1
|
Yazdanpanah N, Rezaei N. The multidisciplinary approach to diagnosing inborn errors of immunity: a comprehensive review of discipline-based manifestations. Expert Rev Clin Immunol 2024; 20:1237-1259. [PMID: 38907993 DOI: 10.1080/1744666x.2024.2372335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/21/2024] [Indexed: 06/24/2024]
Abstract
INTRODUCTION Congenital immunodeficiency is named primary immunodeficiency (PID), and more recently inborn errors of immunity (IEI). There are more than 485 conditions classified as IEI, with a wide spectrum of clinical and laboratory manifestations. AREAS COVERED Regardless of the developing knowledge of IEI, many physicians do not think of IEI when approaching the patient's complaint, which leads to delayed diagnosis, misdiagnosis, serious infectious and noninfectious complications, permanent end-organ damage, and even death. Due to the various manifestations of IEI and the wide spectrum of associated conditions, patients refer to specialists in different disciplines of medicine and undergo - mainly symptomatic - treatments, and because IEI are not included in physicians' differential diagnosis, the main disease remains undiagnosed. EXPERT OPINION A multidisciplinary approach may be a proper solution. Manifestations and the importance of a multidisciplinary approach in the diagnosis of main groups of IEI are discussed in this article.
Collapse
Affiliation(s)
- Niloufar Yazdanpanah
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Tsilifis C, Spegarova JS, Good R, Griffin H, Engelhardt KR, Graham S, Hughes S, Arkwright PD, Hambleton S, Gennery AR. Omenn Syndrome in Two Infants with Different Hypomorphic Variants in Janus Kinase 3. J Clin Immunol 2024; 44:98. [PMID: 38598033 PMCID: PMC11006754 DOI: 10.1007/s10875-024-01699-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 04/02/2024] [Indexed: 04/11/2024]
Abstract
Biallelic null or hypomorphic variants in JAK3 cause SCID and less frequently Omenn syndrome. We investigated homozygous hypomorphic JAK3 mutations in two patients, and expression and function of a novel JAK3R431P variant in Omenn syndrome. Immunophenotyping of PBMC from the patient with the novel JAK3R431P variant was undertaken, by flow cytometry and Phosflow after stimulation with IL-2, IL-7, and IL-15. JAK3 expression was investigated by Western blotting. We report two patients with homozygous hypomorphic JAK3 variants and clinical features of Omenn syndrome. One patient had a previously described JAK3R775H variant, and the second had a novel JAK3R431P variant. One patient with a novel JAK3R431P variant had normal expression of JAK3 in immortalised EBV-LCL cells but reduced phosphorylation of STAT5 after stimulation with IL-2, IL-7, and IL-15 consistent with impaired kinase activity. These results suggest the JAK3R431P variant to be hypomorphic. Both patients are alive and well after allogeneic haematopoietic stem cell transplantation. They have full donor chimerism, restitution of thymopoiesis and development of appropriate antibody responses following vaccination. We expand the phenotype of hypomorphic JAK3 deficiency and demonstrate the importance of functional testing of novel variants in disease-causing genes.
Collapse
Affiliation(s)
- Christo Tsilifis
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Victoria Wing, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Ross Good
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Helen Griffin
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Karin R Engelhardt
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Sophie Graham
- Faculty of Medical Sciences, Medical School, Newcastle University, Newcastle Upon Tyne, UK
| | - Stephen Hughes
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Peter D Arkwright
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Sophie Hambleton
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Victoria Wing, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Andrew R Gennery
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Victoria Wing, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK.
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
3
|
Liongue C, Ratnayake T, Basheer F, Ward AC. Janus Kinase 3 (JAK3): A Critical Conserved Node in Immunity Disrupted in Immune Cell Cancer and Immunodeficiency. Int J Mol Sci 2024; 25:2977. [PMID: 38474223 DOI: 10.3390/ijms25052977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
The Janus kinase (JAK) family is a small group of protein tyrosine kinases that represent a central component of intracellular signaling downstream from a myriad of cytokine receptors. The JAK3 family member performs a particularly important role in facilitating signal transduction for a key set of cytokine receptors that are essential for immune cell development and function. Mutations that impact JAK3 activity have been identified in a number of human diseases, including somatic gain-of-function (GOF) mutations associated with immune cell malignancies and germline loss-of-function (LOF) mutations associated with immunodeficiency. The structure, function and impacts of both GOF and LOF mutations of JAK3 are highly conserved, making animal models highly informative. This review details the biology of JAK3 and the impact of its perturbation in immune cell-related diseases, including relevant animal studies.
Collapse
Affiliation(s)
- Clifford Liongue
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3216, Australia
| | | | - Faiza Basheer
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3216, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3216, Australia
| |
Collapse
|
4
|
Oftedal BE, Sjøgren T, Wolff ASB. Interferon autoantibodies as signals of a sick thymus. Front Immunol 2024; 15:1327784. [PMID: 38455040 PMCID: PMC10917889 DOI: 10.3389/fimmu.2024.1327784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
Type I interferons (IFN-I) are key immune messenger molecules that play an important role in viral defense. They act as a bridge between microbe sensing, immune function magnitude, and adaptive immunity to fight infections, and they must therefore be tightly regulated. It has become increasingly evident that thymic irregularities and mutations in immune genes affecting thymic tolerance can lead to the production of IFN-I autoantibodies (autoAbs). Whether these biomarkers affect the immune system or tissue integrity of the host is still controversial, but new data show that IFN-I autoAbs may increase susceptibility to severe disease caused by certain viruses, including SARS-CoV-2, herpes zoster, and varicella pneumonia. In this article, we will elaborate on disorders that have been identified with IFN-I autoAbs, discuss models of how tolerance to IFN-Is is lost, and explain the consequences for the host.
Collapse
Affiliation(s)
- Bergithe E. Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Thea Sjøgren
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S. B. Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
5
|
Justiz-Vaillant AA, Gopaul D, Akpaka PE, Soodeen S, Arozarena Fundora R. Severe Combined Immunodeficiency-Classification, Microbiology Association and Treatment. Microorganisms 2023; 11:1589. [PMID: 37375091 DOI: 10.3390/microorganisms11061589] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Severe combined immunodeficiency (SCID) is a primary inherited immunodeficiency disease that presents before the age of three months and can be fatal. It is usually due to opportunistic infections caused by bacteria, viruses, fungi, and protozoa resulting in a decrease in number and impairment in the function of T and B cells. Autosomal, X-linked, and sporadic forms exist. Evidence of recurrent opportunistic infections and lymphopenia very early in life should prompt immunological investigation and suspicion of this rare disorder. Adequate stem cell transplantation is the treatment of choice. This review aimed to provide a comprehensive approach to the microorganisms associated with severe combined immunodeficiency (SCID) and its management. We describe SCID as a syndrome and summarize the different microorganisms that affect children and how they can be investigated and treated.
Collapse
Affiliation(s)
- Angel A Justiz-Vaillant
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Darren Gopaul
- Department of Internal Medicine, Port of Spain General Hospital, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Patrick Eberechi Akpaka
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
- Eric Williams Medical Sciences Complex, North Central Regional Health Authority, Champs Fleurs, Trinidad and Tobago
| | - Sachin Soodeen
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Rodolfo Arozarena Fundora
- Eric Williams Medical Sciences Complex, North Central Regional Health Authority, Champs Fleurs, Trinidad and Tobago
- Department of Clinical and Surgical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| |
Collapse
|
6
|
Jain NK, Tailang M, Jain HK, Chandrasekaran B, Sahoo BM, Subramanian A, Thangavel N, Aldahish A, Chidambaram K, Alagusundaram M, Kumar S, Selvam P. Therapeutic implications of current Janus kinase inhibitors as anti-COVID agents: A review. Front Pharmacol 2023; 14:1135145. [PMID: 37021053 PMCID: PMC10067607 DOI: 10.3389/fphar.2023.1135145] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/09/2023] [Indexed: 04/07/2023] Open
Abstract
Severe cases of COVID-19 are characterized by hyperinflammation induced by cytokine storm, ARDS leading to multiorgan failure and death. JAK-STAT signaling has been implicated in immunopathogenesis of COVID-19 infection under different stages such as viral entry, escaping innate immunity, replication, and subsequent inflammatory processes. Prompted by this fact and prior utilization as an immunomodulatory agent for several autoimmune, allergic, and inflammatory conditions, Jakinibs have been recognized as validated small molecules targeting the rapid release of proinflammatory cytokines, primarily IL-6, and GM-CSF. Various clinical trials are under investigation to evaluate Jakinibs as potential candidates for treating COVID-19. Till date, there is only one small molecule Jakinib known as baricitinib has received FDA-approval as a standalone immunomodulatory agent in treating critical COVID-19 patients. Though various meta-analyses have confirmed and validated the safety and efficacy of Jakinibs, further studies are required to understand the elaborated pathogenesis of COVID-19, duration of Jakinib treatment, and assess the combination therapeutic strategies. In this review, we highlighted JAK-STAT signalling in the pathogenesis of COVID-19 and clinically approved Jakinibs. Moreover, this review described substantially the promising use of Jakinibs and discussed their limitations in the context of COVID-19 therapy. Hence, this review article provides a concise, yet significant insight into the therapeutic implications of Jakinibs as potential anti-COVID agents which opens up a new horizon in the treatment of COVID-19, effectively.
Collapse
Affiliation(s)
- Nem Kumar Jain
- School of Pharmacy, ITM University, Gwalior, Madhya Pradesh, India
- School of Studies in Pharmaceutical Sciences, Jiwaji University, Gwalior, Madhya Pradesh, India
| | - Mukul Tailang
- School of Studies in Pharmaceutical Sciences, Jiwaji University, Gwalior, Madhya Pradesh, India
| | - Hemant Kumar Jain
- Department of General Medicine, Government Medical College, Datia, Madhya Pradesh, India
| | - Balakumar Chandrasekaran
- Faculty of Pharmacy, Philadelphia University, Amman, Jordan
- *Correspondence: Balakumar Chandrasekaran, ; Palani Selvam,
| | - Biswa Mohan Sahoo
- Roland Institute of Pharmaceutical Sciences, Berhampur, Odisha, India
| | - Anandhalakshmi Subramanian
- Department of Microbiology and Clinical Parasitology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Neelaveni Thangavel
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Afaf Aldahish
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Kumarappan Chidambaram
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - M. Alagusundaram
- School of Pharmacy, ITM University, Gwalior, Madhya Pradesh, India
| | - Santosh Kumar
- School of Sciences, ITM University, Gwalior, Madhya Pradesh, India
| | - Palani Selvam
- School of Medicine, College of Medicine and Health Sciences, Jijiga University, Jijiga, Ethiopia
- *Correspondence: Balakumar Chandrasekaran, ; Palani Selvam,
| |
Collapse
|
7
|
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]
|
8
|
Matza Porges S, Shamriz O. Genetics of Immune Dysregulation and Cancer Predisposition: Two Sides of the Same Coin. Clin Exp Immunol 2022; 210:114-127. [PMID: 36165533 PMCID: PMC9750831 DOI: 10.1093/cei/uxac089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/17/2022] [Accepted: 09/23/2022] [Indexed: 01/25/2023] Open
Abstract
Approximately 10% of cancers have a hereditary predisposition. However, no genetic diagnosis is available in 60%-80% of familial cancers. In some of these families, immune dysregulation-mediated disease is frequent. The immune system plays a critical role in identifying and eliminating tumors; thus, dysregulation of the immune system can increase the risk of developing cancer. This review focuses on some of the genes involved in immune dysregulation the promote the risk for cancer. Genetic counseling for patients with cancer currently focuses on known genes that raise the risk of cancer. In missing hereditary familial cases, the history family of immune dysregulation should be recorded, and genes related to the immune system should be analyzed in relevant families. On the other hand, patients with immune disorders diagnosed with a pathogenic mutation in an immune regulatory gene may have an increased risk of cancer. Therefore, those patients need to be under surveillance for cancer. Gene panel and exome sequencing are currently standard methods for genetic diagnosis, providing an excellent opportunity to jointly test cancer and immune genes.
