1
|
Chen S, Tan Y, Tian L. Immunophenotypes in psychosis: is it a premature inflamm-aging disorder? Mol Psychiatry 2024; 29:2834-2848. [PMID: 38532012 PMCID: PMC11420084 DOI: 10.1038/s41380-024-02539-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/28/2024]
Abstract
Immunopsychiatric field has rapidly accumulated evidence demonstrating the involvement of both innate and adaptive immune components in psychotic disorders such as schizophrenia. Nevertheless, researchers are facing dilemmas of discrepant findings of immunophenotypes both outside and inside the brains of psychotic patients, as discovered by recent meta-analyses. These discrepancies make interpretations and interrogations on their roles in psychosis remain vague and even controversial, regarding whether certain immune cells are more activated or less so, and whether they are causal or consequential, or beneficial or harmful for psychosis. Addressing these issues for psychosis is not at all trivial, as immune cells either outside or inside the brain are an enormously heterogeneous and plastic cell population, falling into a vast range of lineages and subgroups, and functioning differently and malleably in context-dependent manners. This review aims to overview the currently known immunophenotypes of patients with psychosis, and provocatively suggest the premature immune "burnout" or inflamm-aging initiated since organ development as a potential primary mechanism behind these immunophenotypes and the pathogenesis of psychotic disorders.
Collapse
Affiliation(s)
- Song Chen
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, PR China
| | - Yunlong Tan
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, PR China
| | - Li Tian
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
2
|
Lev A, Somech R, Somekh I. Newborn screening for severe combined immunodeficiency and inborn errors of immunity. Curr Opin Pediatr 2023; 35:692-702. [PMID: 37707504 DOI: 10.1097/mop.0000000000001291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
PURPOSE OF REVIEW Severe combined immune deficiency (SCID) is the most devastating genetic disease of the immune system with an unfavorable outcome unless diagnosed early in life. Newborn screening (NBS) programs play a crucial role in facilitating early diagnoses and timely interventions for affected infants. RECENT FINDINGS SCID marked the pioneering inborn error of immunity (IEI) to undergo NBS, a milestone achieved 15 years ago through the enumeration of T-cell receptor excision circles (TRECs) extracted from Guthrie cards. This breakthrough has revolutionized our approach to SCID, enabling not only presymptomatic identification and prompt treatments (including hematopoietic stem cell transplantation), but also enhancing our comprehension of the global epidemiology of SCID. SUMMARY NBS is continuing to evolve with the advent of novel diagnostic technologies and treatments. Following the successful implementation of SCID-NBS programs, a call for the early identification of additional IEIs is the next step, encompassing a broader spectrum of IEIs, facilitating early diagnoses, and preventing morbidity and mortality.
Collapse
Affiliation(s)
- Atar Lev
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Affiliated to the Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | | | | |
Collapse
|
3
|
Levy-Mendelovich S, Lev A, Avishai E, Budnik I, Dardik R, Barg AA, Somech R, Kenet G. Can T-cell and B-cell excision circles predict development of inhibitors in pediatric hemophilia A? Pediatr Res 2023; 93:1546-1550. [PMID: 36045224 DOI: 10.1038/s41390-022-02268-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/06/2022] [Accepted: 08/05/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Hemophilia A (HA) therapy requires intravenous replacement infusions of factor (F) VIII concentrate. Inhibitors are high-affinity immunoglobulin G that are directed against FVIII and thereby render replacement therapy ineffective. This complication has significant prognostic implications. We aimed to examine the immune system involvement in inhibitor formation specifically T-cell excision circles (TRECs) and B-cell excision circles (KRECs), markers of new T and B cells, respectively, and examine them as surrogate markers for inhibitor formation. METHODS Blood samples were collected from 35 children with severe HA. Children were divided into two groups: with FVIII inhibitors and without FVIII inhibitors. TRECs and KRECs were measured in peripheral blood. RESULTS A total of 11 patients with inhibitors and 24 without were evaluated. Children with inhibitors had higher levels of TRECs however not statistically significant (p = 0.085). CjKREC levels were higher in the inhibitor patients (p = 0.003). Moreover, the sj/cjKREC ratio was lower in the inhibitor patients (p = 0.015). CONCLUSIONS Our findings may add to the notion that inhibitor formation is attributed to humoral immunity due to peripheral B-cell expansion and loss of peripheral tolerance. Improved knowledge regarding the involvement of the immune system in the formation of FVIII inhibitors will enable better therapy tailoring in the era of non-replacement therapies. IMPACT The etiology of FVIII inhibitor formation is multifactorial, in which the immune system plays a pivotal role. Our findings may add to the notion that inhibitor formation is attributed to humoral immunity due to peripheral B-cell expansion and production of antibodies against FVIII. Improved knowledge regarding the involvement of the immune system in the development of FVIII inhibitors will enable the identification of patients prone to inhibitor development and better therapy tailoring in the new era of non-replacement therapies.
Collapse
Affiliation(s)
- Sarina Levy-Mendelovich
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel.
- Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
- The Sheba Talpiot Medical Leadership Program, Tel Hashomer, Israel.
| | - Atar Lev
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation canter, Edmond and Lily Safra Children's Hospital, Sheba Medical Center affiliated with Sackler school of medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Einat Avishai
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel
- Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ivan Budnik
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Rima Dardik
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel
- Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Asaaf Arie Barg
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel
- Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Raz Somech
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation canter, Edmond and Lily Safra Children's Hospital, Sheba Medical Center affiliated with Sackler school of medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Gili Kenet
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel
- Amalia Biron Research Institute of Thrombosis and Hemostasis, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Fisher IF, Shemer R, Dor Y. Epigenetic liquid biopsies: a novel putative biomarker in immunology and inflammation. Trends Immunol 2023; 44:356-364. [PMID: 37012121 DOI: 10.1016/j.it.2023.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 04/03/2023]
Abstract
Immune and inflammatory processes occurring within tissues are often undetectable by blood cell counts, standard circulating biomarkers, or imaging, representing an unmet biomedical need. Here, we outline recent advances indicating that liquid biopsies can broadly inform human immune system dynamics. Nucleosome-size fragments of cell-free DNA (cfDNA) released from dying cells into blood contain rich epigenetic information such as methylation, fragmentation, and histone mark patterns. This information allows to infer the cfDNA cell of origin, as well as pre-cell death gene expression patterns. We propose that the analysis of epigenetic features of immune cell-derived cfDNA can shed light on immune cell turnover dynamics in healthy people, and inform the study and diagnosis of cancer, local inflammation, infectious or autoimmune diseases, as well as responses to vaccination.
Collapse
|
5
|
Sadeghalvad M, Rezaei N. Immunodeficiencies. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
6
|
McCullough KM, Katrinli S, Hartmann J, Lori A, Klengel C, Missig G, Klengel T, Langford NA, Newman EL, Anderson KJ, Smith AK, Carroll FI, Ressler KJ, Carlezon WA. Blood levels of T-Cell Receptor Excision Circles (TRECs) provide an index of exposure to traumatic stress in mice and humans. Transl Psychiatry 2022; 12:423. [PMID: 36192377 PMCID: PMC9530209 DOI: 10.1038/s41398-022-02159-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022] Open
Abstract
Exposure to stress triggers biological changes throughout the body. Accumulating evidence indicates that alterations in immune system function are associated with the development of stress-associated illnesses such as major depressive disorder and post-traumatic stress disorder, increasing interest in identifying immune markers that provide insight into mental health. Recombination events during T-cell receptor rearrangement and T-cell maturation in the thymus produce circular DNA fragments called T-cell receptor excision circles (TRECs) that can be utilized as indicators of thymic function and numbers of newly emigrating T-cells. Given data suggesting that stress affects thymus function, we examined whether blood levels of TRECs might serve as a quantitative peripheral index of cumulative stress exposure and its physiological correlates. We hypothesized that chronic stress exposure would compromise thymus function and produce corresponding decreases in levels of TRECs. In male mice, exposure to chronic social defeat stress (CSDS) produced thymic involution, adrenal hypertrophy, and decreased levels of TRECs in blood. Extending these studies to humans revealed robust inverse correlations between levels of circulating TRECs and childhood emotional and physical abuse. Cell-type specific analyses also revealed associations between TREC levels and blood cell composition, as well as cell-type specific methylation changes in CD4T + and CD8T + cells. Additionally, TREC levels correlated with epigenetic age acceleration, a common biomarker of stress exposure. Our findings demonstrate alignment between findings in mice and humans and suggest that blood-borne TRECs are a translationally-relevant biomarker that correlates with, and provides insight into, the cumulative physiological and immune-related impacts of stress exposure in mammals.
