence and adulthood. This study investigated the influence of poor virological response (low-level viremia (LLV) and virological failure (VF)) on the immune system of these patients. METHODS HIV-infected, ART-experienced pediatric patients (n=206) were enrolled in this cross-sectional study. The patients were subdivided into school-age children/early adolescents, middle adolescents, and late adolescents/young adults according to their age, and further classified into virological suppression (VS), LLV, and VF groups according to plasma viral load (pVL) measurement. Thymic output, T cells subsets, and immune activation were analyzed by flow cytometry. RESULTS Compared with VS patients, VF patients displayed decreased CD4+ T cell counts, while LLV and VS patients had comparable CD4+ T cell counts regardless of age. Compared with VS patients, LLV and VF patients had higher percentages of CD8+HLA-DR+ and CD8+CD38high T cells, and the immune activation was positively correlated with pVL in VF and LLV patients. Thymic output levels (CD31+) and regulatory T cell subpopulations in LLV and VF patients were comparable to those in VS patients. LLV patients showed comparable percentages of T cell subsets (TN, TCM, TEMRA, and TEM) as VS patients in all age groups. CONCLUSIONS LLV causes excessive immune activation although it does not impair T cell recovery or naïve-to-memory T cell conversion in pediatric patients living with HIV. Therefore, T cell immune activation should be monitored at the management of LLV during ART.
Collapse
Affiliation(s)
- Junyan Han
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Weiwei Mu
- Division of Treatment and Care, National Center for AIDS/STD Control and Prevention, Beijing 102206, China
| | - Hongxin Zhao
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yu Hao
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Chuan Song
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Haiwei Zhou
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Xin Sun
- Division of Treatment and Care, National Center for AIDS/STD Control and Prevention, Beijing 102206, China
| | - Guoli Li
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Guorui Dai
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yu Zhang
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Fujie Zhang
- Division of Treatment and Care, National Center for AIDS/STD Control and Prevention, Beijing 102206, China; Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China.
| | - Hui Zeng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China.
| |
Collapse
|
16
|
Baliu-Piqué M, Kurniawan H, Ravesloot L, Verheij MW, Drylewicz J, Lievaart-Peterson K, Borghans JAM, Koets A, Tesselaar K. Age-related distribution and dynamics of T-cells in blood and lymphoid tissues of goats. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 93:1-10. [PMID: 30550777 DOI: 10.1016/j.dci.2018.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/10/2018] [Accepted: 12/10/2018] [Indexed: 06/09/2023]
Abstract
Neonatal mammals have increased disease susceptibility and sub-optimal vaccine responses. This raises problems in both humans and farm animals. The high prevalence of paratuberculosis in goats and the lack of an effective vaccine against it have a strong impact on the dairy sector, and calls for vaccines optimized for the neonatal immune system. We characterized the composition of the T-cell pool in neonatal kids and adult goats and quantified their turnover rates using in vivo deuterium labelling. From birth to adulthood, CD4+ T-cells were the predominant subset in the thymus and lymph nodes, while spleen and bone marrow contained mainly CD8+ lymphocytes. In blood, CD4+ T-cells were the predominant subset during the neonatal period, while CD8+ T-cells predominated in adults. We observed that thymic mass and cellularity increased during the first 5 months after birth, but decreased later in life. Deuterium labelling revealed that T-cell turnover rates in neonatal kids are considerably higher than in adult animals.
Collapse
Affiliation(s)
- Mariona Baliu-Piqué
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Henry Kurniawan
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Lars Ravesloot
- Department of Bacteriology and Epidemiology, Wageningen Bioveterinary Research, Lelystad, the Netherlands; Department of Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Myrddin W Verheij
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Julia Drylewicz
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - José A M Borghans
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ad Koets
- Department of Bacteriology and Epidemiology, Wageningen Bioveterinary Research, Lelystad, the Netherlands; Department of Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Kiki Tesselaar
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
17
|
Banfai K, Ernszt D, Pap A, Bai P, Garai K, Belharazem D, Pongracz JE, Kvell K. "Beige" Cross Talk Between the Immune System and Metabolism. Front Endocrinol (Lausanne) 2019; 10:369. [PMID: 31275241 PMCID: PMC6591453 DOI: 10.3389/fendo.2019.00369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/24/2019] [Indexed: 12/25/2022] Open
Abstract
With thymic senescence the epithelial network shrinks to be replaced by adipose tissue. Transcription factor TBX-1 controls thymus organogenesis, however, the same TBX-1 has also been reported to orchestrate beige adipose tissue development. Given these different roles of TBX-1, we have assessed if thymic TBX-1 expression persists and demonstrates this dualism during adulthood. We have also checked whether thymic adipose involution could yield beige adipose tissue. We have used adult mouse and human thymus tissue from various ages to evaluate the kinetics of TBX-1 expression, as well as mouse (TEP1) and human (1889c) thymic epithelial cells (TECs) for our studies. Electron micrographs show multi-locular lipid deposits typical of beige adipose cells. Histology staining shows the accumulation of neutral lipid deposits. qPCR measurements show persistent and/or elevating levels of beige-specific and beige-indicative markers (TBX-1, EAR-2, UCP-1, PPAR-gamma). We have performed miRNome profiling using qPCR-based QuantStudio platform and amplification-free NanoString platform. We have observed characteristic alterations, including increased miR21 level (promoting adipose tissue development) and decreased miR34a level (bias toward beige adipose tissue differentiation). Finally, using the Seahorse metabolic platform we have recorded a metabolic profile (OCR/ECAR ratio) indicative of beige adipose tissue. In summary, our results support that thymic adipose tissue emerging with senescence is bona fide beige adipose tissue. Our data show how the borders blur between a key immune tissue (the thymus) and a key metabolic tissue (beige adipose tissue) with senescence. Our work contributes to the understanding of cross talk between the immune system and metabolism.
Collapse
Affiliation(s)
- Krisztina Banfai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
| | - David Ernszt
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
- Department of Physiology, Medical School, University of Pécs, Pécs, Hungary
| | - Attila Pap
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Bai
- Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, Debrecen, Hungary
- MTA-DE Lendulet Laboratory of Cellular Metabolism, Debrecen, Hungary
- Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Kitti Garai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
| | - Djeda Belharazem
- Department of Pathology, University Hospital of Mannheim, Mannheim, Germany
| | - Judit E. Pongracz
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
| | - Krisztian Kvell
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
- Szentagothai Research Center, University of Pécs, Pécs, Hungary
- *Correspondence: Krisztian Kvell
| |
Collapse
|
18
|
Moreira-Filho CA, Bando SY, Bertonha FB, Ferreira LR, Vinhas CDF, Oliveira LHB, Zerbini MCN, Furlanetto G, Chaccur P, Carneiro-Sampaio M. Minipuberty and Sexual Dimorphism in the Infant Human Thymus. Sci Rep 2018; 8:13169. [PMID: 30177771 PMCID: PMC6120939 DOI: 10.1038/s41598-018-31583-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022] Open
Abstract
AIRE expression in thymus is downregulated by estrogen after puberty, what probably renders women more susceptible to autoimmune disorders. Here we investigated the effects of minipuberty on male and female infant human thymic tissue in order to verify if this initial transient increase in sex hormones - along the first six months of life - could affect thymic transcriptional network regulation and AIRE expression. Gene co-expression network analysis for differentially expressed genes and miRNA-target analysis revealed sex differences in thymic tissue during minipuberty, but such differences were not detected in the thymic tissue of infants aged 7-18 months, i.e. the non-puberty group. AIRE expression was essentially the same in both sexes in minipuberty and in non-puberty groups, as assessed by genomic and immunohistochemical assays. However, AIRE-interactors networks showed several differences in all groups regarding gene-gene expression correlation. Therefore, minipuberty and genomic mechanisms interact in shaping thymic sexual dimorphism along the first six months of life.