Collapse
Affiliation(s)
- Sigal Matza Porges
- Department of Human Genetics, Institute for Medical Research, the Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Biotechnology, Hadassah Academic College, Jerusalem, Israel
| | - Oded Shamriz
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah Medical Organization, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
9
|
Sjøgren T, Bratland E, Røyrvik EC, Grytaas MA, Benneche A, Knappskog PM, Kämpe O, Oftedal BE, Husebye ES, Wolff ASB. Screening patients with autoimmune endocrine disorders for cytokine autoantibodies reveals monogenic immune deficiencies. J Autoimmun 2022; 133:102917. [PMID: 36191466 DOI: 10.1016/j.jaut.2022.102917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Autoantibodies against type I interferons (IFN) alpha (α) and omega (ω), and interleukins (IL) 17 and 22 are a hallmark of autoimmune polyendocrine syndrome type 1 (APS-1), caused by mutations in the autoimmune regulator (AIRE) gene. Such antibodies are also seen in a number of monogenic immunodeficiencies. OBJECTIVES To determine whether screening for cytokine autoantibodies (anti-IFN-ω and anti-IL22) can be used to identify patients with monogenic immune disorders. METHODS A novel ELISA assay was employed to measure IL22 autoantibodies in 675 patients with autoimmune primary adrenal insufficiency (PAI) and a radio immune assay (RIA) was used to measure autoantibodies against IFN-ω in 1778 patients with a variety of endocrine diseases, mostly of autoimmune aetiology. Positive cases were sequenced for all coding exons of the AIRE gene. If no AIRE mutations were found, we applied next generation sequencing (NGS) to search for mutations in immune related genes. RESULTS We identified 29 patients with autoantibodies against IFN-ω and/or IL22. Of these, four new APS-1 cases with disease-causing variants in AIRE were found. In addition, we identified two patients with pathogenic heterozygous variants in CTLA4 and NFKB2, respectively. Nine rare variants in other immune genes were identified in six patients, although further studies are needed to determine their disease-causing potential. CONCLUSION Screening of cytokine autoantibodies can efficiently identify patients with previously unknown monogenic and possible oligogenic causes of autoimmune and immune deficiency diseases. This information is crucial for providing personalised treatment and follow-up of patients and their relatives.
Collapse
Affiliation(s)
- Thea Sjøgren
- Department of Clinical Science, University of Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway
| | - Eirik Bratland
- Department of Clinical Science, University of Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway; Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Ellen C Røyrvik
- Department of Clinical Science, University of Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway
| | - Marianne Aa Grytaas
- Department of Medicine, Haukeland University Hospital, Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway
| | - Andreas Benneche
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Per M Knappskog
- Department of Clinical Science, University of Bergen, Norway; Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Olle Kämpe
- KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway; Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bergithe E Oftedal
- Department of Clinical Science, University of Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway
| | - Eystein S Husebye
- Department of Clinical Science, University of Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway.
| | - Anette S B Wolff
- Department of Clinical Science, University of Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway.
| |
Collapse
|
10
|
Zebrafish Model of Severe Combined Immunodeficiency (SCID) Due to JAK3 Mutation. Biomolecules 2022; 12:biom12101521. [PMID: 36291730 PMCID: PMC9599616 DOI: 10.3390/biom12101521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/18/2022] Open
Abstract
JAK3 is principally activated by members of the interleukin-2 receptor family and plays an essential role in lymphoid development, with inactivating JAK3 mutations causing autosomal-recessive severe combined immunodeficiency (SCID). This study aimed to generate an equivalent zebrafish model of SCID and to characterize the model across the life-course. Genome editing of zebrafish jak3 created mutants similar to those observed in human SCID. Homozygous jak3 mutants showed reduced embryonic T lymphopoiesis that continued through the larval stage and into adulthood, with B cell maturation and adult NK cells also reduced and neutrophils impacted. Mutant fish were susceptible to lymphoid leukemia. This model has many of the hallmarks of human SCID resulting from inactivating JAK3 mutations and will be useful for a variety of pre-clinical applications.
Collapse
|
11
|
Galarreta CI, Kennedy C, Blair DR, Slavotinek A. Expanding the phenotype of PIK3C2A related syndrome: Report of two siblings with novel features and genotype. Am J Med Genet A 2022; 188:2724-2731. [PMID: 35770347 DOI: 10.1002/ajmg.a.62881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/05/2022] [Accepted: 04/15/2022] [Indexed: 01/25/2023]
Abstract
A pair of siblings was ascertained due to multiple congenital anomalies, including strikingly similar facial, skeletal, and ocular abnormalities. Exome sequencing of both the children and their mother revealed two novel PIK3C2A variants in the siblings, c.4381delC (p.Arg1461Glufs*31) and c.1555C > T (p.Arg519Ter). PIK3C2A belongs to the Class IIa family of Phosphatidylinositol-3-kinases, which create second messenger lipids that regulate a wide range of downstream signaling pathways involved in cell growth, survival and migration. Tiosano et al. (2019) identified the first monogenic disorder associated with biallelic PIK3C2A loss-of-function variants (oculoskeletodental syndrome). The novel syndrome was characterized by short stature, coarse facial features, ocular and skeletal abnormalities. This report describes two additional siblings affected by the PIK3C2A-related syndrome, confirms core clinical features, establishes intrafamilial variability and expands the phenotype to include proteinuria.
Collapse
Affiliation(s)
- Carolina I Galarreta
- Medical Genetics and Metabolism Department, Valley Children's Hospital, Madera, California, USA
| | - Colleen Kennedy
- Medical Genetics and Metabolism Department, Valley Children's Hospital, Madera, California, USA
| | - David R Blair
- Department of Pediatrics, Division of Genetics, University of California, San Francisco, California, USA
| | - Anne Slavotinek
- Department of Pediatrics, Division of Genetics, University of California, San Francisco, California, USA
| |
Collapse
|
12
|
Sekine Y, Kikkawa K, Witthuhn BA, Kashiwakura JI, Muromoto R, Kitai Y, Fujimuro M, Oritani K, Matsuda T. A novel intramolecular negative regulation of mouse Jak3 activity by tyrosine 820. Int Immunol 2022; 34:303-312. [PMID: 35192696 DOI: 10.1093/intimm/dxac005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Jak3, a member of the Janus kinase family, is essential for the cytokine receptor common gamma (γ) chain-mediated signaling. During activation of Jak3, tyrosine residues are phosphorylated and potentially regulate its kinase activity. We identified a novel tyrosine phosphorylation site within mouse Jak3, Y820, which is conserved in human Jak3, Y824. IL-2-induced tyrosine phosphorylation of Jak3 Y824 in human T cell line HuT78 cells was detected by using a phosphospecific, pY824, antibody. Mutation of mouse Jak3 Y820 to alanine (Y820A) showed increased autophosphorylation of Jak3 and enhanced STAT5 tyrosine phosphorylation and transcriptional activation. Stably expressed Jak3 Y820A in F7 cells, an IL-2 responsive mouse pro-B cell line Ba/F3, exhibited enhanced IL-2-dependent cell growth. Mechanistic studies demonstrated that interaction between Jak3 and STAT5 increased in Jak3 Y820A compared to Jak3 WT. These data suggest that Jak3 Y820 plays a role in negative regulation of Jak3-mediated STAT5 signaling cascade upon IL-2-stimulation. We speculate that this occurs through an interaction promoted by the tyrosine phosphorylated Y820 or a conformational change by Y820 mutation with either the STAT directly or with the recruitment of molecules such as phosphatases via a SH2 interaction. Additional studies will focus on these interactions as Jak3 plays a crucial role in disease and health.
Collapse
Affiliation(s)
- Yuichi Sekine
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kazuna Kikkawa
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Bruce A Witthuhn
- Center for Mass Spectrometry and Proteomics, University of Minnesota, Minneapolis, MN, USA
| | - Jun-Ichi Kashiwakura
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Ryuta Muromoto
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yuichi Kitai
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Masahiro Fujimuro
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kenji Oritani
- Department of Hematology, International University of Health and Welfare, 4-3 Kouzunomori, Narita, Chiba, Japan
| | - Tadashi Matsuda
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
13
|
Pan Y, Pan H, Lian C, Wu B, Lin J, Huang G, Cui B. Case Report: Mutations in JAK3 causing severe combined immunodeficiency complicated by disseminated Bacille Calmette-Guérin disease and Pneumocystis pneumonia. Front Immunol 2022; 13:1055607. [PMID: 36466884 PMCID: PMC9712176 DOI: 10.3389/fimmu.2022.1055607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND As a form of severe combined immunodeficiency (SCID), Janus kinase 3 (JAK3) deficiency can be fatal during severe infections in children, especially after inoculation of live-attenuated vaccines. We report a unique case of JAK3 deficiency with two compound heterozygous JAK3 mutations complicated by disseminated Bacille Calmette-Guérin (BCG) disease and Pneumocystis pneumonia. CASE DESCRIPTION A 5-month-old Chinese girl presented with recurring fever and productive cough after BCG vaccination and ineffective antibiotic treatment. Chest CT demonstrated bilateral infiltrations, enlarged mediastinal and axillary lymph nodes, and hypoplasia of the thymus. Mycobacterium tuberculosis and Pneumocystis jirovecii were detected from blood samples by sequencing. Acid-fast bacilli were also found from the sputum aspirate and gastric aspirate. Lymphocyte subset analyses indicated T-B+NK- immunodeficiency, and gene sequencing identified two heterozygous missense mutations (one unreported globally) in the Janus homology 7 (JH7) domain of JAK3. The patient received rifampicin, isoniazid, ethambutol, and trimethoprim/sulfamethoxazole and was discharged after improvements but against advice. OUTCOME The patient died at 13 months of age due to severe infections and hepatic damage. DISCUSSION SCID should be recognized before inoculation of live-attenuated vaccines in children. Newborn screening for SCID is advocated. Further investigations are needed to better understand the pathogenicity of the variants and molecular mechanism of the JH7 domain of JAK3.