Collapse
Affiliation(s)
- Kenneth M McCullough
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Seyma Katrinli
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Jakob Hartmann
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Adriana Lori
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Claudia Klengel
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Galen Missig
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Torsten Klengel
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Nicole A Langford
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Emily L Newman
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Kasey J Anderson
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Alicia K Smith
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - F Ivy Carroll
- Center for Organic and Medicinal Chemistry, Research Triangle Institute, Research Triangle Park, NC, USA
| | - Kerry J Ressler
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - William A Carlezon
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
7
|
Lev A, Sharir I, Simon AJ, Levy S, Lee YN, Frizinsky S, Daas S, Saraf-Levy T, Broides A, Nahum A, Hanna S, Stepensky P, Toker O, Dalal I, Etzioni A, Stein J, Adam E, Hendel A, Marcus N, Almashanu S, Somech R. Lessons Learned From Five Years of Newborn Screening for Severe Combined Immunodeficiency in Israel. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:2722-2731.e9. [PMID: 35487367 DOI: 10.1016/j.jaip.2022.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/03/2022] [Accepted: 04/05/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Implementation of newborn screening (NBS) programs for severe combined immunodeficiency (SCID) have advanced the diagnosis and management of affected infants and undoubtedly improved their outcomes. Reporting long-term follow-up of such programs is of great importance. OBJECTIVE We report a 5-year summary of the NBS program for SCID in Israel. METHODS Immunologic and genetic assessments, clinical analyses, and outcome data from all infants who screened positive were evaluated and summarized. RESULTS A total of 937,953 Guthrie cards were screened for SCID. A second Guthrie card was requested on 1,169 occasions (0.12%), which resulted in 142 referrals (0.015%) for further validation tests. Flow cytometry immune-phenotyping, T cell receptor excision circle measurement in peripheral blood, and expression of TCRVβ repertoire for the validation of positive cases revealed a specificity and sensitivity of 93.7% and 75.9%, respectively, in detecting true cases of SCID. Altogether, 32 SCID and 110 non-SCID newborns were diagnosed, making the incidence of SCID in Israel as high as 1:29,000 births. The most common genetic defects in this group were associated with mutations in DNA cross-link repair protein 1C and IL-7 receptor α (IL-7Rα) genes. No infant with SCID was missed during the study time. Twenty-two SCID patients underwent hematopoietic stem cell transplantation, which resulted in a 91% survival rate. CONCLUSIONS Newborn screening for SCID should ultimately be applied globally, specifically to areas with high rates of consanguineous marriages. Accumulating data from follow-up studies on NBS for SCID will improve diagnosis and treatment and enrich our understanding of immune development in health and disease.
Collapse
Affiliation(s)
- Atar Lev
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Mina and Everard Goodman Faculty of Life Sciences, Advanced Materials and Nanotechnology Institute, Bar-Ilan University, Ramat-Gan, Israel
| | - Idan Sharir
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Amos J Simon
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Hemato-Immunology Unit, Hematology Lab, Sheba Medical Center, Tel HaShomer, Israel
| | - Shiran Levy
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yu Nee Lee
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shirly Frizinsky
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Suha Daas
- National Newborn Screening Program, Ministry of Health, Tel-HaShomer, Israel
| | - Talia Saraf-Levy
- National Newborn Screening Program, Ministry of Health, Tel-HaShomer, Israel
| | - Arnon Broides
- Pediatric Immunology, Soroka University Medical Center, Beer-Sheva, Israel; Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY
| | - Amit Nahum
- Pediatric Immunology, Soroka University Medical Center, Beer-Sheva, Israel; Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Primary Immunodeficiency Research Laboratory, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Suhair Hanna
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Ruth Children Hospital, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Polina Stepensky
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Department of Bone Marrow Transplantation, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ori Toker
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Faculty of Medicine, Hebrew University of Jerusalem, Israel; Allergy and Immunology Unit, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Ilan Dalal
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Department of Pediatrics, Pediatric Allergy Unit, E. Wolfson Medical Center, Holon, Israel, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Amos Etzioni
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Ruth Children Hospital, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Jerry Stein
- Department for Hemato-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Etai Adam
- Division of Pediatric Hematology and Oncology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel
| | - Ayal Hendel
- Mina and Everard Goodman Faculty of Life Sciences, Advanced Materials and Nanotechnology Institute, Bar-Ilan University, Ramat-Gan, Israel
| | - Nufar Marcus
- Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; Allergy and Immunology Unit, Schneider Children's Medical Center of Israel, Felsenstein Medical Research Center, Kipper Institute of Immunology, Petach Tikva, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Shlomo Almashanu
- National Newborn Screening Program, Ministry of Health, Tel-HaShomer, Israel.
| | - Raz Somech
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Jeffrey Modell Foundation Israeli Network for Primary Immunodeficiency, New York, NY; National Lab for Confirming Primary Immunodeficiency in Newborn Screening Center for Newborn Screening, Ministry of Health, Tel HaShomer, Israel.
| |
Collapse
|
8
|
Kashatnikova DA, Khadzhieva MB, Kolobkov DS, Belopolskaya OB, Smelaya TV, Gracheva AS, Kalinina EV, Larin SS, Kuzovlev AN, Salnikova LE. Pneumonia and Related Conditions in Critically Ill Patients—Insights from Basic and Experimental Studies. Int J Mol Sci 2022; 23:ijms23179896. [PMID: 36077293 PMCID: PMC9456259 DOI: 10.3390/ijms23179896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
Pneumonia is an acute infectious disease with high morbidity and mortality rates. Pneumonia’s development, severity and outcome depend on age, comorbidities and the host immune response. In this study, we combined theoretical and experimental investigations to characterize pneumonia and its comorbidities as well as to assess the host immune response measured by TREC/KREC levels in patients with pneumonia. The theoretical study was carried out using the Columbia Open Health Data (COHD) resource, which provides access to clinical concept prevalence and co-occurrence from electronic health records. The experimental study included TREC/KREC assays in young adults (18–40 years) with community-acquired (CAP) (n = 164) or nosocomial (NP) (n = 99) pneumonia and healthy controls (n = 170). Co-occurring rates between pneumonia, sepsis, acute respiratory distress syndrome (ARDS) and some other related conditions common in intensive care units were the top among 4170, 3382 and 963 comorbidities in pneumonia, sepsis and ARDS, respectively. CAP patients had higher TREC levels, while NP patients had lower TREC/KREC levels compared to controls. Low TREC and KREC levels were predictive for the development of NP, ARDS, sepsis and lethal outcome (AUCTREC in the range 0.71–0.82, AUCKREC in the range 0.67–0.74). TREC/KREC analysis can be considered as a potential prognostic test in patients with pneumonia.
Collapse
Affiliation(s)
- Darya A. Kashatnikova
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Maryam B. Khadzhieva
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
- The Laboratory of Molecular Immunology, Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow 117198, Russia
| | - Dmitry S. Kolobkov
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Olesya B. Belopolskaya
- The Resource Center “Bio-Bank Center”, Research Park of St. Petersburg State University, St. Petersburg 199034, Russia
| | - Tamara V. Smelaya
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Alesya S. Gracheva
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Ekaterina V. Kalinina
- The Laboratory of Molecular Immunology, Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow 117198, Russia
| | - Sergey S. Larin
- The Laboratory of Molecular Immunology, Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow 117198, Russia
| | - Artem N. Kuzovlev
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Lyubov E. Salnikova
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
- The Laboratory of Molecular Immunology, Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow 117198, Russia
- Correspondence:
| |
Collapse
|
9
|
Galimberti S, Balducci S, Guerrini F, Del Re M, Cacciola R. Digital Droplet PCR in Hematologic Malignancies: A New Useful Molecular Tool. Diagnostics (Basel) 2022; 12:1305. [PMID: 35741115 PMCID: PMC9221914 DOI: 10.3390/diagnostics12061305] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/21/2022] [Accepted: 05/22/2022] [Indexed: 01/27/2023] Open
Abstract
Digital droplet PCR (ddPCR) is a recent version of quantitative PCR (QT-PCR), useful for measuring gene expression, doing clonality assays and detecting hot spot mutations. In respect of QT-PCR, ddPCR is more sensitive, does not need any reference curve and can quantify one quarter of samples already defined as "positive but not quantifiable". In the IgH and TCR clonality assessment, ddPCR recapitulates the allele-specific oligonucleotide PCR (ASO-PCR), being not adapt for detecting clonal evolution, that, on the contrary, does not represent a pitfall for the next generation sequencing (NGS) technique. Differently from NGS, ddPCR is not able to sequence the whole gene, but it is useful, cheaper, and less time-consuming when hot spot mutations are the targets, such as occurs with IDH1, IDH2, NPM1 in acute leukemias or T315I mutation in Philadelphia-positive leukemias or JAK2 in chronic myeloproliferative neoplasms. Further versions of ddPCR, that combine different primers/probes fluorescences and concentrations, allow measuring up to four targets in the same PCR reaction, sparing material, time, and money. ddPCR is also useful for quantitating BCR-ABL1 fusion gene, WT1 expression, donor chimerism, and minimal residual disease, so helping physicians to realize that "patient-tailored therapy" that is the aim of the modern hematology.