Collapse
Affiliation(s)
| | - Silvia Yumi Bando
- Departament of Pediatrics, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | | | | | | | | | - Paulo Chaccur
- Instituto Dante Pazzanese de Cardiologia, São Paulo, SP, Brazil
| | - Magda Carneiro-Sampaio
- Departament of Pediatrics, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
19
|
Sadighi Akha AA. Aging and the immune system: An overview. J Immunol Methods 2018; 463:21-26. [PMID: 30114401 DOI: 10.1016/j.jim.2018.08.005] [Citation(s) in RCA: 232] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 07/17/2018] [Accepted: 08/08/2018] [Indexed: 12/13/2022]
Abstract
The world is witnessing a rapid demographic shift towards an older population, a trend with major medical, social, economic and political implications. Aging is a multifaceted process, involving numerous molecular and cellular mechanisms in the context of different organ systems. A crucial component of aging is a set of functional and structural alterations in the immune system that can manifest as a decreased ability to fight infection, diminished response to vaccination, increased incidence of cancer, higher prevalence of autoimmunity and constitutive low-grade inflammation, among others. In addition to cell-intrinsic changes in both innate and adaptive immune cells, alterations in the stromal microenvironment in primary and secondary lymphoid organs play an important role in age-associated immune dysfunction. This article will provide a broad overview of these phenomena and point out some of their clinical and therapeutic implications.
Collapse
Affiliation(s)
- Amir A Sadighi Akha
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, United States.
| |
Collapse
|
20
|
Iacobazzi D, Swim MM, Albertario A, Caputo M, Ghorbel MT. Thymus-Derived Mesenchymal Stem Cells for Tissue Engineering Clinical-Grade Cardiovascular Grafts. Tissue Eng Part A 2018; 24:794-808. [DOI: 10.1089/ten.tea.2017.0290] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Dominga Iacobazzi
- Bristol Medical School, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| | - Megan M. Swim
- Bristol Medical School, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| | - Ambra Albertario
- Bristol Medical School, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| | - Massimo Caputo
- Bristol Medical School, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| | - Mohamed T. Ghorbel
- Bristol Medical School, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
21
|
Fishbein KW, Makrogiannis SK, Lukas VA, Okine M, Ramachandran R, Ferrucci L, Egan JM, Chia CW, Spencer RG. Measurement of fat fraction in the human thymus by localized NMR and three-point Dixon MRI techniques. Magn Reson Imaging 2018; 50:110-118. [PMID: 29605590 DOI: 10.1016/j.mri.2018.03.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 03/27/2018] [Accepted: 03/29/2018] [Indexed: 11/28/2022]
Abstract
PURPOSE To develop a protocol to non-invasively measure and map fat fraction, fat/(fat+water), as a function of age in the adult thymus for future studies monitoring the effects of interventions aimed at promoting thymic rejuvenation and preservation of immunity in older adults. MATERIALS AND METHODS Three-dimensional spoiled gradient echo 3T MRI with 3-point Dixon fat-water separation was performed at full inspiration for thymus conspicuity in 36 volunteers 19 to 56 years old. Reproducible breath-holding was facilitated by real-time pressure recording external to the console. The MRI method was validated against localized spectroscopy in vivo, with ECG triggering to compensate for stretching during the cardiac cycle. Fat fractions were corrected for T1 and T2 bias using relaxation times measured using inversion recovery-prepared PRESS with incremented echo time. RESULTS In thymus at 3 T, T1water = 978 ± 75 ms, T1fat = 323 ± 37 ms, T2water = 43.4 ± 9.7 ms and T2fat = 52.1 ± 7.6 ms were measured. Mean T1-corrected MRI fat fractions varied from 0.2 to 0.8 and were positively correlated with age, weight and body mass index (BMI). In subjects with matching MRI and MRS fat fraction measurements, the difference between these measurements exhibited a mean of -0.008 with a 95% confidence interval of (0.123, -0.138). CONCLUSIONS 3-point Dixon MRI of the thymus with T1 bias correction produces quantitative fat fraction maps that correlate with T2-corrected MRS measurements and show age trends consistent with thymic involution.
Collapse
Affiliation(s)
- Kenneth W Fishbein
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD 21224, USA.
| | - Sokratis K Makrogiannis
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Vanessa A Lukas
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Marilyn Okine
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Ramona Ramachandran
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Luigi Ferrucci
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Josephine M Egan
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Chee W Chia
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Richard G Spencer
- Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD 21224, USA
| |
Collapse
|
22
|
Zhao T, Wu J, Liu X, Zhang L, Chen G, Lu H. Diagnosis of thymic epithelial tumor subtypes by a quantitative proteomic approach. Analyst 2018; 143:2491-2500. [PMID: 29645059 DOI: 10.1039/c8an00218e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study shows the first depth proteomic profiling of all TET subtypes and six candidate biomarkers were identified and validated.
Collapse
Affiliation(s)
- Ting Zhao
- Institutes of Biomedical Sciences and Department of Chemistry
- Fudan University
- Shanghai 200032
- P.R. China
| | - Jie Wu
- Department of Pathology
- Zhongshan Hospital
- Fudan University
- Shanghai 200032
- P.R. China
| | - Xiaohui Liu
- Institutes of Biomedical Sciences and Department of Chemistry
- Fudan University
- Shanghai 200032
- P.R. China
| | - Lei Zhang
- Institutes of Biomedical Sciences and Department of Chemistry
- Fudan University
- Shanghai 200032
- P.R. China
| | - Gang Chen
- Department of Pathology
- Zhongshan Hospital
- Fudan University
- Shanghai 200032
- P.R. China
| | - Haojie Lu
- Institutes of Biomedical Sciences and Department of Chemistry
- Fudan University
- Shanghai 200032
- P.R. China
- Key Laboratory of Glycoconjugates Research Ministry of Public Health
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW This review summarizes research on the physiological changes that occur with aging and the resulting effects on fracture healing. RECENT FINDINGS Aging affects the inflammatory response during fracture healing through senescence of the immune response and increased systemic pro-inflammatory status. Important cells of the inflammatory response, macrophages, T cells, mesenchymal stem cells, have demonstrated intrinsic age-related changes that could impact fracture healing. Additionally, vascularization and angiogenesis are impaired in fracture healing of the elderly. Finally, osteochondral cells and their progenitors demonstrate decreased activity and quantity within the callus. Age-related changes affect many of the biologic processes involved in fracture healing. However, the contributions of such changes do not fully explain the poorer healing outcomes and increased morbidity reported in elderly patients. Future research should address this gap in understanding in order to provide improved and more directed treatment options for the elderly population.
Collapse
Affiliation(s)
- Dan Clark
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA
- Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital and Trauma Center, 2550 23rd St, Building 9, San Francisco, CA, 94110, USA
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California at San Francisco, 513 Parnassus Ave., Rm. S-619A, San Francisco, CA, 94143, USA
| | - Mary Nakamura
- Department of Medicine, University of California at San Francisco, San Francisco, CA, 94143-0451, USA
- Department of Pathology, VA Medical Center, University of California San Francisco & Pathology Service, San Francisco, CA, 94121, USA
| | - Ted Miclau
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA
- Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital and Trauma Center, 2550 23rd St, Building 9, San Francisco, CA, 94110, USA
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA.
- Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital and Trauma Center, 2550 23rd St, Building 9, San Francisco, CA, 94110, USA.
| |
Collapse
|
24
|
Ernszt D, Banfai K, Kellermayer Z, Pap A, Lord JM, Pongracz JE, Kvell K. PPARgamma Deficiency Counteracts Thymic Senescence. Front Immunol 2017; 8:1515. [PMID: 29163553 PMCID: PMC5681731 DOI: 10.3389/fimmu.2017.01515] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/26/2017] [Indexed: 12/30/2022] Open
Abstract
Thymic senescence contributes to increased incidence of infection, cancer and autoimmunity at senior ages. This process manifests as adipose involution. As with other adipose tissues, thymic adipose involution is also controlled by PPARgamma. This is supported by observations reporting that systemic PPARgamma activation accelerates thymic adipose involution. Therefore, we hypothesized that decreased PPARgamma activity could prevent thymic adipose involution, although it may trigger metabolic adverse effects. We have confirmed that both human and murine thymic sections show marked staining for PPARgamma at senior ages. We have also tested the thymic lobes of PPARgamma haplo-insufficient and null mice. Supporting our working hypothesis both adult PPARgamma haplo-insufficient and null mice show delayed thymic senescence by thymus histology, thymocyte mouse T-cell recombination excision circle qPCR and peripheral blood naive T-cell ratio by flow-cytometry. Delayed senescence showed dose-response with respect to PPARgamma deficiency. Functional immune parameters were also evaluated at senior ages in PPARgamma haplo-insufficient mice (null mice do not reach senior ages due to metabolic adverse affects). As expected, sustained and elevated T-cell production conferred oral tolerance and enhanced vaccination efficiency in senior PPARgamma haplo-insufficient, but not in senior wild-type littermates according to ELISA IgG measurements. Of note, humans also show increased oral intolerance issues and decreased protection by vaccines at senior ages. Moreover, PPARgamma haplo-insufficiency also exists in human known as a rare disease (FPLD3) causing metabolic adverse effects, similar to the mouse. When compared to age- and metabolic disorder-matched other patient samples (FPLD2 not affecting PPARgamma activity), FPLD3 patients showed increased human Trec (hTrec) values by qPCR (within healthy human range) suggesting delayed thymic senescence, in accordance with mouse results and supporting our working hypothesis. In summary, our experiments prove that systemic decrease of PPARgamma activity prevents thymic senescence, albeit with metabolic drawbacks. However, thymic tissue-specific PPARgamma antagonism would likely solve the issue.
Collapse
Affiliation(s)
- David Ernszt
- Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, University of Pecs, Pecs, Hungary.,Szentagothai Research Center, University of Pecs, Pecs, Hungary
| | - Krisztina Banfai
- Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, University of Pecs, Pecs, Hungary.,Szentagothai Research Center, University of Pecs, Pecs, Hungary
| | - Zoltan Kellermayer
- Faculty of Medicine, Department of Immunology and Biotechnology, University of Pecs, Pecs, Hungary
| | - Attila Pap
- Faculty of Medicine, Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Janet M Lord
- College of Medical and Dental Sciences, Institute of Inflammation and Aging, University of Birmingham, Birmingham, United Kingdom
| | - Judit E Pongracz
- Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, University of Pecs, Pecs, Hungary.,Szentagothai Research Center, University of Pecs, Pecs, Hungary
| | - Krisztian Kvell
- Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, University of Pecs, Pecs, Hungary.,Szentagothai Research Center, University of Pecs, Pecs, Hungary
| |
Collapse
|
25
|
Weinberger B, Keller M, Grubeck-Loebenstein B. Long-term maintenance of diphtheria-specific antibodies after booster vaccination is hampered by latent infection with Cytomegalovirus. IMMUNITY & AGEING 2017; 14:16. [PMID: 28670328 PMCID: PMC5485602 DOI: 10.1186/s12979-017-0099-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/19/2017] [Indexed: 11/23/2022]
Abstract
Many currently used vaccines are less immunogenic in the elderly compared to young adults. The impact of latent infection with Cytomegalovirus (CMV) on vaccine-induced antibody responses has been discussed controversially. We have demonstrated that recall responses to diphtheria vaccination are frequently insufficient in elderly persons and that antibody concentrations decline substantially within 5 years. In the current study we show that within a cohort of healthy elderly (n = 87; median age 71 years, range 66–92) antibody responses to a booster vaccination against diphtheria do not differ between CMV-negative and CMV-positive individuals 4 weeks after vaccination.. However, the goal of diphtheria-vaccination is long-term protection and this is achieved by circulating anti-toxin antibodies. Diphtheria-specific antibody concentrations decline faster in CMV-positive compared to CMV-negative older adults leading to an increased proportion of persons without protective antibody concentrations 5 years after booster vaccination and endangering long-term protection. This finding could be relevant for vaccination schedules.
Collapse
Affiliation(s)
- Birgit Weinberger
- Institute for Biomedical Aging Research, Universität Innsbruck, Rennweg 10, A-6020 Innsbruck, Austria
| | - Michael Keller
- Institute for Biomedical Aging Research, Universität Innsbruck, Rennweg 10, A-6020 Innsbruck, Austria
| | | |
Collapse
|
26
|
Guo D, Ye Y, Qi J, Tan X, Zhang Y, Ma Y, Li Y. Age and sex differences in microRNAs expression during the process of thymus aging. Acta Biochim Biophys Sin (Shanghai) 2017; 49:409-419. [PMID: 28369179 DOI: 10.1093/abbs/gmx029] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Indexed: 12/18/2022] Open
Abstract
The gender-biased thymus involution and the importance of microRNAs (miRNAs, miRs) expression in modulating the thymus development have been reported in many studies. However, how males and females differ in so many ways in thymus involution remains unclear. To address this question, we investigated the miRNA expression profiles in both untreated 3- and 12-month-old female and male mice thymuses. The results showed that 7 and 18 miRNAs were defined as the sex- and age-specific miRNAs, respectively. The expression of miR-181c-5p, miR-20b-5p, miR-98b-5p, miR-329-3p, miR-341-5p, and miR-2137 showed significant age-difference in mice thymus by quantitative polymerase chain reaction. High expression levels of miR-2137 were detected in mice thymic epithelial cells and gradually increased during the process of thymus aging. MiR-27b-3p and miR-378a-3p of the female-biased miRNAs were confirmed as the sex- and estrogen-responsive miRNAs in mice thymus in vivo. Their potential target genes and the pathway were identified by the online software. Possible regulation roles of sex- and age-specific miRNA expression during the process of thymus aging were discussed. Our results suggested that these miRNAs may be potential biomarkers for the study of sex- and age-specific thymus aging and involution.
Collapse
Affiliation(s)
- Dongguang Guo
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yaqiong Ye
- Department of Basic Veterinary Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Junjie Qi
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xiaotong Tan
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yuan Zhang
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yongjiang Ma
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yugu Li
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
27
|
Jacqueline C, Bourfia Y, Hbid H, Sorci G, Thomas F, Roche B. Interactions between immune challenges and cancer cells proliferation: timing does matter! EVOLUTION MEDICINE AND PUBLIC HEALTH 2016; 2016:299-311. [PMID: 27535084 PMCID: PMC5046994 DOI: 10.1093/emph/eow025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Indexed: 12/17/2022]
Abstract
The immune system is a key component of malignant cell control and it is also involved in the elimination of pathogens that threaten the host. Despite our body is permanently exposed to a myriad of pathogens, the interference of such infections with the immune responses against cancer has been poorly investigated. Through a mathematical model, we show that the frequency, the duration and the action (positive or negative) of immune challenges may significantly impact tumor proliferation. First, we observe that a long immunosuppressive challenge increases accumulation of cancerous cells only if it occurs 14 years after the beginning of immunosenescence. However, short immune challenges result in an even greater accumulation of cancerous cells for the same total duration of immunosuppression. Finally, we show that short challenges of immune activation could lead to a slightly decrease in cancerous cell accumulation compared to a long one. Our results predict that frequent and acute immune challenges could have a different and in some extent higher impact on cancer risk than persistent ones even they have been much less studied in cancer epidemiology. These results are discussed regarding the existing empirical evidences and we suggest potential novel indirect role of infectious diseases on cancer incidence which should be investigated to improve prevention strategies against cancer.