Collapse
Affiliation(s)
- Ying Pan
- The Department of Pediatrics, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Hui Pan
- The Outpatient Department, Shantou Longhu People’s Hospital, Shantou, Guangdong, China
- The Clinical Research Unit, Shantou University Medical College, Shantou, Guangdong, China
| | - Chunan Lian
- The Department of Pediatrics, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Beiyan Wu
- The Department of Pediatrics, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jieying Lin
- The Department of Pediatrics, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Guang Huang
- The Department of Pediatrics, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Binglin Cui
- The Department of Pediatrics, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- *Correspondence: Binglin Cui,
| |
Collapse
|
14
|
Fathi E, Farahzadi R, Valipour B. Alginate/gelatin encapsulation promotes NK cells differentiation potential of bone marrow resident C-kit + hematopoietic stem cells. Int J Biol Macromol 2021; 177:317-327. [PMID: 33621568 DOI: 10.1016/j.ijbiomac.2021.02.131] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/16/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023]
Abstract
The ability of natural killer (NK) cells to destroy cancerous cells with no prior sensitization has made them attractive candidates for cell therapy. The application of hydrogels must be notified as cell delivery vehicles in cell differentiation. The present study was conducted to investigate the effect of alginate-gelatin encapsulation on NK cell differentiation potential of C-kit+ cells. C-kit+ cells were differentiated to NK cells under both encapsulated and un-encapsulated conditions. Next, the cells were subjected to real-time polymerase chain reaction (PCR) and western blotting for the assessment of their telomere length and protein expressions, respectively. Afterward, culture medium was collected to measure cytokines levels. Thereafter, the differentiated NK cells were co-cultured with Molt-4 cells to investigate the potency of cell apoptosis by Annexin V/PI assay. A significant change was observed in the protein expression of Janus kinase/Signal transducers (JAK/STAT) pathway components. Additionally, the encapsulation caused an increase in the apoptosis of Molt-4 cells and telomere length of NK cells differentiated C-kit+ cells. Therefore, it can be concluded that the effects of encapsulation on NK cell's differentiation of C-kit+ cells could be resulted from the secreted cytokines of interleukin (IL)-2, IL-3, IL-7, and IL-12 as well as the increased telomere length.
Collapse
Affiliation(s)
- Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Behnaz Valipour
- Department of Histopathology and Anatomy, Faculty of Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| |
Collapse
|
15
|
Chauhan P, Nair A, Patidar A, Dandapat J, Sarkar A, Saha B. A primer on cytokines. Cytokine 2021; 145:155458. [PMID: 33581983 DOI: 10.1016/j.cyto.2021.155458] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/19/2022]
Abstract
Cytokines are pleiotropic polypeptides that control the development of and responses mediated by immune cells. Cytokine classification predominantly relies on [1] the target receptor(s), [2] the primary structural features of the extracellular domains of their receptors, and [3] their receptor composition. Functionally, cytokines are either pro-inflammatory or anti-inflammatory, hematopoietic colony-stimulating factors, developmental and would healing maintaining immune homeostasis. When the balance in C can form complex networks amongst themselves that may affect the homeostasis and diseases. Cytokines can affect resistance and susceptibility for many diseases and their availability in the host cytokine production and interaction is disturbed, immunopathogenesis sets in. Therefore, cytokine-targeting bispecific, and chimeric antibodies form a significant mode of immnuo-therapeutics Although the field has grown deep and wide, many areas of cytokine biology remain unknown. Here, we have reviewed these cytokines along with the organization, signaling, and functions through respective cytokine-receptor-families. Being part of the special issue on the Role of Cytokines in Leishmaniasis, this review is intended to be used as an organized primer on cytokines and not a resource for detailed discussion- for which a two-volume Handbook of cytokines is available- on each of the cytokines. Priming the readers on cytokines, we next brief the role of cytokines in Leishmaniasis. In the brief, we do not provide an account of each of the involved cytokines known to date, instead, we offer a temporal relationship between the cytokines and the progress of the infection towards the alternate outcomes- healing or non-healing- of the infection.
Collapse
Affiliation(s)
- Prashant Chauhan
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Arathi Nair
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Ashok Patidar
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Jagneshwar Dandapat
- P.G. Department of Biotechnology, Utkal University, Bhubaneswar 751004, India
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar 751024, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India; Trident Academy of Creative Technology, Bhubaneswar 751024, India; Department of Allied Health Sciences, BLDE (Deemed University), Vijayapura 562135, India.
| |
Collapse
|
16
|
Khanolkar A, Wilks JD, Liu G, Simpson BM, Caparelli EA, Kirschmann DA, Bergerson J, Fuleihan RL. A case of aberrant CD8 T cell-restricted IL-7 signaling with a Janus kinase 3 defect-associated atypical severe combined immunodeficiency. Immunol Res 2020; 68:13-27. [PMID: 32215810 DOI: 10.1007/s12026-020-09123-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Severe combined immunodeficiency (SCID) disorders compromise lymphocyte numbers and/or function. One subset of SCID typically affects T cell and Natural Killer (NK) cell development in tandem (T-B+NK-) due to mutations arising in the genes encoding the common γ chain or Janus Kinase 3 (JAK3). In rare circumstances, mutations in the JAK3 gene have been reported to cause atypical SCID that selectively affects T cells (T-B+NK+). Here we describe a case involving a female infant who was referred to our institution on day nine of life following an abnormal newborn screen result for T-SCID. Immunological assessments revealed a T-B+NK+ phenotype and molecular analyses, including whole exome sequencing, identified compound heterozygous JAK3 variants (R117C and E658K). Pre-transplant phosflow analyses revealed a persistent IL-7 signaling defect, based on phospho-STAT5 measurements, only in CD8 but not CD4 T cells. Intriguingly, phospho-STAT5 signals in response to IL-2 stimulation were not affected in either CD4 or CD8 T cells. The pre-transplant clinical course was unremarkable, and the patient received a cord-blood stem cell transplant on day 716 of life. Post-transplant monitoring revealed that despite normalization of lymphocyte counts, the CD8 T cell-restricted IL-7 signaling defect was still evident at day 627 post-transplant (phospho-STAT5 signal in CD8 T cells was > 60% reduced compared with CD4 T cells). The post-transplant clinical course has also been complicated by identification of autoimmune responses and likely GVHD-induced ichthyosis. To the best of our knowledge, this report represents the third case of JAK3-associated atypical SCID reported in the literature.
Collapse
Affiliation(s)
- Aaruni Khanolkar
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, 60611, USA. .,Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Chicago, IL, 60611, USA.
| | - Jeffrey D Wilks
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, 60611, USA
| | - Guorong Liu
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, 60611, USA
| | - Bridget M Simpson
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, 60611, USA
| | - Edward A Caparelli
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, 60611, USA
| | - Dawn A Kirschmann
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, 60611, USA
| | - Jenna Bergerson
- Department of Pediatrics, Division of Allergy and Immunology, Feinberg School of Medicine, Chicago, IL, 60611, USA.,Primary Immunodeficiency Clinic, NIH/NIAID, 9000 Rockville Pike, Bldg. 10, Room 11N244A MSC 1960, Bethesda, MD, 20892, USA
| | - Ramsay L Fuleihan
- Department of Pediatrics, Division of Allergy and Immunology, Feinberg School of Medicine, Chicago, IL, 60611, USA.,Division of Allergy & Immunology, Sidra Medicine, Doha, Qatar
| |
Collapse
|
17
|
Goldberg L, Simon AJ, Lev A, Barel O, Stauber T, Kunik V, Rechavi G, Somech R. Atypical immune phenotype in severe combined immunodeficiency patients with novel mutations in IL2RG and JAK3. Genes Immun 2020; 21:326-334. [PMID: 32921793 DOI: 10.1038/s41435-020-00111-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/14/2020] [Accepted: 09/02/2020] [Indexed: 11/09/2022]
Abstract
Mutations in the common gamma chain of the interleukin 2 receptor (IL2RG) or the associated downstream signaling enzyme Janus kinase 3 (JAK3) genes are typically characterized by a T cell-negative, B cell-positive, natural killer (NK) cell-negative (T-B+NK-) severe combined immunodeficiency (SCID) immune phenotype. We report clinical course, immunological, genetic and proteomic work-up of two patients with different novel mutations in the IL-2-JAK3 pathway with a rare atypical presentation of T-B+NK- SCID. Lymphocyte subpopulation revealed significant T cells lymphopenia, normal B cells, and NK cells counts (T-B+NK+SCID). Despite the presence of B cells, IgG levels were low and IgA and IgM levels were undetectable. T-cell proliferation in response to mitogens in patient 1 was very low and T-cell receptor V-beta chain repertoire in patient 2 was polyclonal. Whole-exome sequencing revealed novel mutations in both patients (patient 1-c.923delC frame-shift mutation in the IL2RG gene, patient 2-c.G172A a homozygous missense mutation in the JAK3 gene). Bioinformatic analysis of the JAK3 mutation indicated deleterious effect and 3D protein modeling located the mutation to a surface exposed alpha-helix structure. Our findings help to link between genotype and phenotype, which is a key factor for the diagnosis and treatment of SCID patients.
Collapse
Affiliation(s)
- Lior Goldberg
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation (JMF) Center, Sheba Medical Center, Tel HaShomer, Israel
| | - Amos J Simon
- Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation (JMF) Center, Sheba Medical Center, Tel HaShomer, Israel.,Sheba Cancer Research Center, Sheba Medical Center, Tel HaShomer, Israel
| | - Atar Lev
- Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation (JMF) Center, Sheba Medical Center, Tel HaShomer, Israel
| | - Ortal Barel
- Sheba Cancer Research Center, Sheba Medical Center, Tel HaShomer, Israel.,The Wohl Institute for Translational Medicine, Sheba Medical Center, Tel HaShomer, Israel
| | - Tali Stauber
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation (JMF) Center, Sheba Medical Center, Tel HaShomer, Israel
| | - Vered Kunik
- Bioinformatics Consulting, Gat Rimon, Israel
| | - Gideon Rechavi
- Sheba Cancer Research Center, Sheba Medical Center, Tel HaShomer, Israel.,The Wohl Institute for Translational Medicine, Sheba Medical Center, Tel HaShomer, Israel
| | - Raz Somech
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation (JMF) Center, Sheba Medical Center, Tel HaShomer, Israel.
| |
Collapse
|
18
|
Human diseases caused by impaired signal transducer and activator of transcription and Janus kinase signaling. Curr Opin Pediatr 2019; 31:843-850. [PMID: 31693596 DOI: 10.1097/mop.0000000000000841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW The Janus kinase (JAK) and signal transducer of activation (STAT) pathway plays a key role in the immune system. It is employed by diverse cytokines, interferons, growth factors and related molecules. Mutations in JAK/STAT pathway have been implicated in human disease. Here we review JAK/STAT biology and diseases associated with mutations in this pathway. RECENT FINDINGS Over the past 10 years, many mutations in JAK/STAT pathway has been discovered. These disorders have provided insights to human immunology. SUMMARY In this review, we summarize the biology of each STAT and JAK as well as discuss the human disease that results from somatic or germline mutations to include typical presentation, immunological parameters and treatment.
