1
|
Mazur A, Skrzeczynska-Moncznik J, Mavroudi I, Perraki CM, Korkmaz B, Papadaki HA, Cichy J. Neutrophils from patients with acquired neutropenia exhibit alterations in serine protease immunostaining and activity. Br J Haematol 2024. [PMID: 39359123 DOI: 10.1111/bjh.19815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Affiliation(s)
- Angelika Mazur
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Joanna Skrzeczynska-Moncznik
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Irene Mavroudi
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, Heraklion, Greece
- Department of Hematology, University Hospital of Heraklion, Heraklion, Greece
| | - Christina Maria Perraki
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, Heraklion, Greece
- Department of Hematology, University Hospital of Heraklion, Heraklion, Greece
| | - Brice Korkmaz
- INSERM, Respiratory Disease Research Centre, U1100, Tours, France
- University of Tours, Tours, France
| | - Helen A Papadaki
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, Heraklion, Greece
- Department of Hematology, University Hospital of Heraklion, Heraklion, Greece
| | - Joanna Cichy
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
2
|
Shu Z, Deng M, Han T, Mao H. De Novo Deep Intron ELANE Mutation Resulting in Severe Congenital Neutropenia. J Clin Immunol 2024; 44:183. [PMID: 39172268 DOI: 10.1007/s10875-024-01783-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024]
Abstract
Severe congenital neutropenia (SCN) comprises a diverse range of rare hematological disorders characterized by recurrent, often life-threatening infections that manifest within the first months of life. Mutations in the ELANE gene are the most prevalent cause of SCN. While over 230 mutations in ELANE have been documented, including substitutions, frameshifts, nonsense mutations, and splice site alterations, the occurrence of deep intronic mutations has not been previously reported. Herein, we present the case of a young girl who exhibited recurrent fever, respiratory infections, skin abscesses, and gingivitis shortly after birth. Laboratory analysis revealed markedly diminished neutrophil levels alongside elevated monocyte and eosinophil counts. Bone marrow examination disclosed a halt in myelopoiesis maturation. ELANE gene full-length sequencing identified a novel de novo deep intron mutation in ELANE (c.598 + 79G > T), subsequently confirmed by Sanger sequencing. cDNA sequencing of the patient demonstrated aberrant gene splicing. Utilizing a mini-gene splicing assay for ELANE intronic variants, we identified a mutant ELANE allele (c.597 + 1_597 + 83ins) leading to the creation of a premature termination codon (p.Gly200ValfsTer40). Confocal microscopy revealed heightened expression of myeloperoxidase and neutrophil elastase in the patient, suggesting a potential role for the unfolded protein response in the pathogenesis of the deep intron ELANE mutation. In summary, our findings illustrate the first reported instance of de novo deep intron ELANE mutations associated with SCN, underscoring the importance of exploring deep intronic regions in SCN patients lacking identifiable disease-causing gene mutations.
Collapse
Affiliation(s)
- Zhou Shu
- Department of Immunology, Ministry of Education Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China
| | - Mengyue Deng
- Department of Immunology, Ministry of Education Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China
| | - Tongxin Han
- Department of Immunology, Ministry of Education Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China
| | - Huawei Mao
- Department of Immunology, Ministry of Education Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nanlishi Road, Beijing, 100045, China.
- Beijing Key Laboratory for Genetics of Birth Defects, No. 56 Nanlishi Road, Beijing, 100045, China.