Collapse
Affiliation(s)
- Camille Jacqueline
- CREEC, 911 Avenue Agropolis, BP 64501, Montpellier, Cedex 5 34394, France MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, Montpellier, Cedex 5 34394, France
| | - Youssef Bourfia
- Laboratoire Jacques-Louis Lions (LJLL), UMR 7598 Université Pierre et Marie Curie (UPMC), Paris 6, Boîte courrier 187, ;Paris, Cedex 05 75252, France Université Cadi Ayyad Laboratoire de Mathématiques et Dynamique de Populations, Cadi Ayyad University, Marrakech, Morocco
| | - Hassan Hbid
- Université Cadi Ayyad Laboratoire de Mathématiques et Dynamique de Populations, Cadi Ayyad University, Marrakech, Morocco International Center for Mathematical and Computational Modeling of Complex Systems (UMI IRD/UPMC UMMISCO), 32 Avenue Henri Varagnat, Bondy Cedex 93143, France
| | - Gabriele Sorci
- BiogéoSciences, CNRS UMR 6282, Université de Bourgogne, 6 Boulevard Gabriel, Dijon 21000, France
| | - Frédéric Thomas
- CREEC, 911 Avenue Agropolis, BP 64501, Montpellier, Cedex 5 34394, France MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, Montpellier, Cedex 5 34394, France
| | - Benjamin Roche
- CREEC, 911 Avenue Agropolis, BP 64501, Montpellier, Cedex 5 34394, France International Center for Mathematical and Computational Modeling of Complex Systems (UMI IRD/UPMC UMMISCO), 32 Avenue Henri Varagnat, Bondy Cedex 93143, France
| |
Collapse
|
28
|
Mostafa M, Vali R, Chan J, Omarkhail Y, Shammas A. Variants and pitfalls on radioiodine scans in pediatric patients with differentiated thyroid carcinoma. Pediatr Radiol 2016; 46:1579-89. [PMID: 27488505 DOI: 10.1007/s00247-016-3655-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 04/28/2016] [Accepted: 06/06/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND Potentially false-positive findings on radioiodine scans in children with differentiated thyroid carcinoma can mimic functioning thyroid tissue and functioning thyroid carcinomatous tissue. Such false-positive findings comprise variants and pitfalls that can vary slightly in children as compared with adults. OBJECTIVE To determine the patterns and frequency of these potential false-positive findings on radioiodine scans in children with differentiated thyroid carcinoma. MATERIALS AND METHODS We reviewed a total of 223 radioiodine scans from 53 pediatric patients (mean age 13.3 years, 37 girls) with differentiated thyroid carcinoma. Focal or regional activity that likely did not represent functioning thyroid tissue or functioning thyroid carcinomatous tissue were categorized as variants or pitfalls. The final diagnosis was confirmed by reviewing the concurrent and follow-up clinical data, correlative ultrasonography, CT scanning, serum thyroglobulin and antithyroglobulin antibody levels. We calculated the frequency of these variants and pitfalls from diagnostic and post-therapy radioiodine scans. RESULTS The most common variant on the radioiodine scans was the thymic activity (24/223, 10.8%) followed by the cardiac activity (8/223, 3.6%). Salivary contamination and star artifact, caused by prominent thyroid remnant, were the most important observed pitfalls. CONCLUSION Variants and pitfalls that mimic functioning thyroid tissue or functioning thyroid carcinomatous tissue on radioiodine scan in children with differentiated thyroid carcinoma are not infrequent, but they decrease in frequency on successive radioiodine scans. Potential false-positive findings can be minimized with proper knowledge of the common variants and pitfalls in children and correlation with clinical, laboratory and imaging data.
Collapse
Affiliation(s)
- Mohamed Mostafa
- Nuclear Medicine Division, Department of Medical Imaging, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.
| | - Reza Vali
- Nuclear Medicine Division, Department of Medical Imaging, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Jeffrey Chan
- Nuclear Medicine Division, Department of Medical Imaging, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Yusuaf Omarkhail
- Nuclear Medicine Division, Department of Medical Imaging, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Amer Shammas
- Nuclear Medicine Division, Department of Medical Imaging, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| |
Collapse
|
29
|
Lee JH, Kim TJ, Kim JW, Yoon JS, Kim HS, Lee KM. The Anti-apoptotic Effect of Ghrelin on Restraint Stress-Induced Thymus Atrophy in Mice. Immune Netw 2016; 16:242-8. [PMID: 27574503 PMCID: PMC5002450 DOI: 10.4110/in.2016.16.4.242] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 07/29/2016] [Accepted: 08/04/2016] [Indexed: 12/22/2022] Open
Abstract
Thymic atrophy is a complication that results from exposure to many environmental stressors, disease treatments, and microbial challenges. Such acute stress-associated thymic loss can have a dramatic impact on the host's ability to replenish the necessary naïve T cell output to reconstitute the peripheral T cell numbers and repertoire to respond to new antigenic challenges. We have previously reported that treatment with the orexigenic hormone ghrelin results in an increase in the number and proliferation of thymocytes after dexamethasone challenge, suggesting a role for ghrelin in restraint stress-induced thymic involution and cell apoptosis and its potential use as a thymostimulatory agent. In an effort to understand how ghrelin suppresses thymic T cell apoptosis, we have examined the various signaling pathways induced by receptor-specific ghrelin stimulation using a restraint stress mouse model. In this model, stress-induced apoptosis in thymocytes was effectively blocked by ghrelin. Western blot analysis demonstrated that ghrelin prevents the cleavage of pro-apoptotic proteins such as Bim, Caspase-3, and PARP. In addition, ghrelin stimulation activates the Akt and Mitogen-activated protein kinases (MAPK) signaling pathways in a time/dose-dependent manner. Moreover, we also revealed the involvement of the FoxO3a pathway in the phosphorylation of Akt and ERK1/2. Together, these findings suggest that ghrelin inhibits apoptosis by modulating the stress-induced apoptotic signal pathway in the restraint-induced thymic apoptosis.