Collapse
|
19
|
Pseudokinases: From Allosteric Regulation of Catalytic Domains and the Formation of Macromolecular Assemblies to Emerging Drug Targets. Catalysts 2019. [DOI: 10.3390/catal9090778] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pseudokinases are a member of the kinase superfamily that lack one or more of the canonical residues required for catalysis. Protein pseudokinases are widely distributed across species and are present in proteins that perform a great diversity of roles in the cell. They represent approximately 10% to 40% of the kinome of a multicellular organism. In the human, the pseudokinase subfamily consists of approximately 60 unique proteins. Despite their lack of one or more of the amino acid residues typically required for the productive interaction with ATP and metal ions, which is essential for the phosphorylation of specific substrates, pseudokinases are important functional molecules that can act as dynamic scaffolds, competitors, or modulators of protein–protein interactions. Indeed, pseudokinase misfunctions occur in diverse diseases and represent a new therapeutic window for the development of innovative therapeutic approaches. In this contribution, we describe the structural features of pseudokinases that are used as the basis of their classification; analyse the interactome space of human pseudokinases and discuss their potential as suitable drug targets for the treatment of various diseases, including metabolic, neurological, autoimmune, and cell proliferation disorders.
Collapse
|
20
|
Hammarén HM, Virtanen AT, Raivola J, Silvennoinen O. The regulation of JAKs in cytokine signaling and its breakdown in disease. Cytokine 2019; 118:48-63. [DOI: 10.1016/j.cyto.2018.03.041] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/29/2018] [Accepted: 03/30/2018] [Indexed: 01/12/2023]
|
21
|
Vargas-Hernández A, Forbes LR. JAK/STAT proteins and their biological impact on NK cell development and function. Mol Immunol 2019; 115:21-30. [PMID: 30704805 DOI: 10.1016/j.molimm.2018.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/04/2018] [Accepted: 12/08/2018] [Indexed: 02/07/2023]
Abstract
NK cells are important early effectors in the innate immune response to a variety of viral infections and for elimination of tumor cells. The JAK/STAT signaling cascade is critical for NK cell development, maturation, survival, and proliferation, therefore, it is important to understand the role of this pathway in NK cell biology. Many cytokines can activate multiple JAK/STAT protein family members, creating a severe phenotype when mutations impair their function or expression. Here we discuss the impact of defective JAK/STAT signaling pathways on NK cell development, activation and cytotoxicity.
Collapse
Affiliation(s)
- Alexander Vargas-Hernández
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Center for Human Immunobiology, Department of Allergy, Immunology and Rheumatology, Houston, TX, USA
| | - Lisa R Forbes
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Center for Human Immunobiology, Department of Allergy, Immunology and Rheumatology, Houston, TX, USA.
| |
Collapse
|
22
|
Lu S, Zhao K, Wang X, Liu H, Ainiwaer X, Xu Y, Ye M. Use of Laplacian Heat Diffusion Algorithm to Infer Novel Genes With Functions Related to Uveitis. Front Genet 2018; 9:425. [PMID: 30349554 PMCID: PMC6186792 DOI: 10.3389/fgene.2018.00425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 09/10/2018] [Indexed: 12/17/2022] Open
Abstract
Uveitis is the inflammation of the uvea and is a serious eye disease that can cause blindness for middle-aged and young people. However, the pathogenesis of this disease has not been fully uncovered and thus renders difficulties in designing effective treatments. Completely identifying the genes related to this disease can help improve and accelerate the comprehension of uveitis. In this study, a new computational method was developed to infer potential related genes based on validated ones. We employed a large protein–protein interaction network reported in STRING, in which Laplacian heat diffusion algorithm was applied using validated genes as seed nodes. Except for the validated ones, all genes in the network were filtered by three tests, namely, permutation, association, and function tests, which evaluated the genes based on their specialties and associations to uveitis. Results indicated that 59 inferred genes were accessed, several of which were confirmed to be highly related to uveitis by literature review. In addition, the inferred genes were compared with those reported in a previous study, indicating that our reported genes are necessary supplements.
Collapse
Affiliation(s)
- Shiheng Lu
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, China
| | - Ke Zhao
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, China
| | - Xuefei Wang
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, China
| | - Hui Liu
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, China
| | - Xiamuxiya Ainiwaer
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, China
| | - Yan Xu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Min Ye
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, China
| |
Collapse
|
23
|
Di Matteo G, Chiriaco M, Scarselli A, Cifaldi C, Livadiotti S, Di Cesare S, Ferradini V, Aiuti A, Rossi P, Finocchi A, Cancrini C. JAK3 mutations in Italian patients affected by SCID: New molecular aspects of a long-known gene. Mol Genet Genomic Med 2018; 6:713-721. [PMID: 30032486 PMCID: PMC6160700 DOI: 10.1002/mgg3.391] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/20/2018] [Accepted: 02/27/2018] [Indexed: 01/01/2023] Open
Abstract
Background Mutations in the Janus Kinase 3 (JAK3) gene cause an autosomal recessive form of severe combined immunodeficiency (SCID) usually characterized by the absence of both T and NK cells, but preserved numbers of B lymphocytes (T‐B+NK‐SCID). The detection of larger (>100 bp) genomic duplications or deletions can be more difficult to be detected by PCR‐based methods or standard NGS protocols, and a broad range of mutation detection techniques are necessary. Methods We report four unrelated Italian patients (two females and two males) with SCID phenotype. Protein expression, functional studies, molecular analysis by standard methods and NGS, and transcripts studies were performed to obtain a definitive diagnosis. Results Here, we describe four JAK3‐deficient patients from four unrelated families. The first patient is homozygous for the known c.1951 C>T mutation causing the amino acidic change p.R651W. The other two patients, originating from the same small Italian town, resulted compound heterozygotes for the same g.15410_16542del deletion and two different novel mutations, g.13319_13321delTTC and c.933T>G (p.F292V), respectively. The fourth patient was compound heterozygous for the novel mutations p.V599G and p.W709R. Defective STAT5 phosphorylation after IL2 or IL15 stimulation corroborated the mutation pathogenicity. Concerning g.15410_16542del mutation, probably due to an unequal homologous recombination between Alu elements of JAK3 gene, microsatellites analysis revealed that both unrelated Pt2 and Pt3 and their carrier family members shared the same haplotype. These data support the hypothesis of a founder effect for the g.15410_16542del mutation that might have inherited in both unrelated families from the same ancient progenitor. Conclusion Different molecular techniques are still required to obtain a definitive diagnosis of AR‐SCID particularly in all cases in which a monoallelic mutation is found by standard mutation scanning methods.
Collapse
Affiliation(s)
- Gigliola Di Matteo
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Maria Chiriaco
- Department of Pediatrics, Children's Hospital Bambino Gesù, Rome, Italy
| | - Alessia Scarselli
- Department of Pediatrics, Children's Hospital Bambino Gesù, Rome, Italy
| | - Cristina Cifaldi
- Department of Pediatrics, Children's Hospital Bambino Gesù, Rome, Italy
| | | | - Silvia Di Cesare
- Department of Pediatrics, Children's Hospital Bambino Gesù, Rome, Italy
| | - Valentina Ferradini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Pediatric Immunohematology Unit, San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Rossi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Department of Pediatrics, Children's Hospital Bambino Gesù, Rome, Italy
| | - Andrea Finocchi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Department of Pediatrics, Children's Hospital Bambino Gesù, Rome, Italy
| | - Caterina Cancrini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Department of Pediatrics, Children's Hospital Bambino Gesù, Rome, Italy
| |
Collapse
|
24
|
Are Janus Kinase Inhibitors Superior over Classic Biologic Agents in RA Patients? BIOMED RESEARCH INTERNATIONAL 2018; 2018:7492904. [PMID: 29862290 PMCID: PMC5971265 DOI: 10.1155/2018/7492904] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 03/28/2018] [Indexed: 12/13/2022]
Abstract
The Janus Kinases (JAKs) are a family of intracellular tyrosine kinases that provide transmission signals from cytokine, interferons, and many hormones receptors to the nucleus resulting in synthesis of many biologically active compounds and changing cell metabolism and function. That was theoretical background to synthetize the JAK inhibitors (Jakinibs). In recent years a substantial battery of evidence has been collected indicating the potential role of Jakinibs to interact with the specific elements of the immune system, therefore changing the inflammatory response. JAK kinase blockade offers a unique opportunity to block most of the key cytokines enabling the deep interaction into immune system functioning. Following discovery first Jakinibs were intensively studied in various forms of autoimmune diseases, including rheumatoid arthritis, and finally two Jakinibs tofacitinib and Baricitinib have been approved for the treatment of rheumatoid arthritis. Some clinical data indicated that under special circumstances Jakinibs may be even superior to biologics in the treatment of RA; however this suggestion should be verified in large clinical and observational studies.
Collapse
|
25
|
Zhong L, Wang W, Ma M, Gou L, Tang X, Song H. Chronic active Epstein-Barr virus infection as the initial symptom in a Janus kinase 3 deficiency child: Case report and literature review. Medicine (Baltimore) 2017; 96:e7989. [PMID: 29049190 PMCID: PMC5662356 DOI: 10.1097/md.0000000000007989] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
RATIONALE With the progress of sequencing technology, an increasing number of atypical primary immunodeficiency (PID) patients have been discovered, including Janus kinase 3 (JAK3) gene deficiency. PATIENT CONCERNS We report a patient who presented with chronic active Epstein-Barr virus (CAEBV) infection but responded poorly to treatment with ganciclovir. DIAGNOSES Next-generation sequencing (NGS) was performed, including all known PID genes, after which Sanger sequencing was performed to verify the results. Genetic analysis revealed that our patient had 2 novel compound heterozygous mutations of JAK3, a gene previously reported to cause a rare form of autosomal recessive severe combined immunodeficiency with recurrent infections. The p.H27Q mutation came from his father, while p. R222H from his mother. Thus, his diagnosis was corrected for JAK3-deficiency PID and CAEBV. INTERVENTIONS Maintenance treatment of subcutaneous injection of recombinant human interferon α-2a was given to our patient with 2 MU, 3 times a week. OUTCOMES Interferon alpha was applied and the EBV infection was gradually controlled and his symptoms ameliorated remarkably. Our patient is in good health now and did not have relapses. LESSONS The diagnoses of PID should be taken into consideration when CAEBV patients respond poorly to conventional treatments. Good results of our patient indicate that interferon α-2a may be an alternative treatment for those who are unwilling to accept hematopoietic stem cell transplantation (HSCT) like our patient. Literature review identified 59 additional cases of JAK3 deficiency with various infections.