| |
Collapse
|
3
|
Doll L, Welte K, Skokowa J, Bajoghli B. A JAGN1-associated severe congenital neutropenia zebrafish model revealed an altered G-CSFR signaling and UPR activation. Blood Adv 2024; 8:4050-4065. [PMID: 38739706 PMCID: PMC11342096 DOI: 10.1182/bloodadvances.2023011656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
ABSTRACT A variety of autosomal recessive mutations in the JAGN1 gene cause severe congenital neutropenia (CN). However, the underlying pathomechanism remains poorly understood, mainly because of the limited availability of primary hematopoietic stem cells from JAGN1-CN patients and the absence of animal models. In this study, we aimed to address these limitations by establishing a zebrafish model of JAGN1-CN. We found 2 paralogs of the human JAGN1 gene, namely jagn1a and jagn1b, which play distinct roles during zebrafish hematopoiesis. Using various approaches such as morpholino-based knockdown, CRISPR/Cas9-based gene editing, and misexpression of a jagn1b harboring a specific human mutation, we successfully developed neutropenia while leaving other hematopoietic lineages unaffected. Further analysis of our model revealed significant upregulation of apoptosis and genes involved in the unfolded protein response (UPR). However, neither UPR nor apoptosis is the primary mechanism that leads to neutropenia in zebrafish. Instead, Jagn1b has a critical role in granulocyte colony-stimulating factor receptor signaling and steady-state granulopoiesis, shedding light on the pathogenesis of neutropenia associated with JAGN1 mutations. The establishment of a zebrafish model for JAGN1-CN represents a significant advancement in understanding the specific pathologic pathways underlying the disease. This model provides a valuable in vivo tool for further investigation and exploration of potential therapeutic strategies.
Collapse
Affiliation(s)
- Larissa Doll
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Karl Welte
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Children’s Hospital, University Hospital Tuebingen, Tuebingen, Germany
| | - Julia Skokowa
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
- Gene and RNA Therapy Center, Tuebingen University, Tuebingen, Germany
| | - Baubak Bajoghli
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
- Austrian BioImaging/CMI, Vienna, Austria
| |
Collapse
|
4
|
Nasri M, Ritter MU, Mir P, Dannenmann B, Kaufmann MM, Arreba-Tutusaus P, Xu Y, Borbaran-Bravo N, Klimiankou M, Lengerke C, Zeidler C, Cathomen T, Welte K, Skokowa J. CRISPR-Cas9n-mediated ELANE promoter editing for gene therapy of severe congenital neutropenia. Mol Ther 2024; 32:1628-1642. [PMID: 38556793 PMCID: PMC11184331 DOI: 10.1016/j.ymthe.2024.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/07/2023] [Accepted: 03/28/2024] [Indexed: 04/02/2024] Open
Abstract
Severe congenital neutropenia (CN) is an inherited pre-leukemia bone marrow failure syndrome commonly caused by autosomal-dominant ELANE mutations (ELANE-CN). ELANE-CN patients are treated with daily injections of recombinant human granulocyte colony-stimulating factor (rhG-CSF). However, some patients do not respond to rhG-CSF, and approximately 15% of ELANE-CN patients develop myelodysplasia or acute myeloid leukemia. Here, we report the development of a curative therapy for ELANE-CN through inhibition of ELANE mRNA expression by introducing two single-strand DNA breaks at the opposing DNA strands of the ELANE promoter TATA box using CRISPR-Cas9D10A nickases-termed MILESTONE. This editing effectively restored defective neutrophil differentiation of ELANE-CN CD34+ hematopoietic stem and progenitor cells (HSPCs) in vitro and in vivo, without affecting the functions of the edited neutrophils. CRISPResso analysis of the edited ELANE-CN CD34+ HSPCs revealed on-target efficiencies of over 90%. Simultaneously, GUIDE-seq, CAST-Seq, and rhAmpSeq indicated a safe off-target profile with no off-target sites or chromosomal translocations. Taken together, ex vivo gene editing of ELANE-CN HSPCs using MILESTONE in the setting of autologous stem cell transplantation could be a universal, safe, and efficient gene therapy approach for ELANE-CN patients.