Collapse
Affiliation(s)
- Jun Ho Lee
- Global Research Laboratory, Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea
| | - Tae-Jin Kim
- Global Research Laboratory, Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea
| | - Jie Wan Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13524, Korea
| | - Jeong Seon Yoon
- Department of Anatomy, School of Medicine, Ajou University, Suwon 16499, Korea
| | - Hyuk Soon Kim
- Department of Immunology, School of Medicine, Konkuk University, Chungju 27478, Korea
| | - Kyung-Mi Lee
- Global Research Laboratory, Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea
| |
Collapse
|
30
|
Mehta S. Assessment of thymus size in healthy term neonates using sonography - A study from tertiary care hospital of Central India. J Neonatal Perinatal Med 2015; 8:143-7. [PMID: 26410439 DOI: 10.3233/npm-15814067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE A cross-sectional study was done to assess the size of thymus in term healthy infants using sonography and to assess its variation in size, shape, location, echogenicity and echotexture for both male and female reference population. MATERIAL AND METHODS A total of 250 term healthy neonates of either sex were subjected to thymic sonography. The shape, size in maximal anteroposterior, longitudinal, and transverse dimensions of both right and left lobes of thymus was evaluated. Their echotexture and echogenicity were determined and standards for reference population were calculated. The results were statistically evaluated. RESULTS According to the shape; the thymus was quadrilateral in 156 neonates (62%), round in 37 (15%), bilobate in 32 (13%), and crescent shaped in 25 (10%) in transverse scan. According to the echo texture, the thymus echogenicity was homogeneous and almost similar or slightly less to that of the liver and spleen in most cases 200 [80%] and was coarse in 50 (20%). In all subjects, the echogenicity of the thymus was less than that of the thyroid gland. Variations in location of the thymus were found in 70 (28%) neonates. The range of mean AP diameter was 0.39-2.36 cm (mean 1.43 ± 0.3), that of transverse 1.4 - 4.3 cm (mean 2.16 ± 0.54) and that of thymic index 1.2-5.1 cm2 (mean 2.98 ± 0.64). CONCLUSIONS A normative range was calculated for standard population, which can be used as a reference for comparing thymus size for both normal and diseased neonates for various immunological diseases.
Collapse
|
31
|
Abstract
Age-related changes in the immune system, commonly termed "immunosenescence," contribute to deterioration of the immune response and fundamentally impact the health and survival of elderly individuals. Immunosenescence affects both the innate and adaptive immune systems; however, the most notable changes are in T cell immunity and include thymic involution, the collapse of T cell receptor (TCR) diversity, an imbalance in T cell populations, and the clonal expansion of senescent T cells. Senescent T cells have the ability to produce large quantities of proinflammatory cytokines and cytotoxic mediators; thus, they have been implicated in the pathogenesis of many chronic inflammatory diseases. Recently, an increasing body of evidence has suggested that senescent T cells also have pathogenic potential in cardiovascular diseases, such as hypertension, atherosclerosis, and myocardial infarction, underscoring the detrimental roles of these cells in various chronic inflammatory responses. Given that cardiovascular disease is the number one cause of death worldwide, there is great interest in understanding the contribution of age-related immunological changes to its pathogenesis. In this review, we discuss general features of age-related alterations in T cell immunity and the possible roles of senescent T cells in the pathogenesis of cardiovascular disease.
Collapse
|
32
|
Nigar Z, Itrat M. Evaluation of a Unani polyherbal formulation (Tiryaqe wabai) as an immunostimulator in elderly persons. Anc Sci Life 2014; 33:119-22. [PMID: 25284946 PMCID: PMC4171853 DOI: 10.4103/0257-7941.139054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background and Aims: Elderly subjects have a dysregulation of immune response mainly due to the changes in cell - mediated immunity. Due to their weakened immune response, the elderly are at increased risk of infection and related complications. In Unani medicine Tiryaq wabai was used for the prevention of epidemic diseases during outbreaks, but it has not been explored scientifically so far. The study was aimed to evaluate the immune-stimulating effect of Tiryaq wabai in elderly. Materials and Methods: A randomized placebo controlled trial was conducted at National Institute of Unani Medicine Hospital, Bangalore. Thirty immunocompromised elderly persons were selected on the basis of clinical examination considering parameters like history of recurrent infection, unexplained weight loss, persistent diarrhea etc. They were randomly assigned, 20 in test and 10 in the control group. Tiryaq wabai was given to test group 500 mg orally thrice in a week for 45 days. Placebo was given orally to the control group at a dose of 500 mg thrice in a week for 45 days. Response was assessed by total leucocyte count (TLC), lymphocyte percentage, absolute lymphocyte count (ALC), CD4 and CD8 count. The results were analyzed statistically using Graph Pad InStat 3. Results: The test drug showed statistically significant increase in TLC (P < 0.001), lymphocyte percentage (P < 0.001),ALC (P < 0.001), CD4 count (P < 0.001) in comparison to control group, but increase in CD8 count was not statistically significant. No major adverse effect was observed throughout the study. Conclusion: The findings outlined above indicate immune- stimulating activity of Tiryaq wabai and supports its use in conditions where immunostimulation is required and thus is suggestive of therapeutic usefulness.
Collapse
Affiliation(s)
- Zar Nigar
- Departments of Tahaffuzi wa Samaji Tib National Institute of Unani Medicine, Bengaluru, Karnataka, India
| | - Malik Itrat
- Departments of Tahaffuzi wa Samaji Tib National Institute of Unani Medicine, Bengaluru, Karnataka, India
| |
Collapse
|
33
|
Nikolich-Žugich J. Aging of the T cell compartment in mice and humans: from no naive expectations to foggy memories. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:2622-9. [PMID: 25193936 PMCID: PMC4157314 DOI: 10.4049/jimmunol.1401174] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Until the mid-20th century, infectious diseases were the major cause of morbidity and mortality in humans. Massive vaccination campaigns, antibiotics, antivirals, and advanced public health measures drastically reduced sickness and death from infections in children and younger adults. However, older adults (>65 y of age) remain vulnerable to infections, and infectious diseases remain among the top 5-10 causes of death in this population. The aging of the immune system, often referred to as immune senescence, is the key phenomenon underlying this vulnerability. This review centers on age-related changes in T cells, which are dramatically and reproducibly altered with aging. I discuss changes in T cell production, maintenance, function, and response to latent persistent infection, particularly against CMV, which exerts a profound influence on the aging T cell pool, concluding with a brief list of measures to improve immune function in older adults.
Collapse
Affiliation(s)
- Janko Nikolich-Žugich
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724; and Arizona Center on Aging, University of Arizona College of Medicine, Tucson, AZ 85724
| |
Collapse
|
34
|
Shao H, Ou Y, Wang T, Shen H, Wu F, Zhang W, Tao C, Yuan Y, Bo H, Wang H, Huang S. Differences in TCR-Vβ repertoire and effector phenotype between tumor infiltrating lymphocytes and peripheral blood lymphocytes increase with age. PLoS One 2014; 9:e102327. [PMID: 25019226 PMCID: PMC4096599 DOI: 10.1371/journal.pone.0102327] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 06/16/2014] [Indexed: 12/16/2022] Open
Abstract
Tumor infiltrating lymphocytes (TIL) reflect the host's anti-tumor immune response, and can be a valuable predictor of prognosis. However, many properties of TIL are not fully understood. In the present study, TCR-Vβ repertoires of cancer patients were primarily analyzed by flow cytometry. Abnormally expressed TCR-Vβ subfamilies were generally found in both TIL and peripheral blood lymphocytes (PBL) of each patient. Of note, increased patient age was associated with increasingly biased TCR-Vβ repertoire in TIL but not in PBL, and the dispersion degree of the differences of TCR-Vβ subfamilies between TIL and PBL correlated positively with age (P = 0.007). Utilizing immunoscope analysis, we identified the age-related reduction in TCR-Vβ diversity, but polyclonal pattern was predominant in significantly expanded TCR-Vβ subfamilies. In addition, we found that older patients possessed a decreased ratio of CD8+CD62L+ non-effector cells in TIL compared to PBL, implying age-related increase of CD8+CD62L- effector cells in TIL. The colocalization analysis of CD8 and CD3, however, suggested the suppressed activity of these effector cells in tumor microenvironment. These findings further elucidate the properties of TIL, showing an increasing difference between TIL and PBL with age, which may provide insight for the development of effective immunotherapies for cancer patients of different ages.