Collapse
Affiliation(s)
- Sara Galimberti
- Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, 56126 Pisa, Italy; (S.G.); (S.B.); (F.G.); (M.D.R.)
| | - Serena Balducci
- Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, 56126 Pisa, Italy; (S.G.); (S.B.); (F.G.); (M.D.R.)
| | - Francesca Guerrini
- Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, 56126 Pisa, Italy; (S.G.); (S.B.); (F.G.); (M.D.R.)
| | - Marzia Del Re
- Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, 56126 Pisa, Italy; (S.G.); (S.B.); (F.G.); (M.D.R.)
| | - Rossella Cacciola
- Department of Clinical and Experimental Medicine, Section of Hemostasis, University of Catania, 95123 Catania, Italy
| |
Collapse
|
10
|
Weiss B, Ben-Horin S, Lev A, Broide E, Yavzori M, Lahat A, Kopylov U, Picard O, Eliakim R, Ron Y, Avni-Biron I, Yerushalmy-Feler A, Assa A, Somech R, Bar-Gil Shitrit A. Immune function in newborns with in-utero exposure to anti-TNFα therapy. Front Pediatr 2022; 10:935034. [PMID: 36120653 PMCID: PMC9470929 DOI: 10.3389/fped.2022.935034] [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: 05/03/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND AND AIM Anti-TNFα is measurable in infants exposed in utero up to 12 months of age. Data about the exposure effect on the infant's adaptive immunity are limited. We aimed to prospectively evaluate the distribution and function of T and B cells, in infants of females with inflammatory bowel disease, in utero exposed to anti-TNFα or azathioprine. METHODS A prospective multi-center study conducted 2014-2017. Anti-TNFα levels were measured in cord blood, and at 3 and 12 months. T-cell repertoire and function were analyzed at 3 and 12 months by flow-cytometry, expression of diverse T cell receptors (TCR) and T-cell receptor excision circles (TREC) quantification assay. Serum immunoglobulins and antibodies for inactivated vaccines were measured at 12 months. Baseline clinical data were retrieved, and 2-monthly telephonic interviews were performed regarding child infections and growth. RESULTS 24 pregnant females, age 30.6 (IQR 26.5-34.5) years were recruited, 20 with anti-TNFα (infliximab 8, adalimumab 12), and 4 with azathioprine treatment. Cord blood anti-TNFα was higher than maternal blood levels [4.3 (IQR 2.3-9.2) vs. 2.5 (IQR 1.3-9.7) mcg/ml], declining at 3 and 12 months. All infants had normal number of B-cells (n = 17), adequate levels of immunoglobulins (n = 14), and protecting antibody levels to Tetanus, Diphtheria, Hemophilus influenza-B and hepatitis B (n = 17). All had normal CD4+, CD8+ T-cells, and TREC numbers. TCR repertoire was polyclonal in 18/20 and slightly skewed in 2/20 infants. No serious infections requiring hospitalization were recorded. CONCLUSION We found that T-cell and B-cell immunity is fully mature and immune function is normal in infants exposed in utero to anti-TNFα, as in those exposed to azathioprine. Untreated controls and large-scale studies are needed to confirm these results.
Collapse
Affiliation(s)
- Batia Weiss
- Division of Pediatric Gastroenterology and Nutrition, Edmond and Lily Safra Children's Hospital, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shomron Ben-Horin
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Chaim Sheba Medical Center, Institute of Gastroenterology, Ramat Gan, Israel
| | - Atar Lev
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Pediatric Immunology Laboratory, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Efrat Broide
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Division of Pediatric Gastroenterology, Assaf-Harofe Medical Center, Ramat Gan, Israel
| | - Miri Yavzori
- Chaim Sheba Medical Center, Institute of Gastroenterology, Ramat Gan, Israel
| | - Adi Lahat
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Chaim Sheba Medical Center, Institute of Gastroenterology, Ramat Gan, Israel
| | - Uri Kopylov
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Chaim Sheba Medical Center, Institute of Gastroenterology, Ramat Gan, Israel
| | - Orit Picard
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Chaim Sheba Medical Center, Institute of Gastroenterology, Ramat Gan, Israel
| | - Rami Eliakim
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Chaim Sheba Medical Center, Institute of Gastroenterology, Ramat Gan, Israel
| | - Yulia Ron
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Souraski Medical Center, Institute of Gastroenterology, Tel Aviv, Israel
| | - Irit Avni-Biron
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel
| | - Anat Yerushalmy-Feler
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Division of Pediatric Gastroenterology, Souraski Medical Center, Dana Duek Children's Hospital, Tel Aviv, Israel
| | - Amit Assa
- Schneider Children's Hospital, Institute of Pediatric Gastroenterology, Hepatology and Nutrition, Petah Tikva, Israel.,The Juliet Keidan Institute of Pediatric Gastroenterology and Nutrition, Shaare Zedek Medical Center, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Raz Somech
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Pediatric Immunology Laboratory, Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Pediatrics A, Edmond and Lily Safra Children's Hospital, Ramat Gan, Israel
| | - Ariella Bar-Gil Shitrit
- The Juliet Keidan Institute of Pediatric Gastroenterology and Nutrition, Shaare Zedek Medical Center, The Hebrew University of Jerusalem, Jerusalem, Israel.,Medical School, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
11
|
Khalturina EO, Degtyareva ND, Bairashevskaia AV, Mulenkova AV, Degtyareva AV. Modern diagnostic capabilities of neonatal screening for primary immunodeficiencies in newborns. Clin Exp Pediatr 2021; 64:504-510. [PMID: 33781055 PMCID: PMC8498015 DOI: 10.3345/cep.2020.01270] [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: 07/19/2020] [Accepted: 02/05/2021] [Indexed: 11/27/2022] Open
Abstract
Population screening of newborns is an extremely important and informative diagnostic approach that allows early identification of babies who are predisposed to the development of a number of serious diseases. Some of these diseases are known and have effective treatment methods. Neonatal screening enables the early diagnosis and subsequent timely initiation of therapy. This helps to prevent serious complications and reduce the percentage of disability and deaths among newborns and young children. Primary immunodeficiency diseases and primary immunodeficiency syndrome (PIDS) are a heterogeneous group of diseases and conditions based on impaired immune system function associated with developmental defects and characterized by various combinations of recurrent infections, development of autoimmune and lymphoproliferative syndromes (genetic defects in apoptosis, gene mutation Fas receptor or ligand), granulomatous process, and malignant neoplasms. Most of these diseases manifest in infancy and lead to serious illness, disability, and high mortality rates. Until recently, it was impossible to identify children with PIDS before the onset of the first clinical symptoms, which are usually accompanied by complications in the form of severe coinfections of a viral-bacterial-fungal etiology. Modern advances in medical laboratory technology have allowed the identification of children with severe PIDS, manifested by T- and/or B-cell lymphopenia and other disorders of the immune system. This review discusses the main existing strategies and directions used in PIDS screening programs for newborns, including approaches to screening based on excision of T-cell receptors and kappa-recombination excision circles, as well as the potential role and place of next-generation sequencing technology to increase the diagnostic accuracy of these diseases.
Collapse
Affiliation(s)
- Evgenia Olegovna Khalturina
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia.,National Medical Research Center for Obstetrics, Gynecology, and Perinatology named after Academician V.I. Kulakov of the Ministry of Health of the Russian Federation; Department of Pediatrics and Neonatology, Moscow, Russia
| | - Natalia Dmitrievna Degtyareva
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Anastasiia Vasi'evna Bairashevskaia
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Alena Valerievna Mulenkova
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Anna Vladimirovna Degtyareva
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia.,National Medical Research Center for Obstetrics, Gynecology, and Perinatology named after Academician V.I. Kulakov of the Ministry of Health of the Russian Federation; Department of Pediatrics and Neonatology, Moscow, Russia
| |
Collapse
|
12
|
Exploring genetic defects in adults who were clinically diagnosed as severe combined immune deficiency during infancy. Immunol Res 2021; 69:145-152. [PMID: 33599911 DOI: 10.1007/s12026-021-09179-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/03/2021] [Indexed: 12/16/2022]
Abstract
Genetic diagnostic tools including whole-exome sequencing (WES) have advanced our understanding in human diseases and become common practice in diagnosing patients with suspected primary immune deficiencies. Establishing a genetic diagnosis is of paramount importance for tailoring adequate therapeutic regimens, including identifying the need for hematopoietic stem cell transplantation (HSCT) and genetic-based therapies. Here, we genetically studied two adult patients who were clinically diagnosed during infancy with severe combined immune deficiency (SCID). Two unrelated patients, both of consanguineous kindred, underwent WES in adulthood, 2 decades after their initial clinical manifestations. Upon clinical presentation, immunological workup was performed, which led to a diagnosis of SCID. The patients presented during infancy with failure to thrive, generalized erythematous rash, and recurrent gastrointestinal and respiratory tract infections, including episodes of Pneumocystis pneumonia infection and Candida albicans fungemia. Hypogammaglobulinemia and T-cell lymphopenia were detected. Both patients were treated with a 10/10 HLA matched sibling donor unconditioned HSCT. Retrospective genetic workup revealed homozygous bi-allelic mutations in IL7RA in one patient and in RAG2 in the other. Our study exemplifies the impact of retrospectively establishing a genetic diagnosis. Pinpointing the genetic cause raises several issues including optimized surveillance and treatment, understanding disease mechanisms and outcomes, future family planning, and social and psychological considerations.