Collapse
Affiliation(s)
- Masoud Nasri
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany.
| | - Malte U Ritter
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany.
| | - Perihan Mir
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Benjamin Dannenmann
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Masako M Kaufmann
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106 Freiburg, Germany; Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Spemann Graduate School of Biology and Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Patricia Arreba-Tutusaus
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Yun Xu
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Natalia Borbaran-Bravo
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Maksim Klimiankou
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Claudia Lengerke
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Cornelia Zeidler
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; Pediatric Hematology and Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106 Freiburg, Germany; Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Karl Welte
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Children`s Hospital, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Julia Skokowa
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, 72076 Tübingen, Germany; Gene and RNA Therapy Center (GRTC), University Hospital Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
5
|
Xiao Y, Wang N, Jin X, Liu A, Zhang Z. Clinical relevance of SCN and CyN induced by ELANE mutations: a systematic review. Front Immunol 2024; 15:1349919. [PMID: 38840904 PMCID: PMC11150597 DOI: 10.3389/fimmu.2024.1349919] [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: 12/05/2023] [Accepted: 04/29/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction According to the PRISMA criteria, a systematic review has been conducted to investigate the clinical relevance between patients with severe congenital neutropenia (SCN) and cyclic congenital neutropenia (CyN) induced by ELANE mutations. Methods We have searched PubMed, EMBASE, Web of Science, Scopus, Cochrane, CNKI, Wanfang Medicine, and VIP for ELANE mutation related literature published from 1997 to 2022. Using Microsoft Excel collect and organize data, SPSS 25, GraphPad Prism 8.0.1, and Omap analyze and plot statistical. Compare the gender, age, geography, mutation sites, infection characteristics, treatment, and other factors of SCN and CyN patients induced by ELANE mutations, with a focus on exploring the relationship between genotype and clinical characteristics, genotype and prognosis. Results This study has included a total of 467 patients with SCN and 90 patients with CyN. The onset age of SCN and CyN are both less than 1 year old, and the onset and diagnosis age of SCN are both younger than CyN. The mutation of ELANE gene is mainly missense mutation, and hot spot mutations include S126L, P139L, G214R, c.597+1G>A. The high-frequency mutations with severe outcomes are A57V, L121H, L121P, c.597+1G>A, c.597+1G>T, S126L, C151Y, C151S, G214R, C223X. Respiratory tract, skin and mucosa are the most common infection sites, Staphylococcus aureus, Pseudomonas aeruginosa and Escherichia coli are the most common. Discussion Patients with refractory G-CSF are more likely to develop severe outcomes. The commonly used pre-treatment schemes for transplantation are Bu-Cy-ATG and Flu-Bu-ATG. The prognosis of transplantation is mostly good, but the risk of GVHD is high. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/. PROSPERO, identifier CRD42023434656.
Collapse
Affiliation(s)
- Yufan Xiao
- Department of Rheumatology and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Nandi Wang
- Department of Rheumatology and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xinghao Jin
- Department of Rheumatology and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Anna Liu
- Department of Rheumatology and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiyong Zhang
- Department of Rheumatology and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Caglayan M, Ozden S. Potential impacts of bisphenols on prostate cells: An overview of cytotoxicity, proliferation, oxidative stress, apoptosis, and ER-stress response activation. Food Chem Toxicol 2024; 184:114416. [PMID: 38134982 DOI: 10.1016/j.fct.2023.114416] [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: 09/07/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
This study aimed to evaluate the toxic effects of Bisphenol A (BPA), Bisphenol F (BPF) and Bisphenol S (BPS) on PNT1A and PC-3 cells, focusing on their effects on endoplasmic reticulum (ER) stress and related pathways. PNT1A and PC-3 were treated with BPA, BPF and BPS at concentrations of 0.1, 1 and 10 μM for 48 h cytotoxicity, BrdU cell proliferation, ROS generation, apoptosis detection, gene expression analysis and Western blot analysis were performed. BPA induced proliferation and late apoptosis in PNT1A cells, whereas it induced both late apoptosis and early apoptosis in PC-3 cells. BPF and BPS induced late apoptosis in PC-3 cells. Increased ROS levels were observed in PNT1A cells exposed to 1-10 μM BPA. BPA, BPF and BPS increased the expression levels of ER stress-related genes in PNT1A cells. Furthermore, exposure to BPA increased the expression of ER stress-related CHOP/DDIT3 protein in PNT1A cells. These findings highlight the potential health risks associated with BPA, BPF and BPS exposure and emphasize the importance of investigating the underlying mechanisms by which these chemicals may affect human health. Further research is required to comprehensively understand the role of ER stress pathways in cellular responses to these substances.