Collapse
Affiliation(s)
- Hongwei Shao
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangzhou, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yusheng Ou
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangzhou, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Teng Wang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangzhou, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Han Shen
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangzhou, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Fenglin Wu
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangzhou, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenfeng Zhang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangzhou, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Changli Tao
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangzhou, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yin Yuan
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangzhou, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Huaben Bo
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangzhou, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hui Wang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangzhou, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shulin Huang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangzhou, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
- Southern Medical University, Guangzhou, China
- * E-mail:
| |
Collapse
|
35
|
de Araújo AL, Silva LCR, Fernandes JR, Benard G. Preventing or reversing immunosenescence: can exercise be an immunotherapy? Immunotherapy 2014; 5:879-93. [PMID: 23902557 DOI: 10.2217/imt.13.77] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
There is now a strong body of evidence demonstrating that aging is accompanied by severe alterations in the immune system, a process known as immunosenescence. Among these changes are alterations in T-cell subpopulation size, cytokine secretion pattern, cell replicative capacity and antibody production, all of which culminate in a proinflammatory state called 'inflammaging' and a diminished capacity to respond to new antigens. These alterations are closely related to the increased mortality and morbidity rates observed in this population. However, the role of exercise on the prevention or treatment of immunosenescence is virtually unknown. Data gathered from the literature regarding the effects of physical activity on immune system aging are still limited and conflicting, with existing reports either advocating benefits or asserting a lack of evidence. Exercise as part of a healthy lifestyle has already been shown to provide long-term benefits with regard to cardiovascular, cognitive, psychosocial and other aspects of the elderly. If positive effects are also observed for immunosenescence, exercise could be a highly cost-effective measure to improve human quality of life compared with other strategies currently being pursued.
Collapse
Affiliation(s)
- Adriana L de Araújo
- Laboratory of Dermatology & Immunodeficiencies, Dermatology Division, Clinics Hospital, São Paulo, Brazil
| | | | | | | |
Collapse
|
36
|
Ferrando-Martínez S, Ruiz-Mateos E, Dudakov JA, Velardi E, Grillari J, Kreil DP, Muñoz-Fernandez MÁ, van den Brink MRM, Leal M. WNT signaling suppression in the senescent human thymus. J Gerontol A Biol Sci Med Sci 2014; 70:273-81. [PMID: 24657825 DOI: 10.1093/gerona/glu030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human thymus is completely developed in late fetal stages and its function peaks in newborns. After the first year of life, the thymus undergoes a progressive atrophy that dramatically decreases de novo T-lymphocyte maturation. Hormonal signaling and changes in the microRNA expression network are identified as underlying causes of human thymus involution. However, specific pathways involved in the age-related loss of thymic function remain unknown. In this study, we analyzed differential gene-expression profile and microRNA expression in elderly (70 years old) and young (less than 10 months old and 11 years old) human thymic samples. Our data have shown that WNT pathway deregulation through the overexpression of different inhibitors by the nonadipocytic component of the human thymus stimulates the age-related involution. These results are of particular interest because interference of WNT signaling has been demonstrated in both animal models and in vitro studies, with the three major hallmarks of thymic involution: (i) epithelial structure disruption, (ii) adipogenic process, and (iii) thymocyte development arrest. Thus, our results suggest that secreted inhibitors of the WNT pathway could be explored as a novel therapeutical target in the reversal of the age-related thymic involution.
Collapse
Affiliation(s)
- Sara Ferrando-Martínez
- Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain. Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain. Laboratory of Immunovirology, Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine. Institute of Biomedicine of Seville, IBiS, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain.
| | - Ezequiel Ruiz-Mateos
- Laboratory of Immunovirology, Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine. Institute of Biomedicine of Seville, IBiS, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| | - Jarrod A Dudakov
- Department of Immunology and Medicine, Sloan Kettering Institute, New York City, USA
| | - Enrico Velardi
- Department of Immunology and Medicine, Sloan Kettering Institute, New York City, USA
| | - Johannes Grillari
- Department of Biotechnology, VIBT-BOKU, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| | - David P Kreil
- Chair of Bioinformatics, BOKU University Vienna, Austria and Life Sciences, University of Warwick, UK
| | - M Ángeles Muñoz-Fernandez
- Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain. Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | | | - Manuel Leal
- Laboratory of Immunovirology, Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine. Institute of Biomedicine of Seville, IBiS, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| |
Collapse
|
37
|
Klein N, Sefe D, Mosconi I, Zanchetta M, Castro H, Jacobsen M, Jones H, Bernardi S, Pillay D, Giaquinto C, Walker AS, Gibb DM, De Rossi A. The immunological and virological consequences of planned treatment interruptions in children with HIV infection. PLoS One 2013; 8:e76582. [PMID: 24194841 PMCID: PMC3806774 DOI: 10.1371/journal.pone.0076582] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 08/26/2013] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES To evaluate the immunological and viral consequences of planned treatment interruptions (PTI) in children with HIV. DESIGN This was an immunological and virological sub-study of the Paediatric European Network for Treatment of AIDS (PENTA) 11 trial, which compared CD4-guided PTI of antiretroviral therapy (ART) with continuous therapy (CT) in children. METHODS HIV-1 RNA and lymphocyte subsets, including CD4 and CD8 cells, were quantified on fresh samples collected during the study; CD45RA, CD45RO and CD31 subpopulations were evaluated in some centres. For 36 (18 PTI, 18 CT) children, immunophenotyping was performed and cell-associated HIV-1 DNA analysed on stored samples to 48 weeks. RESULTS In the PTI group, CD4 cell count fell rapidly in the first 12 weeks off ART, with decreases in both naïve and memory cells. However, the proportion of CD4 cells expressing CD45RA and CD45RO remained constant in both groups. The increase in CD8 cells in the first 12 weeks off ART in the PTI group was predominantly due to increases in RO-expressing cells. PTI was associated with a rapid and sustained increase in CD4 cells expressing Ki67 and HLA-DR, and increased levels of HIV-1 DNA. CONCLUSIONS PTI in children is associated with rapid changes in CD4 and CD8 cells, likely due to increased cell turnover and immune activation. However, children off treatment may be able to maintain stable levels of naïve CD4 cells, at least in proportion to the memory cell pool, which may in part explain the observed excellent CD4 cell recovery with re-introduction of ART.