Collapse
|
13
|
Cao M, Ruan L, Huang Y, Wang J, Yan J, Sang Y, Li S, Wang G, Wu X. Premature CD4 + T Cells Senescence Induced by Chronic Infection in Patients with Acute Coronary Syndrome. Aging Dis 2020; 11:1471-1480. [PMID: 33269101 PMCID: PMC7673853 DOI: 10.14336/ad.2020.0203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 02/03/2020] [Indexed: 01/10/2023] Open
Abstract
Acquired immune responses mediated by CD4+ T cells contribute to the initiation and progression of acute coronary syndrome (ACS). ACS patients show acquired immune system abnormalities that resemble the characteristics of autoimmune dysfunction described in the elderly. This study aimed to investigate the role of premature CD4+ T cells senescence in ACS and the underlying mechanism. We compared the immunological status of 25 ACS patients, 15 young healthy individuals (C1), and 20 elderly individuals with absence of ACS (C2). The percentages of CD4+ T lymphocyte subsets (including naïve, regulatory, memory and effector T cells) in peripheral blood were analyzed. In ACS patients, a significant expansion of CD4+CD28null effector T cells and a decline of CD4+CD25+CD62L+Treg cells were observed. In addition, patients with ACS showed an accelerated loss of CD4+CD45RA+CD62L+ naïve T cells and a compensatory increase in the number of CD4+CD45RO+ memory T cells. ACS patients demonstrated no significant difference in frequency of T cell receptor excision circles (TRECs) compared to age-matched healthy volunteers. The expression of p16Ink4a was increased while CD62L was decreased in CD4+CD28null T cells of ACS patients. Compared to healthy donors, ACS patients demonstrated the lowest telomerase activity in both CD4+CD28+and CD4+CD28null T cells. The serum levels of C-reactive protein, Cytomegalovirus IgG, Helicobactor pylori IgG and Chlamydia pneumonia IgG were significantly higher in ACS patients. The results suggested that the percentage of CD4+ T cell subpopulations correlated with chronic infection, which contributes to immunosenescence. In conclusion, chronic infection induced senescence of premature CD4+T cells, which may be responsible for the development of ACS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiaofen Wu
- Department of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
14
|
Gomes JAN, da Silva Dias GA, Fujihara S, Yoshikawa GT, Koyama RVL, Sousa RCM, Quaresma JAS, Fuzii HT. Decrease in naïve T cell production due to HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) development. Immunobiology 2020; 226:152050. [PMID: 33338979 DOI: 10.1016/j.imbio.2020.152050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 10/25/2020] [Accepted: 11/29/2020] [Indexed: 10/22/2022]
Abstract
Human T-lymphocytic virus 1 (HTLV-1) is mainly associated with adult T-cell leukemia/lymphoma (ATLL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Patients with HAM/TSP exhibit significant changes in their immune response, and HTLV-1 infection can interfere in cytokine production and perhaps in T cell production. The aims of this study were to evaluate thymic function in HAM/TSP patients and HTLV-1 healthy carriers (HCs) and correlate it to age and interleukin 7 (IL-7) gene expression. Thymic function in 21 HAM/TSP patients and 12 HCs was evaluated by quantifying T cell receptor rearrangement excision circle (TREC) particles and IL-7 gene expression, both measured by quantitative polymerase chain reaction. HAM/TSP patients presented lower TREC particle counts (p = 0.0112) and lower IL-7 expression (p = 0.0102) than HCs. Both TREC particles and IL-7 gene expression were separately analyzed in two age groups: ≤ 59 years and ≥60 years, The ≤59-year-old HAM/TSP patients had a lower TREC count compared with the ≤59-year-old HCs (p = 0.0476). In conclusion, HAM/TSP development could interfere with thymic function because the results showed TREC particle reduction in HAM/TSP patients in relation to HCs, and it could be associated with a concomitant reduction in IL-7 expression.
Collapse
Affiliation(s)
| | | | - Satomi Fujihara
- Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém-Pará, Brazil
| | | | | | | | | | - Hellen Thais Fuzii
- Núcleo de Medicina Tropical, Universidade Federal do Pará, Belém-Pará, Brazil
| |
Collapse
|
15
|
Sommer N, Noack S, Hecker A, Hackstein H, Bein G, Weissmann N, Seeger W, Mayer K, Hecker M. Decreased Thymic Output Contributes to Immune Defects in Septic Patients. J Clin Med 2020; 9:jcm9092695. [PMID: 32825352 PMCID: PMC7565146 DOI: 10.3390/jcm9092695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/02/2020] [Accepted: 08/12/2020] [Indexed: 12/29/2022] Open
Abstract
Background: Prolonged immunosuppression and hypoinflammation, termed compensatory anti-inflammatory response syndrome (CARS), contribute to high morbidity and mortality in the late phase of sepsis. Although apoptosis is a well-known cause of lymphopenia in sepsis, the contribution of thymic output to immune alterations in sepsis and potential compensatory mechanisms are largely unknown. Methods: We investigate the release of CD4+ T cells from the thymus and their peripheral proliferation by evaluating T-cell receptor excision circles (TREC) and the expression of CD31 as markers for recent thymic emigrants (RTE) and their proliferative offspring in septic patients with relevant lymphopenia in the CARS phase. Moreover, we determine the aging of T cells by measuring telomere characteristics. Results: In septic patients, we found decreased CD4+ T-helper cell numbers, while CD8+ T cell numbers were unchanged. As a possible cause, we detected increased apoptosis of CD4+ T-helper cells and decreased levels of IL-7, which promotes the maturation of T cells in the thymus. Accordingly, the relative number of mature CD4+ T cells, TREC-containing CD4+ T cells, and CD31+ RTEs (characteristic of thymic output) was decreased, while the relative number of CD31-T cells (peripherally expanded naïve T cells) was increased. Furthermore, the telomere length decreased, although telomerase activity and markers for the shelterin complex were increased specifically in CD4+ but not in CD8+ T cells. Conclusion: We thus conclude that, in addition to T-cell apoptosis, decreased thymic output and increased aging of CD4+ T cells may contribute to lymphopenia and immunosuppression in sepsis. Increased proliferation of peripheral T cells cannot compensate for these effects.
Collapse
Affiliation(s)
- Natascha Sommer
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (N.S.); (S.N.); (N.W.); (W.S.); (K.M.)
| | - Steffen Noack
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (N.S.); (S.N.); (N.W.); (W.S.); (K.M.)
| | - Andreas Hecker
- Department of General and Thoracic Surgery, Justus-Liebig-University of Giessen, 35392 Giessen, Germany;
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, 91054 Erlangen, Germany;
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University, 35392 Giessen, Germany;
| | - Gregor Bein
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University, 35392 Giessen, Germany;
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (N.S.); (S.N.); (N.W.); (W.S.); (K.M.)
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (N.S.); (S.N.); (N.W.); (W.S.); (K.M.)
| | - Konstantin Mayer
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (N.S.); (S.N.); (N.W.); (W.S.); (K.M.)
- Department of Pulmonary and Sleep Medicine, ViDia hospitals, 76137 Karlsruhe, Germany
| | - Matthias Hecker
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (N.S.); (S.N.); (N.W.); (W.S.); (K.M.)
- Correspondence: ; Tel.: +49-641-985-56755; Fax: +49-641-985-51259
| |
Collapse
|
16
|
Halouani A, Jmii H, Bodart G, Michaux H, Renard C, Martens H, Aouni M, Hober D, Geenen V, Jaïdane H. Assessment of Thymic Output Dynamics After in utero Infection of Mice With Coxsackievirus B4. Front Immunol 2020; 11:481. [PMID: 32300341 PMCID: PMC7142265 DOI: 10.3389/fimmu.2020.00481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 03/02/2020] [Indexed: 01/01/2023] Open
Abstract
The thymus is the main organ of the lymphatic system, in which T cells undergo a rigorous selection to ensure that their receptors (TCRs) will be functional and will not react against the self. Genes encoding for TCR chains are fragmented and must be rearranged by a process of somatic recombination generating TCR rearrangement excision circles (TRECs). We recently documented coxsackievirus B4 (CV-B4) infection of Swiss albino mouse thymus in the course of in utero transmission. In the current study, we intended to evaluate thymic output in this experimental model. For this purpose, pregnant Swiss albino mice were inoculated with CV-B4 at day 10 or 17 of gestation, and thymus and spleen were sampled from offspring at different time points and then subjected to quantification of TREC molecules and Ptk7 gene expression. Results showed a pronounced effect of in utero CV-B4 infection on the thymus with an increase in the cellularity and, consequently, the weight of the organ. sj and DβTREC analysis, by real-time PCR, revealed a significant decrease following CV-B4 infection compared to controls, a decrease which gets worse as time goes by, both in the thymus and in the periphery. Those observations reflect a disturbance in the export of T cells to the periphery and their accumulation within the thymus. The evaluation of Ptk7 transcripts in the thymus, for its part, showed a decrease in expression, especially following an infection at day 10 of gestation, which supports the hypothesis of T cell accumulation in a mature stage in the thymus. The various effects observed correlate either negatively or positively with the viral load in the thymus and spleen. Disruption in thymic export may indeed interfere with T cell maturation. We speculate that this may lead to a premature release of T cells and the possibility of circulating autoreactive or proliferation-impaired T cell clones.
Collapse
Affiliation(s)
- Aymen Halouani
- Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir, Tunisia.,Faculté des Sciences de Tunis, Université de Tunis El Manar, Tunis, Tunisia.,GIGA-I3 Neuroimmunoendocrinology, GIGA Research Institute, University of Liège, Liège, Belgium
| | - Habib Jmii
- Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir, Tunisia.,Faculté des Sciences de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Gwennaëlle Bodart
- GIGA-I3 Neuroimmunoendocrinology, GIGA Research Institute, University of Liège, Liège, Belgium
| | - Hélène Michaux
- GIGA-I3 Neuroimmunoendocrinology, GIGA Research Institute, University of Liège, Liège, Belgium
| | - Chantal Renard
- GIGA-I3 Neuroimmunoendocrinology, GIGA Research Institute, University of Liège, Liège, Belgium
| | - Henri Martens
- GIGA-I3 Neuroimmunoendocrinology, GIGA Research Institute, University of Liège, Liège, Belgium
| | - Mahjoub Aouni
- Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir, Tunisia
| | - Didier Hober
- Université de Lille, CHU de Lille, Laboratoire de Virologie, Lille, France
| | - Vincent Geenen
- GIGA-I3 Neuroimmunoendocrinology, GIGA Research Institute, University of Liège, Liège, Belgium
| | - Hela Jaïdane
- Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir, Tunisia
| |
Collapse
|
17
|
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is the effective mean of immune restoration in severe combined immunodefiency (SCID). Usually, HSCT without cytoreductive conditioning is attempted. Nevertheless, conditioning procedures are still preferred in a subset of patients. Herein, we describe the immunological outcome in a cohort of conditioned and unconditioned patients, from diagnosis, through transplantation, to follow-up. This retrospective study was conducted on 17 patients with SCID (10 conditioned, 7 unconditioned) who later underwent HSCT. Immune reconstitution was assessed in the post-transplant year by quantification of T cell receptor excision circles (TRECs) and kappa-deleting recombination excision circles (KRECs), among additional laboratory and clinical evaluations. Unconditioned patients were diagnosed and transplanted earlier. TREC and KREC quantification showed a gradual increase in both groups, with higher levels in the conditioned group. Engraftment percentages differed drastically between groups, favoring the conditioned group. Unconditioned patients were significantly more dependent on intravenous immunoglobulins (IVIGs). One patient from each group succumbed to disease complications. Conditioning demonstrated superior laboratorial outcomes. Patients with unique characteristics (i.e., consanguinity, Bacillus Calmette-Guérin vaccination, impaired access to IVIG) may require personalized considerations. The effort to implement secondary prevention of SCID with newborn screening should continue.