Collapse
Affiliation(s)
- Mine Caglayan
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, İstanbul University, Istanbul, Turkey; Institute of Graduate Studies in Health Sciences, Istanbul University, Istanbul, Turkey; Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Biruni University, Istanbul, Turkey
| | - Sibel Ozden
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, İstanbul University, Istanbul, Turkey.
| |
Collapse
|
7
|
Oyarbide U, Crane GM, Corey SJ. The metabolic basis of inherited neutropenias. Br J Haematol 2024; 204:45-55. [PMID: 38049194 DOI: 10.1111/bjh.19192] [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: 08/29/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 12/06/2023]
Abstract
Neutrophils are the shortest-lived blood cells, which requires a prodigious degree of proliferation and differentiation to sustain physiologically sufficient numbers and be poised to respond quickly to infectious emergencies. More than 107 neutrophils are produced every minute in an adult bone marrow-a process that is tightly regulated by a small group of cytokines and chemical mediators and dependent on nutrients and energy. Like granulocyte colony-stimulating factor, the primary growth factor for granulopoiesis, they stimulate signalling pathways, some affecting metabolism. Nutrient or energy deficiency stresses the survival, proliferation, and differentiation of neutrophils and their precursors. Thus, it is not surprising that monogenic disorders related to metabolism exist that result in neutropenia. Among these are pathogenic mutations in HAX1, G6PC3, SLC37A4, TAFAZZIN, SBDS, EFL1 and the mitochondrial disorders. These mutations perturb carbohydrate, lipid and/or protein metabolism. We hypothesize that metabolic disturbances may drive the pathogenesis of a subset of inherited neutropenias just as defects in DNA damage response do in Fanconi anaemia, telomere maintenance in dyskeratosis congenita and ribosome formation in Diamond-Blackfan anaemia. Greater understanding of metabolic pathways in granulopoiesis will identify points of vulnerability in production and may point to new strategies for the treatment of neutropenias.
Collapse
Affiliation(s)
- Usua Oyarbide
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Pediatrics, Cleveland Clinic, Cleveland, Ohio, USA
| | - Genevieve M Crane
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Seth J Corey
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Pediatrics, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
8
|
Neutropenia: diagnosis and management. World J Pediatr 2022; 18:771-777. [PMID: 35962272 DOI: 10.1007/s12519-022-00593-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/05/2022] [Indexed: 10/15/2022]
|
9
|
CLPB in neutropenia: 1 gene with many faces. Blood 2022; 139:649-650. [PMID: 35113147 DOI: 10.1182/blood.2021012606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 11/20/2022] Open
|
10
|
Warren JT, Cupo RR, Wattanasirakul P, Spencer DH, Locke AE, Makaryan V, Bolyard AA, Kelley ML, Kingston NL, Shorter J, Bellanné-Chantelot C, Donadieu J, Dale DC, Link DC. Heterozygous variants of CLPB are a cause of severe congenital neutropenia. Blood 2022; 139:779-791. [PMID: 34115842 PMCID: PMC8814677 DOI: 10.1182/blood.2021010762] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/19/2021] [Indexed: 02/05/2023] Open
Abstract
Severe congenital neutropenia is an inborn disorder of granulopoiesis. Approximately one third of cases do not have a known genetic cause. Exome sequencing of 104 persons with congenital neutropenia identified heterozygous missense variants of CLPB (caseinolytic peptidase B) in 5 severe congenital neutropenia cases, with 5 more cases identified through additional sequencing efforts or clinical sequencing. CLPB encodes an adenosine triphosphatase that is implicated in protein folding and mitochondrial function. Prior studies showed that biallelic mutations of CLPB are associated with a syndrome of 3-methylglutaconic aciduria, cataracts, neurologic disease, and variable neutropenia. However, 3-methylglutaconic aciduria was not observed and, other than neutropenia, these clinical features were uncommon in our series. Moreover, the CLPB variants are distinct, consisting of heterozygous variants that cluster near the adenosine triphosphate-binding pocket. Both genetic loss of CLPB and expression of CLPB variants result in impaired granulocytic differentiation of human hematopoietic progenitor cells and increased apoptosis. These CLPB variants associate with wild-type CLPB and inhibit its adenosine triphosphatase and disaggregase activity in a dominant-negative fashion. Finally, expression of CLPB variants is associated with impaired mitochondrial function but does not render cells more sensitive to endoplasmic reticulum stress. Together, these data show that heterozygous CLPB variants are a new and relatively common cause of congenital neutropenia and should be considered in the evaluation of patients with congenital neutropenia.