Collapse
Affiliation(s)
- Nigel Klein
- Institute of Child Health, University College London, London, United Kingdom
- * E-mail:
| | - Delali Sefe
- Institute of Child Health, University College London, London, United Kingdom
| | - Ilaria Mosconi
- AIDS Reference Center, University of Padova, Padova, Italy
| | | | - Hannah Castro
- Medical Research Council Clinical Trials Unit, London, United Kingdom
| | - Marianne Jacobsen
- Institute of Child Health, University College London, London, United Kingdom
| | - Hannah Jones
- Institute of Child Health, University College London, London, United Kingdom
| | | | - Deenan Pillay
- University College London/Medical Research Council Centre for Medical Molecular Virology, University College London Medical School, London, United Kingdom
| | - Carlo Giaquinto
- Department of Paediatrics, University of Padova, Padova, Italy
| | - A. Sarah Walker
- Medical Research Council Clinical Trials Unit, London, United Kingdom
| | - Diana M. Gibb
- Medical Research Council Clinical Trials Unit, London, United Kingdom
| | - Anita De Rossi
- AIDS Reference Center, University of Padova, Padova, Italy
| | | |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Aging of the hematopoietic system is associated with myeloid malignancies, anemia and immune dysfunction. As hematopoietic stem cells (HSCs) generate all cells of the hematopoietic system, age-associated changes in HSCs may underlie many features of the aged hematopoietic system. Recent findings on age-associated changes in HSCs are reviewed here. RECENT FINDINGS Aged HSCs are myeloid biased, have acquired DNA damage and are functionally compromised. However, overall function of the HSC compartment is well maintained through age-associated expansion of HSCs. Many age-related changes in the hematopoietic system, in particular the clonal myeloid bias of HSCs and the decrease in B and T-cell development, in fact begin during development. Furthermore, HSCs possess specific protective mechanisms aimed at maintaining their number, even at the expense of accumulating damaged cells. SUMMARY We argue that age-related changes in HSCs and in the hematopoietic system may not entirely be due to a degenerative aging process, but are the result of developmental and stem cell-protective mechanisms aimed at maximizing fitness during reproductive life. These mechanisms may be disadvantageous later in life as damaged HSCs accumulate and establishment of responses to neoantigens becomes compromised because of the reduced generation of naive T and B cells.
Collapse
|
39
|
Abstract
Age-related regression of the thymus is associated with a decline in naïve T cell output. This is thought to contribute to the reduction in T cell diversity seen in older individuals and linked with increased susceptibility to infection, autoimmune disease, and cancer. Thymic involution is one of the most dramatic and ubiquitous changes seen in the aging immune system, but the mechanisms which underlying this process are poorly understood. However, a picture is emerging, implicating the involvement of both extrinsic and intrinsic factors. In this review we assess the role of the thymic microenvironment as a potential target that regulates thymic involution, question whether thymocyte development in the aged thymus is functionally impaired, and explore the kinetics of thymic involution.
Collapse
Affiliation(s)
- Donald B Palmer
- Infection and Immunity Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London , London , UK
| |
Collapse
|
40
|
Montecino-Rodriguez E, Berent-Maoz B, Dorshkind K. Causes, consequences, and reversal of immune system aging. J Clin Invest 2013; 123:958-65. [PMID: 23454758 DOI: 10.1172/jci64096] [Citation(s) in RCA: 536] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The effects of aging on the immune system are manifest at multiple levels that include reduced production of B and T cells in bone marrow and thymus and diminished function of mature lymphocytes in secondary lymphoid tissues. As a result, elderly individuals do not respond to immune challenge as robustly as the young. An important goal of aging research is to define the cellular changes that occur in the immune system and the molecular events that underlie them. Considerable progress has been made in this regard, and this information has provided the rationale for clinical trials to rejuvenate the aging immune system.
Collapse
Affiliation(s)
- Encarnacion Montecino-Rodriguez
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | | | | |
Collapse
|
41
|
Ferrando-Martínez S, de la Fuente M, Guerrero JM, Leal M, Muñoz-Fernández MÁ. [Impact of thymic function in age-related immune deterioration]. Rev Esp Geriatr Gerontol 2013; 48:232-7. [PMID: 23453427 DOI: 10.1016/j.regg.2012.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 11/14/2012] [Accepted: 11/16/2012] [Indexed: 01/09/2023]
Abstract
Age-related biological deterioration also includes immune system deterioration and, in consequence, a rise in the incidence and prevalence of infections and cancers, as well as low responses to vaccination strategies. Out of all immune cell subsets, T-lymphocytes seem to be involved in most of the age-related defects. Since T-lymphocytes mature during their passage through the thymus, and the thymus shows an age-related process of atrophy, thymic regression has been proposed as the triggering event of this immune deterioration in elderly people. Historically, it has been accepted that the young thymus sets the T-lymphocyte repertoire during the childhood, whereupon atrophy begins until the elderly thymus is a non-functional evolutionary trace. However, a rising body of knowledge points toward the thymus functioning during adulthood. In the elderly, higher thymic function is associated with a younger immune system, while thymic function failure is associated with all-cause mortality. Therefore, any new strategy focused on the improvement of the elderly quality of life, especially those trying to influence the immune system, should take into account, together with peripheral homeostasis, thymus function as a key element in slowing down age-related decline.
Collapse
Affiliation(s)
- Sara Ferrando-Martínez
- Laboratorio de Inmuno-Biología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, España; Laboratorio de Inmunovirología, Unidad Clínica de Enfermedades Infecciosas, Microbiología y Medicina Preventiva, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, España.
| | | | | | | | | |
Collapse
|
42
|
Buchholz VR, Gräf P, Busch DH. The smallest unit: effector and memory CD8(+) T cell differentiation on the single cell level. Front Immunol 2013; 4:31. [PMID: 23424063 PMCID: PMC3573211 DOI: 10.3389/fimmu.2013.00031] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 01/26/2013] [Indexed: 12/16/2022] Open
Abstract
CD8+ T cell immune responses provide immediate protection against primary infection and durable memory capable of rapidly fighting off re-infection. Immediate protection and lasting memory are implemented by phenotypically and functionally distinct T cell subsets. While it is now widely accepted that these diverge from a common source of naïve T cells (Tn), the developmental relation and succession of effector and memory T cell subsets is still under intense debate. Recently, a distinct memory T cell subset has been suggested to possess stem cell-like features, sparking the hope to harness its capacity for self-renewal and diversification for successful therapy of chronic infections or malignant diseases. In this review we highlight current developmental models of memory generation, T cell subset diversification and T cell stemness. We discuss the importance of single cell monitoring techniques for adequately mapping these developmental processes and take a brief look at signaling components active in the putative stem cell-like memory T cell compartment.
Collapse
Affiliation(s)
- Veit R Buchholz
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München Munich, Germany
| | | | | |
Collapse
|
43
|
The needs for HIV treatment and care of children, adolescents, pregnant women and older people in low-income and middle-income countries. AIDS 2012; 26 Suppl 2:S105-16. [PMID: 23303433 DOI: 10.1097/qad.0b013e32835bddfc] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Success in diagnosing and treating HIV-infected adults has, where HIV care and treatment is available, turned HIV into a chronic, rather than life-limiting disease. Progress meeting the needs of HIV-infected children, perinatally and horizontally infected adolescents, pregnant women and older people has lagged behind. We review the special needs and barriers to scaling up care and antiretroviral therapy (ART) coverage in these populations. DESIGN AND METHODS A literature review combined with personal views and operational experience specifically from countries covered by the Evidence for Action programme. RESULTS Challenges include logistics of diagnosis and treatment in pregnancy, difficulties in early infant diagnosis, availability of appropriate paediatric formulations, management of adolescents, and comorbidities in older people. CONCLUSION Priorities for development need to focus upon the simplification of HIV care to allow provision for all ages at the primary healthcare level. Specific priorities include focused use of virological testing in infants, ongoing development of dispersible and scored fixed-dose ART combinations suitable for use across ages, development of 'adolescent-friendly' HIV services catering for perinatally and horizontally infected adolescents to improve adherence and reduce onward transmissions, simplification of referral pathways to ensure all pregnant women are tested for HIV and commenced on ART, and education of healthcare workers on the specific needs of HIV care in older patients. Each priority will be reviewed and potential solutions discussed.