Collapse
|
18
|
Liao CY, Yu HW, Cheng CN, Chen JS, Lin CW, Chen PC, Shieh CC. A novel pathogenic mutation on Interleukin-7 receptor leading to severe combined immunodeficiency identified with newborn screening and whole exome sequencing. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2020; 53:99-105. [DOI: 10.1016/j.jmii.2018.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/25/2017] [Accepted: 02/05/2018] [Indexed: 01/08/2023]
|
19
|
Marcus N, Stauber T, Lev A, Simon AJ, Stein J, Broides A, Somekh I, Almashanu S, Somech R. MHC II deficient infant identified by newborn screening program for SCID. Immunol Res 2019; 66:537-542. [PMID: 30084052 DOI: 10.1007/s12026-018-9019-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Newborn screening (NBS) programs for severe combined immunodeficiency (SCID), using the TREC-based assay, have enabled early diagnosis, prompt treatment, and eventually changed the natural history of affected infants. Nevertheless, it was believed that some affected infants with residual T cell, such as patients with MHC II deficiency, will be misdiagnosed by this assay. A full immune workup and genetic analysis using direct Sanger sequencing and whole exome sequencing have been performed to a patient that was identified by the Israeli NBS program for SCID. The patient was found to have severe CD4 lymphopenia with an inverted CD4/CD8 ratio, low TREC levels in peripheral blood, abnormal response to mitogen stimulation, and a skewed T cell receptor repertoire. HLA-DR expression on peripheral blood lymphocytes was undetectable suggesting a diagnosis of MHC II deficiency. Direct sequencing of the RFX5 gene revealed a stop codon change (p. R239X, c. C715T), which could cause the patient's immune phenotype. His parents were found to be heterozygote carriers for the mutation. Whole exome sequencing could not identify other potential mutations to explain his immunodeficiency. The patient underwent successful conditioned hematopoietic stem cell transplantation from healthy matched unrelated donor and is currently well and alive with full chimerism. Infants with MHC class II deficiency can potentially be identified by the TREC-based assay NBS for SCID. Therefore, MHC II molecules (e.g., HLA-DR) measurement should be part of the confirmatory immune-phenotyping for patients with positive screening results. This will make the diagnosis of such patients straightforward.
Collapse
Affiliation(s)
- Nufar Marcus
- Allergy and Immunology Unit, Felsenstein Medical Research Center, Kipper Institute of Immunology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tali Stauber
- Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation Center, 52621, Tel Hashomer, Israel
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 52621, Tel Hashomer, Israel
| | - Atar Lev
- Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation Center, 52621, Tel Hashomer, Israel
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 52621, Tel Hashomer, Israel
| | - Amos J Simon
- Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation Center, 52621, Tel Hashomer, Israel
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 52621, Tel Hashomer, Israel
| | - Jerry Stein
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department for Hemato-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Arnon Broides
- Pediatric Immunology Clinic, Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ido Somekh
- Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Shlomo Almashanu
- The National Center for Newborn Screening, Ministry of Health, 52621, Tel HaShomer, Israel
| | - Raz Somech
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation Center, 52621, Tel Hashomer, Israel.
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 52621, Tel Hashomer, Israel.
- The National Lab for Confirming Primary Immunodeficiency in Newborn Screening Center for Newborn Screening, Ministry of Health, Tel HaShomer, Israel.
| |
Collapse
|
20
|
Bischof J, Gärtner F, Zeiser K, Kunz R, Schreiner C, Hoffer E, Burster T, Knippschild U, Zimecki M. Immune Cells and Immunosenescence. Folia Biol (Praha) 2019; 65:53-63. [PMID: 31464181 DOI: 10.14712/fb2019065020053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Aging is associated with progressive loss of physiological integrity, leading to impaired physical and mental functions as well as increased morbidity and mortality. With advancing age, the immune system is no longer able to adequately control autoimmunity, infections, or cancer. The abilities of the elderly to slow down undesirable effects of aging may depend on the genetic background, lifestyle, geographic region, and other presently unknown factors. Although most aspects of the immunity are constantly declining in relation to age, some features are retained, while e.g. the ability to produce high levels of cytokines, response to pathogens by increased inflammation, and imbalanced proteolytic activity are found in the elderly, and might eventually cause harm. In this context, it is important to differentiate between the effect of immunosenescence that is contributing to this decline and adaptations of the immune system that can be quickly reversed if necessary.
Collapse
Affiliation(s)
- J Bischof
- Department of General Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm, Germany
| | - F Gärtner
- Department of General Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm, Germany
| | - K Zeiser
- Department of General Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm, Germany
| | - R Kunz
- Department of General Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm, Germany
| | - C Schreiner
- Department of General Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm, Germany
| | - E Hoffer
- Department of General Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm, Germany
| | - T Burster
- Department of Biology, School of Science and Technology, Nazarbayev University, Astana, Kazakhstan Republic
| | - U Knippschild
- Department of General Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm, Germany
| | - M Zimecki
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
21
|
Somekh I, Marquardt B, Liu Y, Rohlfs M, Hollizeck S, Karakukcu M, Unal E, Yilmaz E, Patiroglu T, Cansever M, Frizinsky S, Vishnvenska-Dai V, Rechavi E, Stauber T, Simon AJ, Lev A, Klein C, Kotlarz D, Somech R. Novel Mutations in RASGRP1 are Associated with Immunodeficiency, Immune Dysregulation, and EBV-Induced Lymphoma. J Clin Immunol 2018; 38:699-710. [DOI: 10.1007/s10875-018-0533-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/09/2018] [Indexed: 12/25/2022]
|
22
|
Wilson K, Duque DR, Murphy MS, Hawken S, Pham-Huy A, Kwong J, Deeks SL, Potter BK, Crowcroft NS, Bulman DE, Chakraborty P, Little J. T-cell receptor excision circle levels and safety of paediatric immunization: A population-based self-controlled case series analysis. Hum Vaccin Immunother 2018; 14:1378-1391. [PMID: 29420131 PMCID: PMC6037463 DOI: 10.1080/21645515.2018.1433971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/09/2018] [Accepted: 01/20/2018] [Indexed: 12/21/2022] Open
Abstract
T-cell receptor excision circle levels are a surrogate marker of T-cell production and immune system function. We sought to determine whether non-pathological levels of infant T-cell receptor excision circles were associated with adverse events following immunization. A self-controlled case series design was applied on a sample of 231,693 children who completed newborn screening for severe combined immunodeficiency in Ontario, Canada between August 2013 and December 2015. Exposures included routinely administered pediatric vaccines up to 15 months of age. Main outcomes were combined health services utilization for recognized adverse events following immunization. 1,406,981 vaccination events were included in the final dataset. 103,007 children received the Pneu-C-13 or Men-C-C vaccine and 97,998 received the MMR vaccine at 12 months of age. 67,725 children received the varicella immunization at 15 months. Our analysis identified no association between newborn T-cell receptor excision circle levels and subsequent health services utilization events following DTa-IPV-Hib, Pneu-C-13, and Men-C-C vaccinations at 2-month (RI 0.94[95%CI 0.87-1.02]), 4-month (RI 0.82[95%CI 0.75-0.9]), 6-month (RI 0.63[95%CI 0.57-0.7]) and 12-month (RI 0.49[95%CI 0.44-0.55]). We also found no trends in health services utilization following MMR (RI 1.43[95%1.34-1.52]) or varicella (RI 1.14[95%CI 1.05-1.23]) vaccination. Our findings provide further support for the safety of pediatric vaccinations.