Collapse
|
26
|
A novel mutation in the JH4 domain of JAK3 causing severe combined immunodeficiency complicated by vertebral osteomyelitis. Clin Immunol 2017; 183:198-200. [DOI: 10.1016/j.clim.2017.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 11/23/2022]
|
27
|
De Vries L, Wildenberg M, De Jonge W, D’Haens G. The Future of Janus Kinase Inhibitors in Inflammatory Bowel Disease. J Crohns Colitis 2017; 11:885-893. [PMID: 28158411 PMCID: PMC5881740 DOI: 10.1093/ecco-jcc/jjx003] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/31/2017] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel diseases, such as ulcerative colitis and Crohn's disease, are disabling conditions characterised by chronic, relapsing inflammation of the gastrointestinal tract. Current treatments are not universally effective or, in the case of therapeutic antibodies, are hampered by immune responses. Janus kinase inhibitors are orally delivered small molecules that target cytokine signalling by preventing phosphorylation of Janus kinases associated with the cytokine receptor. Subsequently, phosphorylation of signal transducers and activators of transcription that relay Janus kinase signalling and transcription of cytokines in the nucleus will be diminished. Key cytokines in the pathogenesis of inflammatory bowel diseases are targeted by Janus kinase inhibitors. Several Janus kinase inhibitors are in development for the treatment of inflammatory bowel diseases. Tofacitinib, inhibiting signalling via all Janus kinase family members, was effective in phase 2 and 3 trials in moderate-severe ulcerative colitis. GSK2586184, a Janus kinase 1 selective inhibitor, induced clinical and endoscopic response in ulcerative colitis; however, the study was discontinued at an early stage due to liver toxicity observed in systemic lupus patients receiving the drug. Filgotinib, a Janus kinase 1 selective inhibitor investigated in treatment of Crohn's disease, was superior to placebo. As adverse events associated with the broad immunological effect of these agents have been reported, the future application of these drugs is potentially limited. We will discuss the treatment efficacy of Janus kinase inhibition in inflammatory bowel diseases, how current Janus kinase inhibitors available target immune responses relevant in inflammatory bowel disease, and whether more specific kinase inhibition could be effective.
Collapse
Affiliation(s)
- L.C.S. De Vries
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands,Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - M.E. Wildenberg
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands,Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - W.J. De Jonge
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - G.R. D’Haens
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
28
|
dela Peña-Ponce MG, Rodriguez-Nieves J, Bernhardt J, Tuck R, Choudhary N, Mengual M, Mollan KR, Hudgens MG, Peter-Wohl S, De Paris K. Increasing JAK/STAT Signaling Function of Infant CD4 + T Cells during the First Year of Life. Front Pediatr 2017; 5:15. [PMID: 28271056 PMCID: PMC5318443 DOI: 10.3389/fped.2017.00015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/20/2017] [Indexed: 12/17/2022] Open
Abstract
Most infant deaths occur in the first year of life. Yet, our knowledge of immune development during this period is scarce and derived from cord blood (CB) only. To more effectively combat pediatric diseases, a deeper understanding of the kinetics and the factors that regulate the maturation of immune functions in early life is needed. Increased disease susceptibility of infants is generally attributed to T helper 2-biased immune responses. The differentiation of CD4+ T cells along a specific T helper cell lineage is dependent on the pathogen type, and on costimulatory and cytokine signals provided by antigen-presenting cells. Cytokines also regulate many other aspects of the host immune response. Therefore, toward the goal of increasing our knowledge of early immune development, we defined the temporal development of the Janus kinase (JAK)/signal transducers and activators of transcription (STAT) signaling function of CD4+ T cells using cross-sectional blood samples from healthy infants ages 0 (birth) to 14 months. We specifically focused on cytokines important in T cell differentiation (IFN-γ, IL-12, and IL-4) or in T cell survival and expansion (IL-2 and IL-7) in infant CD4+ T cells. Independent of the cytokine tested, JAK/STAT signaling in infant compared to adult CD4+ T cells was impaired at birth, but increased during the first year, with the most pronounced changes occurring in the first 6 months. The relative change in JAK/STAT signaling of infant CD4+ T cells with age was distinct for each cytokine tested. Thus, while about 60% of CB CD4+ T cells could efficiently activate STAT6 in response to IL-4, less than 5% of CB CD4+ T cells were able to activate the JAK/STAT pathway in response to IFN-γ, IL-12 or IL-2. By 4-6 months of age, the activation of the cytokine-specific STAT molecules was comparable to adults in response to IL-4 and IFN-γ, while IL-2- and IL-12-induced STAT activation remained below adult levels even at 1 year. These results suggest that common developmental and cytokine-specific factors regulate the maturation of the JAK/STAT signaling function in CD4+ T cells during the first year of life.
Collapse
Affiliation(s)
- Myra Grace dela Peña-Ponce
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Jennifer Rodriguez-Nieves
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Janice Bernhardt
- Division of Neonatal Perinatal Medicine, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Ryan Tuck
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Neelima Choudhary
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Michael Mengual
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Katie R. Mollan
- Lineberger Cancer Center, Center for AIDS Research, University of North Carolina, Chapel Hill, NC, USA
| | - Michael G. Hudgens
- Gillings School of Global Public Health, Center for AIDS Research, University of North Carolina, Chapel Hill, NC, USA
| | - Sigal Peter-Wohl
- Division of Neonatal Perinatal Medicine, Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
29
|
Westhovens R, Taylor PC, Alten R, Pavlova D, Enríquez-Sosa F, Mazur M, Greenwald M, Van der Aa A, Vanhoutte F, Tasset C, Harrison P. Filgotinib (GLPG0634/GS-6034), an oral JAK1 selective inhibitor, is effective in combination with methotrexate (MTX) in patients with active rheumatoid arthritis and insufficient response to MTX: results from a randomised, dose-finding study (DARWIN 1). Ann Rheum Dis 2016; 76:998-1008. [PMID: 27993829 DOI: 10.1136/annrheumdis-2016-210104] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 11/01/2016] [Accepted: 11/05/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVES To evaluate the efficacy and safety of different doses and regimens of filgotinib, an oral Janus kinase 1 inhibitor, as add-on treatment to methotrexate (MTX) in patients with active rheumatoid arthritis (RA) and inadequate response to MTX. METHODS In this 24-week phase IIb study, patients with moderate-to-severe active RA receiving a stable dose of MTX were randomised (1:1:1:1:1:1:1) to receive placebo or 50, 100 or 200 mg filgotinib, administered once daily or twice daily. Primary end point was the percentage of patients achieving a week 12 American College of Rheumatology (ACR)20 response. RESULTS Overall, 594 patients were randomised and treated. At week 12, significantly more patients receiving filgotinib 100 mg once daily or 200 mg daily (both regimens) achieved an ACR20 response versus placebo. For other key end points at week 12 (ACR50, ACR-N, Disease Activity Score based on 28 joints and C reactive protein value, Clinical Disease Activity Index, Simplified Disease Activity Index and Health Assessment Questionnaire-Disability Index), differences in favour of 100 or 200 mg filgotinib daily were seen versus placebo; responses were maintained or improved through to week 24. Rapid onset of action and dose-dependent responses were observed for most efficacy end points and were associated with an increased haemoglobin concentration. No significant differences between once-daily and twice-daily regimens were seen. Treatment-emergent adverse event rates were similar in placebo and filgotinib groups. Serious infections occurred in one and five patients in the placebo and filgotinib groups, respectively. No tuberculosis or opportunistic infections were reported. CONCLUSIONS Filgotinib as add-on to MTX improved the signs and symptoms of active RA over 24 weeks and was associated with a rapid onset of action. Filgotinib was generally well tolerated. TRIAL REGISTRATION NUMBER NCT01888874.
Collapse
Affiliation(s)
- R Westhovens
- Department of Development and Regeneration KU Leuven, Skeletal Biology and Engineering Research Center, Leuven, Belgium.,Department of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - P C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - R Alten
- Schlosspark-Klinik Innere Medizin II, Berlin, Germany
| | | | | | - M Mazur
- IMSP Institul de Cardiologie, Chisinau, Moldova
| | - M Greenwald
- Desert Medical Advances, Palm Desert, California, USA
| | | | | | | | | |
Collapse
|
30
|
Moore CA, Iasella CJ, Venkataramanan R, Lakkis FG, Smith RB, McDyer JF, Zeevi A, Ensor CR. Janus kinase inhibition for immunosuppression in solid organ transplantation: Is there a role in complex immunologic challenges? Hum Immunol 2016; 78:64-71. [PMID: 27998802 DOI: 10.1016/j.humimm.2016.12.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/14/2016] [Accepted: 12/15/2016] [Indexed: 01/02/2023]
Abstract
Inhibition of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway for immunosuppression in solid organ transplantation is appealing due to its specificity for immune cell function, particularly for JAK3. This is due to its unique association with only the common gamma chain (γc). The γc is an appealing immunosuppression target in transplantation because of the critically important lymphokines that act at it, including IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Tofacitinib was initially purported to selectively inhibit solely JAK3, but subsequent analyses have also demonstrated its activity at the other members of the JAK family. Clinical outcomes have validated tofacitinib's pan-JAK activity in kidney transplantation after demonstrating an increased risk of infection and malignancy as compared to CNI-based regimens. After these trials, tofacitinib investigation for use in transplantation has effectively ceased. However, a post-hoc analysis has shed new light on the monitoring of tofacitinib exposure in order to predict infection and oncologic events. With new methods to monitor tofacitinib exposure, clinicians may be able to effectively reduce toxicities while providing a high level of immunosuppression. The purpose of this review to identify when, and for whom, JAK inhibitors may provide benefit in solid organ transplantation.