Collapse
Affiliation(s)
- Julia T Warren
- Division of Hematology-Oncology, Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO
| | - Ryan R Cupo
- Department of Biochemistry and Biophysics, Pharmacology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Peeradol Wattanasirakul
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St, MO
| | - David H Spencer
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St, MO
| | - Adam E Locke
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St, MO
| | - Vahagn Makaryan
- Department of Medicine, University of Washington, Seattle, WA
| | | | | | - Natalie L Kingston
- Medical Scientist Training Program, Washington University School of Medicine, St, MO
| | - James Shorter
- Department of Biochemistry and Biophysics, Pharmacology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Christine Bellanné-Chantelot
- Département de Génétique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié Salpêtrière, Sorbonne Université, Paris, France; and
| | - Jean Donadieu
- Sorbonne Université, INSERM, AP-HP, Registre français des Neutropénies Chroniques, Centre de Référence des Neutropénies Chroniques, Hôpital Trousseau, Service Hémato Oncologie Pédiatrique, Paris, France
| | - David C Dale
- Department of Medicine, University of Washington, Seattle, WA
| | - Daniel C Link
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St, MO
| |
Collapse
|
11
|
A patient with severe congenital neutropenia harbors a missense ELANE mutation due to paternal germline mosaicism. Clin Chim Acta 2021; 526:14-20. [PMID: 34968504 DOI: 10.1016/j.cca.2021.12.023] [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: 10/05/2021] [Revised: 12/02/2021] [Accepted: 12/23/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Clinical and genetic characteristics of ELANE mutation of a 3-year-old male who had a severe congenital neutropenia (SCN) were examined. We then investigated whether CRISPR/Cas9-mediated gene editing could correct the mutation. PROCEDURE The proband underwent extensive clinical assessments, such as exome sequencing and bioinformatics analysis, so that pathogenic genes could be identified. Sanger sequencing was also utilized for confirmation. The cell line, 293-ELANE, harboring ELANE mutation was generated, and the mutation was then corrected by CRISPR/Cas9-mediated homology-directed repair (HDR). RESULTS The ELANE gene test in the proband unveiled a heterozygous de novo missense mutation: c. 248T > A (p.V83D), which was not detected in his asymptomatic parents who had provided peripheral blood samples. We found that 46.01% of his father's sperm cells had the same mutation. These results demonstrate that the proband inherited the ELANE mutation from his father, who had an average neutrophil count but had a germline mosaicism. The highest repair efficiency of CRISPR/Cas9-mediated HDR for 293-ELANE is 4.43%. CONCLUSIONS We identified a missense mutation (p.V83D) in ELANE that causes SCN. This is the first report on paternal semen mosaicism of an ELANE mutation. Our study paves the way for preimplantation genetic diagnosis (PGD) based on ELANE mutation prevention and clinical treatment of congenital disabilities.