Collapse
Affiliation(s)
- Kumanan Wilson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | | | - Malia S.Q Murphy
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Steven Hawken
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - Anne Pham-Huy
- Department of Pediatrics, University of Ottawa, Ottawa, Canada
| | - Jeffrey Kwong
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Shelley L. Deeks
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Beth K. Potter
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | | | | | - Pranesh Chakraborty
- Department of Pediatrics, University of Ottawa, Ottawa, Canada
- Newborn Screening Ontario, Children's Hospital of Eastern Ontario, Ottawa, Canada
| | - Julian Little
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| |
Collapse
|
23
|
Mannan T, Ahmed S, Akhtar E, Ahsan KB, Haq A, Kippler M, Vahter M, Raqib R. Associations of Arsenic Exposure With Telomere Length and Naïve T Cells in Childhood—A Birth Cohort Study. Toxicol Sci 2018; 164:539-549. [DOI: 10.1093/toxsci/kfy105] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Tania Mannan
- Infectious Diseases Division, icddr,b, Dhaka 1212, Bangladesh
- Department of Immunology, Bangladesh University of Health Sciences, Dhaka 1216, Bangladesh
| | - Sultan Ahmed
- Infectious Diseases Division, icddr,b, Dhaka 1212, Bangladesh
| | - Evana Akhtar
- Infectious Diseases Division, icddr,b, Dhaka 1212, Bangladesh
| | | | - Ahsanul Haq
- Infectious Diseases Division, icddr,b, Dhaka 1212, Bangladesh
| | - Maria Kippler
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE 171 77, Sweden
| | - Marie Vahter
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE 171 77, Sweden
| | - Rubhana Raqib
- Infectious Diseases Division, icddr,b, Dhaka 1212, Bangladesh
| |
Collapse
|
24
|
Woroniecka KI, Rhodin KE, Chongsathidkiet P, Keith KA, Fecci PE. T-cell Dysfunction in Glioblastoma: Applying a New Framework. Clin Cancer Res 2018; 24:3792-3802. [PMID: 29593027 DOI: 10.1158/1078-0432.ccr-18-0047] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/01/2018] [Accepted: 03/26/2018] [Indexed: 02/06/2023]
Abstract
A functional, replete T-cell repertoire is an integral component to adequate immune surveillance and to the initiation and maintenance of productive antitumor immune responses. Glioblastoma (GBM), however, is particularly adept at sabotaging antitumor immunity, eliciting severe T-cell dysfunction that is both qualitative and quantitative. Understanding and countering such dysfunction are among the keys to harnessing the otherwise stark potential of anticancer immune-based therapies. Although T-cell dysfunction in GBM has been long described, newer immunologic frameworks now exist for reclassifying T-cell deficits in a manner that better permits their study and reversal. Herein, we divide and discuss the various T-cell deficits elicited by GBM within the context of the five relevant categories: senescence, tolerance, anergy, exhaustion, and ignorance. Categorization is appropriately made according to the molecular bases of dysfunction. Likewise, we review the mechanisms by which GBM elicits each mode of T-cell dysfunction and discuss the emerging immunotherapeutic strategies designed to overcome them. Clin Cancer Res; 24(16); 3792-802. ©2018 AACR.
Collapse
Affiliation(s)
- Karolina I Woroniecka
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Kristen E Rhodin
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Pakawat Chongsathidkiet
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Kristin A Keith
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Peter E Fecci
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina. .,Department of Pathology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
25
|
The influence and impact of ageing and immunosenescence (ISC) on adaptive immunity during multiple sclerosis (MS) and the animal counterpart experimental autoimmune encephalomyelitis (EAE). Ageing Res Rev 2018; 41:64-81. [PMID: 29101043 DOI: 10.1016/j.arr.2017.10.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 12/21/2022]
Abstract
The human ageing process encompasses mechanisms that effect a decline in homeostasis with increased susceptibility to disease and the development of chronic life-threatening illness. Increasing age affects the immune system which undergoes a progressive loss of efficiency, termed immunosenescence (ISC), to impact on quantitative and functional aspects of innate and adaptive immunity. The human demyelinating disease multiple sclerosis (MS) and the corresponding animal model experimental autoimmune encephalomyelitis (EAE) are strongly governed by immunological events that primarily involve the adaptive arm of the immune response. MS and EAE are frequently characterised by a chronic pathology and a protracted disease course which thereby creates the potential for exposure to the inherent, on-going effects and consequences of ISC. Collective evidence is presented to confirm the occurrence of established and unendorsed biological markers of ISC during the development of both diseases. Moreover, results are discussed from studies during the course of MS and EAE that reveal a premature upregulation of ISC-related biomarkers which indicates untimely alterations to the adaptive immune system. The effects of ISC and a prematurely aged immune system on autoimmune-associated neurodegenerative conditions such as MS and EAE are largely unknown but current evaluation of data justifies and encourages further investigation.
Collapse
|
26
|
Levy-Mendelovich S, Lev A, Aviner S, Rosenberg N, Kaplinsky C, Sharon N, Miskin H, Dvir A, Kenet G, Schushan IE, Somech R. Quantification of specific T and B cells immunological markers in children with chronic and transient ITP. Pediatr Blood Cancer 2017; 64. [PMID: 28544224 DOI: 10.1002/pbc.26646] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/25/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Immune thrombocytopenic purpura (ITP) is characterized by a transient (nonchronic) or permanent (chronic) decline in the number of platelets. Predicting the course of ITP, at the time of diagnosis, is of importance. Here we studied at diagnosis, clinical and immunological parameters in order to distinguish between different courses. The latter included the measure of new B and T cells using quantification of kappa-deleting recombination excision circles (KRECs) and T-cell receptor excision circles (TRECs), respectively. METHODS Blood samples were collected from 44 children with a clinical diagnosis of ITP. Real-time PCR was performed in order to quantify the number of copies of TREC and KREC followed by collection of clinical data from medical files. The children were retrospectively divided into two groups: chronic and nonchronic. RESULTS Twenty-four patients (54%) were classified as nonchronic ITP and 20 patients (46%) were classified as chronic ITP. We confirmed some clinical parameters (e.g., gender, age) but not others (e.g., preceding infection, level of thrombocytopenia) that distinguish patients with chronic and nonchronic course. While KREC quantification was similar in patients regardless the outcome of their disease, it was significantly higher than the level of controls (P < 0.05). TREC quantification was not different between patients and controls. CONCLUSIONS KREC but not TREC levels are different in patients comparing to controls, pointing to an overreaction of B-cell development as a role in the pathogenesis of ITP. These results may shed more lights on the immune mechanism of ITP.
Collapse
Affiliation(s)
- Sarina Levy-Mendelovich
- Pediatric Department A and the Immunology Services, "Edmond and Lily Safra" Children's Hospital, Jeffrey Modell Foundation Center, Sheba Medical Center, Tel Hashomer affiliated with Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Pediatric Hemato-Oncology Department, "Edmond and Lily Safra" Children's Hospital, Sheba Medical Center, Tel Hashomer affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,National Hemphilia and Thrombosis Institute, Sheba Medical Center, Tel Hashomer affiliated to Sackler School of Medicine, Tel Aviv University, Israel
| | - Atar Lev
- Pediatric Department A and the Immunology Services, "Edmond and Lily Safra" Children's Hospital, Jeffrey Modell Foundation Center, Sheba Medical Center, Tel Hashomer affiliated with Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shraga Aviner
- Department of Pediatrics, Barzilai University Medical Center, Ashkelon affiliated to Ben Gurion University, Beer-Sheva, Israel
| | - Nurit Rosenberg
- Institute of Thrombosis and Hemostasis, Chaim Sheba Medical Center, Tel Hashomer affiliated to Sackler School of Medicine, Tel Aviv University, Israel
| | - Caim Kaplinsky
- Pediatric Hemato-Oncology Department, "Edmond and Lily Safra" Children's Hospital, Sheba Medical Center, Tel Hashomer affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nechama Sharon
- Pediatric Department, Laniado Hospital, Netanya affiliated to Sackler School of Medicine, Tel Aviv University, Israel
| | - Hagit Miskin
- Pediatric Hematoloy Unit, Shaare Zedek Medical Center, Jerusalem affiliated to Hebrew University School of Medicine, Jerusalem, Israel
| | - Aviya Dvir
- Institute of Thrombosis and Hemostasis, Chaim Sheba Medical Center, Tel Hashomer affiliated to Sackler School of Medicine, Tel Aviv University, Israel
| | - Gili Kenet
- National Hemphilia and Thrombosis Institute, Sheba Medical Center, Tel Hashomer affiliated to Sackler School of Medicine, Tel Aviv University, Israel
| | - Irit Eisen Schushan
- Neonatology Department, "Edmond and Lily Safra" Children's Hospital, Sheba Medical Center, Tel Hashomer affiliated to Sackler School of Medicine, Tel Aviv University, Israel
| | - Raz Somech
- Pediatric Department A and the Immunology Services, "Edmond and Lily Safra" Children's Hospital, Jeffrey Modell Foundation Center, Sheba Medical Center, Tel Hashomer affiliated with Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
27
|
Cho S, Seo HJ, Lee JH, Kim MY, Lee SD. Influence of immunologic status on age prediction using signal joint T cell receptor excision circles. Int J Legal Med 2017; 131:1061-1067. [DOI: 10.1007/s00414-017-1540-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/19/2017] [Indexed: 10/20/2022]
|
28
|
Matteucci C, Grelli S, Balestrieri E, Minutolo A, Argaw-Denboba A, Macchi B, Sinibaldi-Vallebona P, Perno CF, Mastino A, Garaci E. Thymosin alpha 1 and HIV-1: recent advances and future perspectives. Future Microbiol 2017; 12:141-155. [PMID: 28106477 DOI: 10.2217/fmb-2016-0125] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In spite of the consistent benefits for HIV-1 infected patients undergoing antiretroviral therapy, a complete immune reconstitution is usually not achieved. Actually, antiretroviral therapy may be frequently accompanied by immunological unresponsiveness, persistent inflammatory conditions and inefficient cytotoxic T-cell response. Thymosin alpha 1 is a thymic peptide that demonstrates a peculiar ability to restore immune system homeostasis in different physiological and pathological conditions (i.e., infections, cancer, immunodeficiency, vaccination and aging) acting as multitasking protein depending on the host state of inflammation or immune dysfunction. This review reports the present knowledge on the in vitro and in vivo studies concerning the use of thymosin alpha 1 in HIV-1 infection. Recent findings and future perspectives of therapeutic intervention are discussed.