Collapse
Affiliation(s)
- Cody A Moore
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.
| | - Carlo J Iasella
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States; Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fadi G Lakkis
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Thomas E. Starzl Transplantation Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Randall B Smith
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - John F McDyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Adriana Zeevi
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Christopher R Ensor
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States; Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
31
|
Woon ST, Ameratunga R. Comprehensive genetic testing for primary immunodeficiency disorders in a tertiary hospital: 10-year experience in Auckland, New Zealand. Allergy Asthma Clin Immunol 2016; 12:65. [PMID: 27980540 PMCID: PMC5142146 DOI: 10.1186/s13223-016-0169-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 11/16/2016] [Indexed: 12/04/2022] Open
Abstract
Background and purpose New Zealand is a developed geographically isolated country in the South Pacific with a population of 4.4 million. Genetic diagnosis is the standard of care for most patients with primary immunodeficiency disorders (PIDs). Methods Since 2005, we have offered a comprehensive genetic testing service for PIDs and other immune-related disorders with a published sequence. Here we present results for this program, over the first decade, between 2005 and 2014. Results We undertook testing in 228 index cases and 32 carriers during this time. The three most common test requests were for X-linked lymphoproliferative (XLP), tumour necrosis factor receptor associated periodic syndrome (TRAPS) and haemophagocytic lymphohistiocytosis (HLH). Of the 32 suspected XLP cases, positive diagnoses were established in only 2 patients. In contrast, genetic defects in 8 of 11 patients with suspected X-linked agammaglobulinemia (XLA) were confirmed. Most XLA patients were initially identified from absence of B cells. Overall, positive diagnoses were made in about 23% of all tests requested. The diagnostic rate was lowest for several conditions with locus heterogeneity. Conclusions Thorough clinical characterisation of patients can assist in prioritising which genes should be tested. The clinician-driven customised comprehensive genetic service has worked effectively for New Zealand. Next generation sequencing will play an increasing role in disorders with locus heterogeneity.
Collapse
Affiliation(s)
- See-Tarn Woon
- Department of Virology and Immunology, LabPLUS, Auckland City Hospital, Grafton, Auckland, 1148 New Zealand
| | - Rohan Ameratunga
- Department of Virology and Immunology, LabPLUS, Auckland City Hospital, Grafton, Auckland, 1148 New Zealand
| |
Collapse
|
32
|
Moore JC, Tang Q, Yordán NT, Moore FE, Garcia EG, Lobbardi R, Ramakrishnan A, Marvin DL, Anselmo A, Sadreyev RI, Langenau DM. Single-cell imaging of normal and malignant cell engraftment into optically clear prkdc-null SCID zebrafish. J Exp Med 2016; 213:2575-2589. [PMID: 27810924 PMCID: PMC5110017 DOI: 10.1084/jem.20160378] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 09/16/2016] [Indexed: 12/03/2022] Open
Abstract
Cell transplantation into immunodeficient mice has revolutionized our understanding of regeneration, stem cell self-renewal, and cancer; yet models for direct imaging of engrafted cells has been limited. Here, we characterize zebrafish with mutations in recombination activating gene 2 (rag2), DNA-dependent protein kinase (prkdc), and janus kinase 3 (jak3). Histology, RNA sequencing, and single-cell transcriptional profiling of blood showed that rag2 hypomorphic mutant zebrafish lack T cells, whereas prkdc deficiency results in loss of mature T and B cells and jak3 in T and putative Natural Killer cells. Although all mutant lines engraft fluorescently labeled normal and malignant cells, only the prkdc mutant fish reproduced as homozygotes and also survived injury after cell transplantation. Engraftment into optically clear casper, prkdc-mutant zebrafish facilitated dynamic live cell imaging of muscle regeneration, repopulation of muscle stem cells within their endogenous niche, and muscle fiber fusion at single-cell resolution. Serial imaging approaches also uncovered stochasticity in fluorescently labeled leukemia regrowth after competitive cell transplantation into prkdc mutant fish, providing refined models to assess clonal dominance and progression in the zebrafish. Our experiments provide an optimized and facile transplantation model, the casper, prkdc mutant zebrafish, for efficient engraftment and direct visualization of fluorescently labeled normal and malignant cells at single-cell resolution.
Collapse
Affiliation(s)
- John C Moore
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Qin Tang
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Nora Torres Yordán
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Harvard University, Cambridge, MA 02138
| | - Finola E Moore
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Elaine G Garcia
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Riadh Lobbardi
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Ashwin Ramakrishnan
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| | - Dieuwke L Marvin
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
| | - Anthony Anselmo
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Ruslan I Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - David M Langenau
- Molecular Pathology, Massachusetts General Hospital, Charlestown, MA 02129
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Stem Cell Institute, Cambridge, MA 02139
| |
Collapse
|
33
|
Iwata S, Tanaka Y. Progress in understanding the safety and efficacy of Janus kinase inhibitors for treatment of rheumatoid arthritis. Expert Rev Clin Immunol 2016; 12:1047-57. [DOI: 10.1080/1744666x.2016.1189826] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
34
|
McNally R, Toms AV, Eck MJ. Crystal Structure of the FERM-SH2 Module of Human Jak2. PLoS One 2016; 11:e0156218. [PMID: 27227461 PMCID: PMC4881981 DOI: 10.1371/journal.pone.0156218] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 04/13/2016] [Indexed: 01/18/2023] Open
Abstract
Jak-family tyrosine kinases mediate signaling from diverse cytokine receptors. Binding of Jaks to their cognate receptors is mediated by their N-terminal region, which contains FERM and SH2 domains. Here we describe the crystal structure of the FERM-SH2 region of Jak2 at 3.0Å resolution. The structure reveals that these domains and their flanking linker segments interact intimately to form an integrated structural module. The Jak2 FERM-SH2 structure closely resembles that recently described for Tyk2, another member of the Jak family. While the overall architecture and interdomain orientations are preserved between Jak2 and Tyk2, we identify residues in the putative receptor-binding groove that differ between the two and may contribute to the specificity of receptor recognition. Analysis of Jak mutations that are reported to disrupt receptor binding reveals that they lie in the hydrophobic core of the FERM domain, and are thus expected to compromise the structural integrity of the FERM-SH2 unit. Similarly, analysis of mutations in Jak3 that are associated with severe combined immunodeficiency suggests that they compromise Jak3 function by destabilizing the FERM-SH2 structure.
Collapse
Affiliation(s)
- Randall McNally
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Angela V. Toms
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michael J. Eck
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
35
|
Sertori R, Liongue C, Basheer F, Lewis KL, Rasighaemi P, de Coninck D, Traver D, Ward AC. Conserved IL-2Rγc Signaling Mediates Lymphopoiesis in Zebrafish. THE JOURNAL OF IMMUNOLOGY 2015; 196:135-43. [PMID: 26590317 DOI: 10.4049/jimmunol.1403060] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 10/20/2015] [Indexed: 01/30/2023]
Abstract
The IL-2 receptor γ common (IL-2Rγc) chain is the shared subunit of the receptors for the IL-2 family of cytokines, which mediate signaling through JAK3 and various downstream pathways to regulate lymphopoiesis. Inactivating mutations in human IL-2Rγc result in SCID, a primary immunodeficiency characterized by greatly reduced numbers of lymphocytes. This study used bioinformatics, expression analysis, gene ablation, and specific pharmacologic inhibitors to investigate the function of two putative zebrafish IL-2Rγc paralogs, il-2rγc.a and il-2rγc.b, and downstream signaling components during early lymphopoiesis. Expression of il-2rγc.a commenced at 16 h post fertilization (hpf) and rose steadily from 4-6 d postfertilization (dpf) in the developing thymus, with il-2rγc.a expression also confirmed in adult T and B lymphocytes. Transcripts of il-2rγc.b were first observed from 8 hpf, but waned from 16 hpf before reaching maximal expression at 6 dpf, but this was not evident in the thymus. Knockdown of il-2rγc.a, but not il-2rγc.b, substantially reduced embryonic lymphopoiesis without affecting other aspects of hematopoiesis. Specific targeting of zebrafish Jak3 exerted a similar effect on lymphopoiesis, whereas ablation of zebrafish Stat5.1 and pharmacologic inhibition of PI3K and MEK also produced significant but smaller effects. Ablation of il-2rγc.a was further demonstrated to lead to an absence of mature T cells, but not B cells in juvenile fish. These results indicate that conserved IL-2Rγc signaling via JAK3 plays a key role during early zebrafish lymphopoiesis, which can be potentially targeted to generate a zebrafish model of human SCID.
Collapse
Affiliation(s)
- Robert Sertori
- School of Medicine, Deakin University, Geelong, Victoria 3216, Australia; Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria 3216, Australia
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, Victoria 3216, Australia; Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria 3216, Australia
| | - Faiza Basheer
- School of Medicine, Deakin University, Geelong, Victoria 3216, Australia; Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria 3216, Australia
| | - Kanako L Lewis
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093; and
| | - Parisa Rasighaemi
- School of Medicine, Deakin University, Geelong, Victoria 3216, Australia; Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria 3216, Australia
| | - Dennis de Coninck
- School of Medicine, Deakin University, Geelong, Victoria 3216, Australia; GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht 6200, the Netherlands
| | - David Traver
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093; and
| | - Alister C Ward
- School of Medicine, Deakin University, Geelong, Victoria 3216, Australia; Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria 3216, Australia;
| |
Collapse
|
36
|
Scarselli A, Di Cesare S, Di Matteo G, De Matteis A, Ariganello P, Romiti ML, Cascioli S, De Vito R, Bertaina A, Locatelli F, Gaspar HB, Aiuti A, Rossi P, Gilmour K, Cancrini C. Combined immunodeficiency due to JAK3 mutation in a child presenting with skin granuloma. J Allergy Clin Immunol 2015; 137:948-51.e5. [PMID: 26545580 DOI: 10.1016/j.jaci.2015.09.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 09/15/2015] [Accepted: 09/21/2015] [Indexed: 10/22/2022]
Affiliation(s)
- Alessia Scarselli
- University Department of Pediatrics, DPUO, Unit of Immune and Infectious Diseases, Bambino Gesù Children's Hospital, Rome, Italy.
| | - Silvia Di Cesare
- University Department of Pediatrics, DPUO, Unit of Immune and Infectious Diseases, Bambino Gesù Children's Hospital, Rome, Italy
| | - Gigliola Di Matteo
- University Department of Pediatrics, DPUO, Unit of Immune and Infectious Diseases, Bambino Gesù Children's Hospital, Rome, Italy
| | - Arianna De Matteis
- University Department of Pediatrics, DPUO, Unit of Immune and Infectious Diseases, Bambino Gesù Children's Hospital, Rome, Italy
| | - Paola Ariganello
- University Department of Pediatrics, DPUO, Unit of Immune and Infectious Diseases, Bambino Gesù Children's Hospital, Rome, Italy
| | - Maria Luisa Romiti
- University Department of Pediatrics, DPUO, Unit of Immune and Infectious Diseases, Bambino Gesù Children's Hospital, Rome, Italy
| | - Simona Cascioli
- Immunology and Pharmacotherapy Area, Bambino Gesù Children's Hospital, Rome, Italy
| | - Rita De Vito
- Department of Pathology, Bambino Gesù Children's Hospital, Rome, Italy
| | - Alice Bertaina
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Rome, Italy
| | - H Bobby Gaspar
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, London, United Kingdom
| | - Alessandro Aiuti
- University Department of Pediatrics, DPUO, Unit of Immune and Infectious Diseases, Bambino Gesù Children's Hospital, Rome, Italy; San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Scientific Institute HS Raffaele, Milan, Italy
| | - Paolo Rossi
- University Department of Pediatrics, DPUO, Unit of Immune and Infectious Diseases, Bambino Gesù Children's Hospital, Rome, Italy
| | - Kimberly Gilmour
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Caterina Cancrini
- University Department of Pediatrics, DPUO, Unit of Immune and Infectious Diseases, Bambino Gesù Children's Hospital, Rome, Italy.