Collapse
|
44
|
Holland AM, Zakrzewski JL, Tsai JJ, Hanash AM, Dudakov JA, Smith OM, West ML, Singer NV, Brill J, Sun JC, van den Brink MRM. Extrathymic development of murine T cells after bone marrow transplantation. J Clin Invest 2012; 122:4716-26. [PMID: 23160195 DOI: 10.1172/jci60630] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 09/27/2012] [Indexed: 11/17/2022] Open
Abstract
Restoring T cell competence is a significant clinical challenge in patients whose thymic function is severely compromised due to age or cytoreductive conditioning. Here, we demonstrate in mice that mesenteric LNs (MLNs) support extrathymic T cell development in euthymic and athymic recipients of bone marrow transplantation (BMT). Furthermore, in aged murine BMT recipients, the contribution of the MLNs to the generation of T cells was maintained, while the contribution of the thymus was significantly impaired. Thymic impairment resulted in a proportional increase in extrathymic-derived T cell progenitors. Extrathymic development in athymic recipients generated conventional naive TCRαβ T cells with a broad Vβ repertoire and intact functional and proliferative potential. Moreover, in the absence of a functional thymus, immunity against known pathogens could be augmented using engineered precursor T cells with viral specificity. These findings demonstrate the potential of extrathymic T cell development for T cell reconstitution in patients with limited thymic function.
Collapse
Affiliation(s)
- Amanda M Holland
- Department of Immunology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
With the improvement of medical care and hygienic conditions, there has been a tremendous increment in human lifespan. However, many of the elderly (>65 years) display chronic illnesses, and a majority requires frequent and longer hospitalization. The robustness of the immune system to eliminate or control infections is often eroded with advancing age. Nevertheless, some elderly individuals do cope better than others. The origin of these inter-individual differences may come from genetic, lifestyle conditions (nutrition, socio-economic parameters), as well as the type, number and recurrence of pathogens encountered during life. The theory we are supporting is that chronic infections, through life, will induce profound changes in the immune system probably due to unbalanced inflammatory profiles. Persistent viruses such a cytomegalovirus are not eliminated and are a driven force to immune exhaustion. Because of their age, elderly individuals may have seen more of these chronic stimulators and have experienced more reactivation episodes ultimately leading to shrinkage of their repertoire and overall immune robustness. This review integrates updates on immunity with advancing age and its impact on associated clinical conditions.
Collapse
|
46
|
Impact of immune-metabolic interactions on age-related thymic demise and T cell senescence. Semin Immunol 2012; 24:321-30. [DOI: 10.1016/j.smim.2012.04.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 03/29/2012] [Accepted: 04/09/2012] [Indexed: 01/13/2023]
|
47
|
Chinn IK, Blackburn CC, Manley NR, Sempowski GD. Changes in primary lymphoid organs with aging. Semin Immunol 2012; 24:309-20. [PMID: 22559987 PMCID: PMC3415579 DOI: 10.1016/j.smim.2012.04.005] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 03/30/2012] [Accepted: 04/09/2012] [Indexed: 12/13/2022]
Abstract
Aging is associated with decreased immune function that leads to increased morbidity and mortality in the elderly. Immune senescence is accompanied by age-related changes in two primary lymphoid organs, bone marrow and thymus, that result in decreased production and function of B and T lymphocytes. In bone marrow, hematopoietic stem cells exhibit reduced self-renewal potential, increased skewing toward myelopoiesis, and decreased production of lymphocytes with aging. These functional sequelae of aging are caused in part by increased oxidative stress, inflammation, adipocyte differentiation, and disruption of hypoxic osteoblastic niches. In thymus, aging is associated with tissue involution, exhibited by a disorganization of the thymic epithelial cell architecture and increased adiposity. This dysregulation correlates with a loss of stroma-thymocyte 'cross-talk', resulting in decreased export of naïve T cells. Mounting evidence argues that with aging, thymic inflammation, systemic stress, local Foxn1 and keratinocyte growth factor expression, and sex steroid levels play critical roles in actively driving thymic involution and overall adaptive immune senescence across the lifespan. With a better understanding of the complex mechanisms and pathways that mediate bone marrow and thymus involution with aging, potential increases for the development of safe and effective interventions to prevent or restore loss of immune function with aging.
Collapse
Affiliation(s)
- Ivan K. Chinn
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, 27710 USA
- Duke Human Vaccine Institute, Box 103020, Duke University Medical Center, Durham, North Carolina, 27710 USA
| | - Clare C. Blackburn
- MRC Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, SCRM Building, 5 Little France Drive, Edinburgh, United Kingdom EH16 4UU
| | - Nancy R. Manley
- Department of Genetics, University of Georgia, 500 DW Brooks Drive, S270B Coverdell Building, Athens, Georgia, 30602 USA
| | - Gregory D. Sempowski
- Duke Human Vaccine Institute, Box 103020, Duke University Medical Center, Durham, North Carolina, 27710 USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, 27710 USA
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, 27710 USA
| |
Collapse
|
48
|
Parietti V, Nelson E, Telliam G, Le Noir S, Pla M, Delord M, Vanneaux V, Mohtashami M, Macintyre EA, Gluckman JC, Asnafi V, Zúñiga-Pflücker JC, Larghero J, Canque B. Dynamics of Human Prothymocytes and Xenogeneic Thymopoiesis in Hematopoietic Stem Cell-Engrafted Nonobese Diabetic-SCID/IL-2rγnullMice. THE JOURNAL OF IMMUNOLOGY 2012; 189:1648-60. [DOI: 10.4049/jimmunol.1201251] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
49
|
Suboptimal immune reconstitution in vertically HIV infected children: a view on how HIV replication and timing of HAART initiation can impact on T and B-cell compartment. Clin Dev Immunol 2012; 2012:805151. [PMID: 22550537 PMCID: PMC3328919 DOI: 10.1155/2012/805151] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 11/21/2011] [Accepted: 12/10/2011] [Indexed: 11/18/2022]
Abstract
Today, HIV-infected children who have access to treatment face a chronic rather than a progressive and fatal disease. As a result, new challenges are emerging in the field. Recent lines of evidence outline several factors that can differently affect the ability of the immune system to fully reconstitute and to mount specific immune responses in children receiving HAART. In this paper, we review the underlying mechanisms of immune reconstitution after HAART initiation among vertically HIV-infected children analyzing the possible causes of suboptimal responses.
Collapse
|
50
|
Cuddihy AR, Suterwala BT, Ge S, Kohn LA, Jang J, Andrade J, Wang X, Crooks GM. Rapid thymic reconstitution following bone marrow transplantation in neonatal mice is VEGF-dependent. Biol Blood Marrow Transplant 2012; 18:683-9. [PMID: 22281302 DOI: 10.1016/j.bbmt.2012.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/10/2012] [Indexed: 12/27/2022]
Abstract
Age-related differences in thymic function influence the rapidity of T cell reconstitution following hematopoietic stem cell transplantation (HSCT). In adults, thymic reconstitution is delayed until after marrow engraftment is established, and is significantly improved by approaches that increase marrow chimerism, such as pretransplantation irradiation. In contrast, we show that neonatal mice undergo more rapid and efficient thymic reconstitution than adults, even when bone marrow (BM) engraftment is minimal and in the absence of pretransplantation radiation. We have previously shown that the neonatal thymus produces high levels of vascular endothelial growth factor (VEGF) that drives angiogenesis locally. In this report, we show that inhibition of VEGF prior to HSCT prevents rapid thymic reconstitution in neonates, but has no effect on thymic reconstitution in adults. These data suggest that the early radiation-independent thymic reconstitution unique to the neonatal host is mediated through VEGF, and reveals a novel pathway that might be targeted to improve immune reconstitution post-HSCT.
Collapse
Affiliation(s)
- Andrew R Cuddihy
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | | | |
Collapse
|