Collapse
Affiliation(s)
- Claudia Matteucci
- Department of Experimental Medicine & Surgery, University of Rome 'Tor Vergata', Via Montepellier, 1, Rome 00133, Italy
| | - Sandro Grelli
- Department of Experimental Medicine & Surgery, University of Rome 'Tor Vergata', Via Montepellier, 1, Rome 00133, Italy
| | - Emanuela Balestrieri
- Department of Experimental Medicine & Surgery, University of Rome 'Tor Vergata', Via Montepellier, 1, Rome 00133, Italy
| | - Antonella Minutolo
- Department of Experimental Medicine & Surgery, University of Rome 'Tor Vergata', Via Montepellier, 1, Rome 00133, Italy
| | - Ayele Argaw-Denboba
- Department of Experimental Medicine & Surgery, University of Rome 'Tor Vergata', Via Montepellier, 1, Rome 00133, Italy
| | - Beatrice Macchi
- Department of System Medicine, University of Rome 'Tor Vergata', Via Montepellier, 1, Rome 00133, Italy
| | - Paola Sinibaldi-Vallebona
- Department of Experimental Medicine & Surgery, University of Rome 'Tor Vergata', Via Montepellier, 1, Rome 00133, Italy.,Institute of Translational Pharmacology, National Research Council, Via Fosso del Cavaliere, 100, Rome 00133, Italy
| | - Carlo Federico Perno
- Department of Experimental Medicine & Surgery, University of Rome 'Tor Vergata', Via Montepellier, 1, Rome 00133, Italy
| | - Antonio Mastino
- Institute of Translational Pharmacology, National Research Council, Via Fosso del Cavaliere, 100, Rome 00133, Italy.,Department of Chemical, Biological, Pharmaceutical & Environmental Sciences, University of Messina, Via F. Stagno d'Alcontres 31, Messina 98166, Italy
| | - Enrico Garaci
- Department of Experimental Medicine & Surgery, University of Rome 'Tor Vergata', Via Montepellier, 1, Rome 00133, Italy.,IRCSS San Raffaele Pisana, Scientific Institute for Research, Hospitalization & Health Care, Via di Val Cannuta, 247, Roma 00166, Italy
| |
Collapse
|
29
|
Gaballa A, Sundin M, Stikvoort A, Abumaree M, Uzunel M, Sairafi D, Uhlin M. T Cell Receptor Excision Circle (TREC) Monitoring after Allogeneic Stem Cell Transplantation; a Predictive Marker for Complications and Clinical Outcome. Int J Mol Sci 2016; 17:E1705. [PMID: 27727179 PMCID: PMC5085737 DOI: 10.3390/ijms17101705] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 09/26/2016] [Accepted: 09/29/2016] [Indexed: 12/22/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a well-established treatment modality for a variety of malignant diseases as well as for inborn errors of the metabolism or immune system. Regardless of disease origin, good clinical effects are dependent on proper immune reconstitution. T cells are responsible for both the beneficial graft-versus-leukemia (GVL) effect against malignant cells and protection against infections. The immune recovery of T cells relies initially on peripheral expansion of mature cells from the graft and later on the differentiation and maturation from donor-derived hematopoietic stem cells. The formation of new T cells occurs in the thymus and as a byproduct, T cell receptor excision circles (TRECs) are released upon rearrangement of the T cell receptor. Detection of TRECs by PCR is a reliable method for estimating the amount of newly formed T cells in the circulation and, indirectly, for estimating thymic function. Here, we discuss the role of TREC analysis in the prediction of clinical outcome after allogeneic HSCT. Due to the pivotal role of T cell reconstitution we propose that TREC analysis should be included as a key indicator in the post-HSCT follow-up.
Collapse
Affiliation(s)
- Ahmed Gaballa
- Department of Oncology and Pathology, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
| | - Mikael Sundin
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
- Pediatric Blood Disorders, Immunodeficiency and Stem Cell Transplantation, Astrid Lindgren Children's Hospital, Karolinska University Hospital, SE-141 86 Stockholm, Sweden.
| | - Arwen Stikvoort
- Department of Oncology and Pathology, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
| | - Muhamed Abumaree
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, KSA-11461 Riyadh, Saudi Arabia.
| | - Mehmet Uzunel
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, SE-141 86 Stockholm, Sweden.
| | - Darius Sairafi
- Department of Oncology and Pathology, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
| | - Michael Uhlin
- Department of Oncology and Pathology, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, SE-141 86 Stockholm, Sweden.
| |
Collapse
|
30
|
Machnes-Maayan D, Lev A, Katz U, Mishali D, Vardi A, Simon AJ, Somech R. Insight into normal thymic activity by assessment of peripheral blood samples. Immunol Res 2015; 61:198-205. [PMID: 25294167 DOI: 10.1007/s12026-014-8558-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The thymus is a highly specialized organ for T cell receptor (TCR) rearrangement and selection mechanisms that ensure the formation of functional and self-tolerant cells. Little is known about how peripheral blood assessment of thymic function reflects thymus activity during infancy. We compared thymic function-related markers in the thymus with those in peripheral blood in order to check their correlations. We concomitantly blood samples from immunocompetent infants who underwent cardiac surgery that involved thymectomy. The studied thymic markers included TCR excision circles (TRECs), four different TCRD (TCR delta chain) gene rearrangements, the TCR repertoire, regulatory T cells (Tregs, defined as the CD4+CD25+FOXP3+ cell population) and real-time quantitative polymerase chain reaction (RQ-PCR) mRNA expression of forkhead box P3 (FOXP3). Twenty patients were enrolled in this study. Their mean age at the time of the surgery was 3 months/5 days ± 3 months/18 days. There was a significant correlation between thymic and peripheral blood levels of TREC, all four TCRD gene rearrangements and the amount of Tregs. The levels of these parameters were significantly higher in the thymus than those detected in the peripheral blood. The TCR repertoire distribution in both samples was similar. FOXP3 mRNA levels in the thymus and peripheral blood correlated well. Our findings demonstrated a strong and significant correlation between peripheral blood and intra-thymic activity parameters during infancy. Assessment of these parameters in peripheral blood can be used to accurately estimate different intra-thymic capacities for assessing T cell function in health and disease.
Collapse
Affiliation(s)
- Diti Machnes-Maayan
- Pediatric Department B, Pediatric Immunology Service, Jeffrey Modell Foundation (JMF) Center, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, 52621, Tel Hashomer, Israel
| | | | | | | | | | | | | |
Collapse
|
31
|
Disturbed B and T cell homeostasis and neogenesis in patients with ataxia telangiectasia. J Clin Immunol 2014; 34:561-72. [DOI: 10.1007/s10875-014-0044-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 04/09/2014] [Indexed: 12/11/2022]
|
32
|
Lyttle A, Roifman C, Dadi H, Wright N, Kavadas F. Canadian society of allergy and clinical immunology annual scientific meeting 2013. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2014; 10:A1-A72. [PMID: 25748655 PMCID: PMC4126037 DOI: 10.1186/1710-1492-10-s1-a1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Alex Lyttle
- Alberta Children’s Hospital, Calgary, Alberta, Canada
| | - Chaim Roifman
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Harjit Dadi
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nicola Wright
- Alberta Children’s Hospital, Calgary, Alberta, Canada
| | | |
Collapse
|
33
|
Abstract
UNLABELLED Quantification of the T cell receptor excision circles (TRECs) has recently emerged as a useful non-invasive clinical and research tool to investigate thymic activity. It allows the identification of T cell production by the thymus. Quantification of TREC copies has recently been implemented as the preferred test to screen neonates with severe combined immunodeficiency (SCID) or significant lymphopenia. Neonatal genetic screening for SCID is highly important in countries with high rates of consanguinous marriages, such as Israel, and can be used for early diagnosis, enabling prompt therapeutic intervention that will save lives and improve the outcome of these patients. TREC measurement is also applicable in clinical settings where T cell immunity is involved, including any T cell immunodeficiencies, HIV infection, the aging process, autoimmune diseases, and immune reconstitution after bone marrow transplantation. TAKE-HOME MESSAGES Severe combined immunodeficiency, a life-threatening condition, can be detected by neonatal screening.The earlier the detection and the quicker the implementation of appropriate treatment, the greater the likelihood for improved outcome, even cure, for the affected children.TRECs and KRECs quantification are useful screening tests for severe T and B cell immunodeficiency and can be used also to evaluate every medical condition involving T and B cell immunity.
Collapse
Affiliation(s)
- Raz Somech
- Pediatric Department B North, Pediatric Immunology Service, Jeffrey Modell Foundation (JMF) Center, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- To whom correspondence should be addressed. E-mail:
| | - Amos Etzioni
- Meyer Children’s Hospital, Rappaport Faculty of Medicine, The Technion – Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
34
|
Matsui K, Giri N, Alter BP, Pinto LA. Cytokine production by bone marrow mononuclear cells in inherited bone marrow failure syndromes. Br J Haematol 2013; 163:81-92. [PMID: 23889587 PMCID: PMC3930339 DOI: 10.1111/bjh.12475] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 06/21/2013] [Indexed: 12/11/2022]
Abstract
Fanconi anaemia (FA), dyskeratosis congenita (DC), Diamond-Blackfan anaemia (DBA), and Shwachman-Diamond syndrome (SDS) are characterized by the progressive development of bone marrow failure. Overproduction of tumour necrosis factor-α (TNF-α) from activated bone marrow T-cells has been proposed as a mechanism of FA-related aplasia. Whether such overproduction occurs in the other syndromes is unknown. We conducted a comparative study on bone marrow mononuclear cells to examine the cellular subset composition and cytokine production. We found lower proportions of haematopoietic stem cells in FA, DC, and SDS, and a lower proportion of monocytes in FA, DC, and DBA compared with controls. The T- and B-lymphocyte proportions were similar to controls, except for low B-cells in DC. We did not observe overproduction of TNF-α or IFN-γ by T-cells in any patients. Induction levels of TNF-α, interleukin (IL)-6, IL-1β, IL-10, granulocyte colony-stimulating factor, and granulocyte-macrophage colony-stimulating factor in monocytes stimulated with high-dose lipopolysaccharide (LPS) were similar at 4 h but lower at 24 h when compared to controls. Unexpectedly, patient samples showed a trend toward higher cytokine level in response to low-dose (0·001 μg/ml) LPS. Increased sensitivity to LPS may have clinical implications and could contribute to the development of pancytopenia by creating a chronic subclinical inflammatory micro-environment in the bone marrow.