| |
Collapse
|
37
|
Chang CW, Lai YS, Westin E, Khodadadi-Jamayran A, Pawlik K, Lamb L, Goldman F, Townes T. Modeling Human Severe Combined Immunodeficiency and Correction by CRISPR/Cas9-Enhanced Gene Targeting. Cell Rep 2015; 12:1668-77. [DOI: 10.1016/j.celrep.2015.08.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/02/2015] [Accepted: 08/04/2015] [Indexed: 12/21/2022] Open
|
38
|
Picard C, Moshous D, Fischer A. The Genetic and Molecular Basis of Severe Combined Immunodeficiency. CURRENT PEDIATRICS REPORTS 2014. [DOI: 10.1007/s40124-014-0070-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
39
|
Berg EL, O'Mahony A. Complex Primary Human Cell Systems for Drug Discovery. HUMAN-BASED SYSTEMS FOR TRANSLATIONAL RESEARCH 2014. [DOI: 10.1039/9781782620136-00088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Phenotypic or biofunctional assays play an important role in drug discovery by helping to bridge the gap between high-throughput, target-based screening assays used for compound identification and more physiologically relevant in vivo disease models used for preclinical development. We have developed a standardised panel of phenotypic assays using primary human cells and co-cultures that model tissue and disease biology for characterization of drug leads. Here we show application of these assays for characterisation of clinical stage kinase inhibitors for rheumatoid arthritis, the recently approved JAK kinase inhibitor, tofacitinib, and the SYK kinase inhibitor, fostamatinib. We demonstrate how profiling in this assay panel can relate to clinical effects, both efficacy and safety related.
Collapse
Affiliation(s)
- Ellen L. Berg
- BioSeek, A Division of DiscoveRx 310 Utah Avenue Suite 100 South San Francisco CA 94080 USA
| | - Alison O'Mahony
- BioSeek, A Division of DiscoveRx 310 Utah Avenue Suite 100 South San Francisco CA 94080 USA
| |
Collapse
|
40
|
Combined immunodeficiency evolving into predominant CD4+ lymphopenia caused by somatic chimerism in JAK3. J Clin Immunol 2014; 34:941-53. [PMID: 25205547 PMCID: PMC4220108 DOI: 10.1007/s10875-014-0088-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 08/13/2014] [Indexed: 12/21/2022]
Abstract
Purpose Idiopathic CD4 lymphopenia constitutes a heterogeneous group of immunodeficiencies with characteristically low CD4+ T-cell counts with largely unknown genetic etiology. We here sought to determine the underlying molecular cause in an index family with two patients suffering from combined immunodeficiency that evolved into predominant CD4+ lymphopenia. The more severely affected index patient also presented with selective antibody deficiency against bacterial polysaccharide antigens. Methods For the genetic analysis, we used combined homozygosity mapping and exome sequencing. Functional assays included immunoblot analysis, flow cytometry and TCR Vβ spectratyping. Results A novel homozygous missense mutation was revealed in the kinase domain of JAK3 (c.T3196C, p.Cys1066Arg). Further analysis showed revertant chimerism in CD8+ T-cells in both patients. The additional presence of revertant CD4+ T-cells was associated with a milder clinical and immunological phenotype in the second patient, although the role somatic chimerism plays in amelioration of disease phenotype is uncertain, as presence of revertant cells had no effect on residual CD4 cell JAK3 signaling function. Residual activity of JAK3-dependent STAT3 and STAT5 signaling was also found in immortalized B-cell lines indicating a hypomorphic nature of the described mutation which likely contributes to the milder clinical phenotype. Conclusions We here present the first case of revertant mosaicism in JAK3 deficiency, manifesting as combined immunodeficiency evolving into predominant CD4+ lymphopenia. Revertant chimerism or hypomorphic mutations in genes typically associated with more severe T-cell deficiency should be considered when assessing patients with milder forms of combined immunodeficiencies. Electronic supplementary material The online version of this article (doi:10.1007/s10875-014-0088-2) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Thiophene carboxamide inhibitors of JAK2 as potential treatments for myleoproliferative neoplasms. Bioorg Med Chem Lett 2014; 24:1968-73. [DOI: 10.1016/j.bmcl.2014.02.064] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 01/30/2023]
|
42
|
Edgar JDM, Buckland M, Guzman D, Conlon NP, Knerr V, Bangs C, Reiser V, Panahloo Z, Workman S, Slatter M, Gennery AR, Davies EG, Allwood Z, Arkwright PD, Helbert M, Longhurst HJ, Grigoriadou S, Devlin LA, Huissoon A, Krishna MT, Hackett S, Kumararatne DS, Condliffe AM, Baxendale H, Henderson K, Bethune C, Symons C, Wood P, Ford K, Patel S, Jain R, Jolles S, El-Shanawany T, Alachkar H, Herwadkar A, Sargur R, Shrimpton A, Hayman G, Abuzakouk M, Spickett G, Darroch CJ, Paulus S, Marshall SE, McDermott EM, Heath PT, Herriot R, Noorani S, Turner M, Khan S, Grimbacher B. The United Kingdom Primary Immune Deficiency (UKPID) Registry: report of the first 4 years' activity 2008-2012. Clin Exp Immunol 2014; 175:68-78. [PMID: 23841717 PMCID: PMC3898556 DOI: 10.1111/cei.12172] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2013] [Indexed: 12/11/2022] Open
Abstract
This report summarizes the establishment of the first national online registry of primary immune deficency in the United Kingdom, the United Kingdom Primary Immunodeficiency (UKPID Registry). This UKPID Registry is based on the European Society for Immune Deficiency (ESID) registry platform, hosted on servers at the Royal Free site of University College, London. It is accessible to users through the website of the United Kingdom Primary Immunodeficiency Network (www.ukpin.org.uk). Twenty-seven centres in the United Kingdom are actively contributing data, with an additional nine centres completing their ethical and governance approvals to participate. This indicates that 36 of 38 (95%) of recognized centres in the United Kingdom have engaged with this project. To date, 2229 patients have been enrolled, with a notable increasing rate of recruitment in the past 12 months. Data are presented on the range of diagnoses recorded, estimated minimum disease prevalence, geographical distribution of patients across the United Kingdom, age at presentation, diagnostic delay, treatment modalities used and evidence of their monitoring and effectiveness.
Collapse
Affiliation(s)
- J D M Edgar
- Regional Immunology Service, The Royal Hospitals, Belfast, East Yorkshire; Centre for Infection and Immunity, Queen's University Belfast, Belfast, East Yorkshire
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yamaoka K, Tanaka Y. Targeting the Janus kinases in rheumatoid arthritis: focus on tofacitinib. Expert Opin Pharmacother 2013; 15:103-13. [DOI: 10.1517/14656566.2014.854771] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
44
|
Plenge RM, Scolnick EM, Altshuler D. Validating therapeutic targets through human genetics. Nat Rev Drug Discov 2013; 12:581-94. [PMID: 23868113 DOI: 10.1038/nrd4051] [Citation(s) in RCA: 447] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
More than 90% of the compounds that enter clinical trials fail to demonstrate sufficient safety and efficacy to gain regulatory approval. Most of this failure is due to the limited predictive value of preclinical models of disease, and our continued ignorance regarding the consequences of perturbing specific targets over long periods of time in humans. 'Experiments of nature' - naturally occurring mutations in humans that affect the activity of a particular protein target or targets - can be used to estimate the probable efficacy and toxicity of a drug targeting such proteins, as well as to establish causal rather than reactive relationships between targets and outcomes. Here, we describe the concept of dose-response curves derived from experiments of nature, with an emphasis on human genetics as a valuable tool to prioritize molecular targets in drug development. We discuss empirical examples of drug-gene pairs that support the role of human genetics in testing therapeutic hypotheses at the stage of target validation, provide objective criteria to prioritize genetic findings for future drug discovery efforts and highlight the limitations of a target validation approach that is anchored in human genetics.
Collapse
Affiliation(s)
- Robert M Plenge
- Division of Rheumatology, Immunology and Allergy, Brigham And Women's Hospital, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
45
|
Cattaneo F, Recher M, Masneri S, Baxi SN, Fiorini C, Antonelli F, Wysocki CA, Calderon JG, Eibel H, Smith AR, Bonilla FA, Tsitsikov E, Giliani S, Notarangelo LD, Pai SY. Hypomorphic Janus kinase 3 mutations result in a spectrum of immune defects, including partial maternal T-cell engraftment. J Allergy Clin Immunol 2013; 131:1136-45. [PMID: 23384681 DOI: 10.1016/j.jaci.2012.12.667] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 10/23/2012] [Accepted: 12/07/2012] [Indexed: 12/31/2022]
Abstract
BACKGROUND Mutations in Janus kinase 3 (JAK3) are a cause of severe combined immunodeficiency, but hypomorphic JAK3 defects can result in a milder clinical phenotype, with residual development and function of autologous T cells. Maternal T-cell engraftment is a common finding in infants with severe combined immunodeficiency but is not typically observed in patients with residual T-cell development. OBJECTIVE We sought to study in detail the molecular, cellular, and humoral immune phenotype and function of 3 patients with hypomorphic JAK3 mutations. METHODS We analyzed the distribution and function of T and B lymphocytes in 3 patients and studied the in vitro and in vivo responses of maternal T lymphocytes in 1 patient with maternal T-cell engraftment and residual production of autologous T lymphocytes. RESULTS B cells were present in normal numbers but with abnormal distribution of marginal zone-like and memory B cells. B-cell differentiation to plasmablasts in vitro in response to CD40 ligand and IL-21 was abolished. In 2 patients the T-cell repertoire was moderately restricted. Surprisingly, 1 patient showed coexistence of maternal and autologous T lymphocytes. By using an mAb recognizing the maternal noninherited HLA-A2 antigen, we found that autologous cells progressively accumulated in vivo but did not compete with maternal cells in vitro. CONCLUSION The study of 3 patients with hypomorphic JAK3 mutations suggests that terminal B-cell maturation/differentiation requires intact JAK3 function, even if partially functioning T lymphocytes are present. Maternal T-cell engraftment can occur in patients with JAK3 mutations despite the presence of autologous T cells.
Collapse
Affiliation(s)
- Federica Cattaneo
- Division of Hematology-Oncology, the Manton Center for Orphan Disease Research, Department of Laboratory Medicine, Boston Children's Hospital, and Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Koo GC, Tan SY, Tang T, Poon SL, Allen GE, Tan L, Chong SC, Ong WS, Tay K, Tao M, Quek R, Loong S, Yeoh KW, Yap SP, Lee KA, Lim LC, Tan D, Goh C, Cutcutache I, Yu W, Ng CCY, Rajasegaran V, Heng HL, Gan A, Ong CK, Rozen S, Tan P, Teh BT, Lim ST. Janus kinase 3-activating mutations identified in natural killer/T-cell lymphoma. Cancer Discov 2012; 2:591-7. [PMID: 22705984 DOI: 10.1158/2159-8290.cd-12-0028] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED The molecular pathogenesis of natural killer/T-cell lymphoma (NKTCL) is not well understood. We conducted whole-exome sequencing and identified Janus kinase 3 (JAK3) somatic-activating mutations (A572V and A573V) in 2 of 4 patients with NKTCLs. Further validation of the prevalence of JAK3 mutations was determined by Sanger sequencing and high-resolution melt (HRM) analysis in an additional 61 cases. In total, 23 of 65 (35.4%) cases harbored JAK3 mutations. Functional characterization of the JAK3 mutations support its involvement in cytokine-independent JAK/STAT constitutive activation leading to increased cell growth. Moreover, treatment of both JAK3-mutant and wild-type NKTCL cell lines with a novel pan-JAK inhibitor, CP-690550, resulted in dose-dependent reduction of phosphorylated STAT5, reduced cell viability, and increased apoptosis. Hence, targeting the deregulated JAK/STAT pathway could be a promising therapy for patients with NKTCLs. SIGNIFICANCE Gene mutations causing NKTCL have not been fully identified. Through exome sequencing, we identified activating mutations of JAK3 that may play a significant role in the pathogenesis of NKTCLs. Our findings have important implications for the management of patients with NKTCLs.
Collapse
Affiliation(s)
- Ghee Chong Koo
- NCCS-VARI Translational Research Laboratory, Department of Medical Sciences, National Cancer Centre Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Bongfen SE, Rodrigue-Gervais IG, Berghout J, Torre S, Cingolani P, Wiltshire SA, Leiva-Torres GA, Letourneau L, Sladek R, Blanchette M, Lathrop M, Behr MA, Gruenheid S, Vidal SM, Saleh M, Gros P. An N-ethyl-N-nitrosourea (ENU)-induced dominant negative mutation in the JAK3 kinase protects against cerebral malaria. PLoS One 2012; 7:e31012. [PMID: 22363534 PMCID: PMC3283600 DOI: 10.1371/journal.pone.0031012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 12/29/2011] [Indexed: 11/19/2022] Open
Abstract
Cerebral malaria (CM) is a lethal neurological complication of malaria. We implemented a genome-wide screen in mutagenized mice to identify host proteins involved in CM pathogenesis and whose inhibition may be of therapeutic value. One pedigree (P48) segregated a resistance trait whose CM-protective effect was fully penetrant, mapped to chromosome 8, and identified by genome sequencing as homozygosity for a mis-sense mutation (W81R) in the FERM domain of Janus-associated kinase 3 (Jak3). The causative effect of Jak3(W81R) was verified by complementation testing in Jak3(W81R/-) double heterozygotes that were fully protected against CM. Jak3(W81R) homozygotes showed defects in thymic development with depletion of CD8(+) T cell, B cell, and NK cell compartments, and defective T cell-dependent production of IFN-γ. Adoptive transfer of normal splenocytes abrogates CM resistance in Jak3(W81R) homozygotes, an effect attributed to the CD8(+) T cells. Jak3(W81R) behaves as a dominant negative variant, with significant CM resistance of Jak3(W81R/+) heterozygotes, compared to CM-susceptible Jak3(+/+) and Jak3(+/-) controls. CM resistance in Jak3(W81R/+) heterozygotes occurs in presence of normal T, B and NK cell numbers. These findings highlight the pathological role of CD8(+) T cells and Jak3-dependent IFN-γ-mediated Th1 responses in CM pathogenesis.
Collapse
Affiliation(s)
- Silayuv E. Bongfen
- Department of Biochemistry, McGill University, Montreal, Canada
- Complex Traits Group, McGill University, Montreal, Canada
| | - Ian-Gael Rodrigue-Gervais
- Department of Medicine, McGill University, Montreal, Canada
- Complex Traits Group, McGill University, Montreal, Canada
| | - Joanne Berghout
- Department of Biochemistry, McGill University, Montreal, Canada
- Complex Traits Group, McGill University, Montreal, Canada
| | - Sabrina Torre
- Department of Human Genetics, McGill University, Montreal, Canada
- Complex Traits Group, McGill University, Montreal, Canada
| | - Pablo Cingolani
- School of Computer Science, McGill University, Montreal, Canada
| | - Sean A. Wiltshire
- Department of Human Genetics, McGill University, Montreal, Canada
- Complex Traits Group, McGill University, Montreal, Canada
| | - Gabriel A. Leiva-Torres
- Department of Human Genetics, McGill University, Montreal, Canada
- Complex Traits Group, McGill University, Montreal, Canada
| | - Louis Letourneau
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Robert Sladek
- Department of Human Genetics, McGill University, Montreal, Canada
| | | | - Mark Lathrop
- Institut de Génomique, Centre National de Génotypage, Evry, France
| | - Marcel A. Behr
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- The McGill University Health Center, Montreal, Canada
| | - Samantha Gruenheid
- Complex Traits Group, McGill University, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Silvia M. Vidal
- Department of Human Genetics, McGill University, Montreal, Canada
- Complex Traits Group, McGill University, Montreal, Canada
| | - Maya Saleh
- Department of Medicine, McGill University, Montreal, Canada
- Complex Traits Group, McGill University, Montreal, Canada
| | - Philippe Gros
- Department of Biochemistry, McGill University, Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
- Complex Traits Group, McGill University, Montreal, Canada
- * E-mail:
| |
Collapse
|
48
|
Aloj G, Giardino G, Valentino L, Maio F, Gallo V, Esposito T, Naddei R, Cirillo E, Pignata C. Severe Combined Immunodeficiences: New and Old Scenarios. Int Rev Immunol 2012; 31:43-65. [DOI: 10.3109/08830185.2011.644607] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
49
|
Yu GP, Nadeau KC, Berk DR, de Saint Basile G, Lambert N, Knapnougel P, Roberts J, Kavanau K, Dunn E, Stiehm ER, Lewis DB, Umetsu DT, Puck JM, Cowan MJ. Genotype, phenotype, and outcomes of nine patients with T-B+NK+ SCID. Pediatr Transplant 2011; 15:733-41. [PMID: 21883749 PMCID: PMC3196791 DOI: 10.1111/j.1399-3046.2011.01563.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There are few reports of clinical presentation, genotype, and HCT outcomes for patients with T-B+NK+ SCID. Between 1981 and 2007, eight of 84 patients with SCID who received and/or were followed after HCT at UCSF had the T-B+NK+ phenotype. One additional patient with T-B+NK+ SCID was identified as the sibling of a patient treated at UCSF. Chart reviews were performed. Molecular analyses of IL7R, IL2RG, JAK3, and the genes encoding the CD3 T-cell receptor components δ (CD3D), ε (CD3E), and ζ (CD3Z) were carried out. IL7R mutations were documented in four patients and CD3D mutations in two others. Three patients had no defects found. Only two of nine patients had an HLA-matched related HCT donor. Both survived, and neither developed GVHD. Five of seven recipients of haploidentical grafts survived. Although the majority of reported cases of T-B+NK+ SCID are caused by defects in IL7R, CD3 complex defects were also found in this series and should be considered when evaluating patients with T-B+NK+ SCID. Additional genes, mutations in which account for T-B+NK+ SCID, remain to be found. Better approaches to early diagnosis and HCT treatment are needed for patients lacking an HLA-matched related donor.
Collapse
Affiliation(s)
- Grace P Yu
- Division of Immunology and Allergy, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital at Stanford
| | - Kari C Nadeau
- Division of Immunology and Allergy, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital at Stanford
| | - David R Berk
- Departments of Medicine and Pediatrics, Divisions of Dermatology, Washington University School of Medicine
| | - Geneviève de Saint Basile
- Inserm, U768, Paris, F-75015 France,Université Paris Descartes, IRNEM (IFR95), Paris, F-75015 France,AP-HP, Hôpital Necker Enfants-Malades, Unité d'Immunologie-Hématologie Pédiatrique, Paris, F-75015 France
| | - Nathalie Lambert
- AP-HP, Hôpital Necker Enfants-Malades, Unité d'Immunologie-Hématologie Pédiatrique, Paris, F-75015 France
| | | | - Joseph Roberts
- Department of Pediatrics and Immunology, Duke University Medical Center
| | - Kristina Kavanau
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco Children's Hospital
| | - Elizabeth Dunn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco Children's Hospital
| | - E. Richard Stiehm
- Divison of Immunology, Allergy and Rheumatology, Department of Pediatrics, Mattel Children's Hospital at the University of California Los Angeles
| | - David B Lewis
- Division of Immunology and Allergy, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital at Stanford
| | - Dale T Umetsu
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital Boston
| | - Jennifer M Puck
- Department of Pediatrics, Institute for Human Genetics, University of California San Francisco Children's Hospital
| | - Morton J Cowan
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco Children's Hospital
| |
Collapse
|
50
|
Alicea-Velázquez NL, Boggon TJ. The use of structural biology in Janus kinase targeted drug discovery. Curr Drug Targets 2011; 12:546-55. [PMID: 21126226 DOI: 10.2174/138945011794751528] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 10/04/2010] [Indexed: 12/12/2022]
Abstract
The Janus kinases (or Jak kinases) mediate cytokine and growth factor signal transduction. Acquired or inherited Jak mutations can result in dysregulation of Jak-mediated signal transduction and can be critical to disease acquisition in neoplasias including acute myeloid, acute lymphoblastic and acute megakaryoblastic leukemias, and in rare X-linked severe combined immunodeficiency. The discovery of an acquired Jak2 point mutation, V617F, in significant numbers of patients with classical myeloproliferative disorders has increased the interest in development of Jak2-specific tyrosine kinase inhibitors and consequently there are now over 20 publically available structures of Jak kinase domains that describe all four family members, Jak1, Jak2, Jak3, and Tyk2. Here we review the recent advances in understanding the druggable structure and function of the Jak family, with a focus on the structural biology of the Jak kinase domain. We will discuss how these advances impact the development of Jak-targeted therapeutics.
Collapse
Affiliation(s)
- Nilda L Alicea-Velázquez
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar St., SHM B-316A, New Haven, CT 06520, USA
| | | |
Collapse
|