Collapse
Affiliation(s)
- Ken Matsui
- Human Papillomavirus Immunology Laboratory, Science Applications
International Corporation (SAIC)-Frederick, Incorporated, Frederick National
Laboratory for Cancer Research, Frederick, MD 21702
| | - Neelam Giri
- Division of Cancer Epidemiology and Genetics Clinical Genetics
Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD,
20892, United States
| | - Blanche P. Alter
- Division of Cancer Epidemiology and Genetics Clinical Genetics
Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD,
20892, United States
| | - Ligia A. Pinto
- Human Papillomavirus Immunology Laboratory, Science Applications
International Corporation (SAIC)-Frederick, Incorporated, Frederick National
Laboratory for Cancer Research, Frederick, MD 21702
| |
Collapse
|
35
|
Reduced T cell receptor excision circle levels in the colonic mucosa of microscopic colitis patients indicate local proliferation rather than homing of peripheral lymphocytes to the inflamed mucosa. BIOMED RESEARCH INTERNATIONAL 2013; 2013:408638. [PMID: 23956982 PMCID: PMC3727133 DOI: 10.1155/2013/408638] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 06/20/2013] [Accepted: 06/24/2013] [Indexed: 12/15/2022]
Abstract
Dysregulated T cell responses in the intestine may lead to chronic bowel inflammation such as collagenous colitis (CC) and lymphocytic colitis (LC), together known as microscopic colitis (MC). Having demonstrated increased local T cell responses in the intestinal mucosa of MC patients, we investigated the recent thymic emigrants by measuring T cell receptor excision circle (TREC) levels in the colonic biopsies from CC (n = 8), LC (n = 5), and CC or LC patients in histopathological remission (CC-HR, n = 3) (LC-HR, n = 6), non-inflamed diarrhoea patients (n = 17), and controls (n = 10) by real-time PCR. We observed lower median TREC levels in both CC and LC patients as well as in LC-HR patients compared to controls. In contrast to MC patients, non-inflamed diarrhoea patients presented with enhanced TREC levels compared to controls. None of the recorded differences did, however, reach statistical significance. A trend towards increased relative expression of CD3 was noted in all MC subgroups examined and reached statistical significance in LC patients compared to controls. In conclusion, reduced TRECs level in the colonic mucosa, together with our previously demonstrated enhanced expression of Ki67+ T cells, suggests local expansion of resident T lymphocytes in the inflamed mucosa of MC patients.
Collapse
|
36
|
Ganaiem H, Eisenstein EM, Tenenbaum A, Somech R, Simanovsky N, Roscioli T, Weintraub M, Stepensky P. The role of hematopoietic stem cell transplantation in SP110 associated veno-occlusive disease with immunodeficiency syndrome. Pediatr Allergy Immunol 2013; 24:250-6. [PMID: 23448538 DOI: 10.1111/pai.12051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/07/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUND Veno-occlusive disease with immunodeficiency (VODI) is an autosomal recessive disorder of combined immunodeficiency (CID) and hepatic injury. Hematopoietic stem cell transplantation (HSCT) - the only definitive treatment for CID - appeared to have a high rate of complications in a previous report. In this study, we describe a new group of patients with VODI highlighting further clinical and immunologic aspects of this disease and re-evaluating the effectiveness of HSCT for the treatment of this disorder. PATIENTS AND METHODS Review of clinical data, immunologic features, molecular studies, treatment, and final outcome of eight kindred members with VODI. RESULTS The patients described had clinical and immunologic findings consistent with VODI. The molecular studies revealed a new mutation in the SP110 gene. HSCT was carried out in five patients and was successful in three. CONCLUSIONS The diagnosis of VODI should be considered in all patients regardless of ethnicity with a severe combined immunodeficiency (SCID)-like presentation, especially with a normal mitogen response, or with signs of hepatic injury. VODI is a primary immune deficiency, which can be successfully corrected by bone marrow transplantation if applied early in the course of disease using appropriate conditioning.
Collapse
Affiliation(s)
- Hammam Ganaiem
- Department of Pediatrics, Hadassah University Hospital, Jerusalem, Israel
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Pace MJ, Graf EH, O'Doherty U. HIV 2-long terminal repeat circular DNA is stable in primary CD4+T Cells. Virology 2013; 441:18-21. [PMID: 23537959 DOI: 10.1016/j.virol.2013.02.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 02/06/2013] [Accepted: 02/28/2013] [Indexed: 12/20/2022]
Abstract
Treatment resistant latent reservoirs remain a barrier to cure HIV, but the maintenance and properties of these reservoirs are not completely understood. 2-LTR circular HIV DNA has been used to assess ongoing viral replication in HAART treated patients. However, the half-life of this DNA form is still debated with conflicting in vivo and in vitro data. Prior in vitro studies have focused on cell lines or short lived activated cells in cultures of brief duration, while in vivo studies have the added complications of cell migration, division, and death. Therefore, we monitored the stability of 2-LTR circles in primary CD4+T cells in a month long culture and compared it to the stability of integrated HIV DNA and T cell receptor excision circles (TRECs), another circular DNA form that is thought to be stable. We found that 2-LTRs, along with TRECs, were stable, suggesting 2-LTRs do not necessarily indicate ongoing replication.
Collapse
Affiliation(s)
- Matthew J Pace
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 422 Curie Blvd, 702 Stellar Chance,. Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
38
|
Lev A, Simon AJ, Broides A, Levi J, Garty BZ, Rosenthal E, Amariglio N, Rechavi G, Somech R. Thymic function in MHC class II–deficient patients. J Allergy Clin Immunol 2013; 131:831-9. [DOI: 10.1016/j.jaci.2012.10.040] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 10/15/2012] [Accepted: 10/18/2012] [Indexed: 11/25/2022]
|
39
|
Current World Literature. Curr Opin Allergy Clin Immunol 2012; 12:670-5. [DOI: 10.1097/aci.0b013e32835af232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
40
|
Punwani D, Gonzalez-Espinosa D, Comeau AM, Dutra A, Pak E, Puck J. Cellular calibrators to quantitate T-cell receptor excision circles (TRECs) in clinical samples. Mol Genet Metab 2012; 107:586-91. [PMID: 23062576 PMCID: PMC3483425 DOI: 10.1016/j.ymgme.2012.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 09/15/2012] [Accepted: 09/15/2012] [Indexed: 11/17/2022]
Abstract
T-cell receptor excision circles (TRECs) are circular DNA molecules formed during rearrangement of the T-cell receptor (TCR) genes during lymphocyte development. Copy number of the junctional portion of the δRec-ψJα TREC, assessed by quantitative PCR (qPCR) using DNA from dried blood spots (DBS), is a biomarker for newly formed T cells and absent or low numbers of TRECs indicate SCID (severe combined immunodeficiency) or T lymphocytopenia. No quantitation standard for TRECs exists. To permit comparison of TREC qPCR results with a reliable method for counting TRECs across different laboratories, we sought to construct a stable cell line containing a normal human chromosomal constitution and a single copy of the TREC junction sequence. A human EBV (Epstein Barr virus)-transformed B-cell line was transduced with a lentivirus encoding mCherry fluorescence, puromycin resistance and the δRec-ψJα TREC sequence. A TREC-EBV cell line, with each cell carrying a single lentiviral insertion was established, expanded and shown to have one TREC copy per diploid genome. Graded numbers of TREC-EBV cells added to aliquots of T lymphocyte depleted blood showed TREC copy number proportional to TREC-EBV cell number. TREC-EBV cells, therefore, constitute a reproducible cellular calibrator for TREC assays, useful for both population-based screening for severe combined immunodeficiency and evaluation of naïve T-cell production in clinical settings.
Collapse
Affiliation(s)
- Divya Punwani
- Dept. of Pediatrics, University of California San Francisco, San Francisco, CA 91413; USA
| | | | - Anne Marie Comeau
- New England Newborn Screening Program, Dept. of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01610; USA
| | - Amalia Dutra
- National Human Genome Research Institute, NIH, Bethesda, MD 20892; USA
| | - Evgenia Pak
- National Human Genome Research Institute, NIH, Bethesda, MD 20892; USA
| | - Jennifer Puck
- Dept. of Pediatrics, University of California San Francisco, San Francisco, CA 91413; USA
- Corresponding author: Jennifer M. Puck, MD, UCSF Department of Pediatrics, Box 0519, 513 Parnassus Avenue, HSE 301A, San Francisco, CA 94143-0519,
| |
Collapse
|
41
|
Lang PO, Govind S, Dramé M, Aspinall R. Comparison of manual and automated DNA purification for measuring TREC in dried blood spot (DBS) samples with qPCR. J Immunol Methods 2012; 384:118-27. [DOI: 10.1016/j.jim.2012.07.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/16/2022]
|