Collapse
|
12
|
Rydzynska Z, Pawlik B, Krzyzanowski D, Mlynarski W, Madzio J. Neutrophil Elastase Defects in Congenital Neutropenia. Front Immunol 2021; 12:653932. [PMID: 33968054 PMCID: PMC8100030 DOI: 10.3389/fimmu.2021.653932] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/06/2021] [Indexed: 12/21/2022] Open
Abstract
Severe congenital neutropenia (SCN) is a rare hematological condition with heterogenous genetic background. Neutrophil elastase (NE) encoded by ELANE gene is mutated in over half of the SCN cases. The role of NE defects in myelocytes maturation arrest in bone marrow is widely investigated; however, the mechanism underlying this phenomenon has still remained unclear. In this review, we sum up the studies exploring mechanisms of neutrophil deficiency, biological role of NE in neutrophil and the effects of ELANE mutation and neutropenia pathogenesis. We also explain the hypotheses presented so far and summarize options of neutropenia therapy.
Collapse
Affiliation(s)
- Zuzanna Rydzynska
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
| | - Bartlomiej Pawlik
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Damian Krzyzanowski
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland.,Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | - Wojciech Mlynarski
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
| | - Joanna Madzio
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
13
|
Cooperating, congenital neutropenia-associated Csf3r and Runx1 mutations activate pro-inflammatory signaling and inhibit myeloid differentiation of mouse HSPCs. Ann Hematol 2020; 99:2329-2338. [PMID: 32821971 PMCID: PMC7481169 DOI: 10.1007/s00277-020-04194-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022]
Abstract
Patients with the pre-leukemia bone marrow failure syndrome called severe congenital neutropenia (CN) have an approximately 15% risk of developing acute myeloid leukemia (AML; called here CN/AML). Most CN/AML patients co-acquire CSF3R and RUNX1 mutations, which play cooperative roles in the development of AML. To establish an in vitro model of leukemogenesis, we utilized bone marrow lin- cells from transgenic C57BL/6-d715 Csf3r mice expressing a CN patient-mimicking truncated CSF3R mutation. We transduced these cells with vectors encoding RUNX1 wild type (WT) or RUNX1 mutant proteins carrying the R139G or R174L mutations. Cells transduced with these RUNX1 mutants showed diminished in vitro myeloid differentiation and elevated replating capacity, compared with those expressing WT RUNX1. mRNA expression analysis showed that cells transduced with the RUNX1 mutants exhibited hyperactivation of inflammatory signaling and innate immunity pathways, including IL-6, TLR, NF-kappaB, IFN, and TREM1 signaling. These data suggest that the expression of mutated RUNX1 in a CSF3R-mutated background may activate the pro-inflammatory cell state and inhibit myeloid differentiation.
Collapse
|
14
|
Garg B, Mehta HM, Wang B, Kamel R, Horwitz MS, Corey SJ. Inducible expression of a disease-associated ELANE mutation impairs granulocytic differentiation, without eliciting an unfolded protein response. J Biol Chem 2020; 295:7492-7500. [PMID: 32299910 PMCID: PMC7247317 DOI: 10.1074/jbc.ra120.012366] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/09/2020] [Indexed: 01/08/2023] Open
Abstract
Severe congenital neutropenia (SCN) is characterized by a near absence of neutrophils, rendering individuals with this disorder vulnerable to recurrent life-threatening infections. The majority of SCN cases arise because of germline mutations in the gene elastase, neutrophil-expressed (ELANE) encoding the neutrophil granule serine protease neutrophil elastase. Treatment with a high dose of granulocyte colony-stimulating factor increases neutrophil production and reduces infection risk. How ELANE mutations produce SCN remains unknown. The currently proposed mechanism is that ELANE mutations promote protein misfolding, resulting in endoplasmic reticulum stress and activation of the unfolded protein response (UPR), triggering death of neutrophil precursors and resulting in neutropenia. Here we studied the ELANE mutation p.G185R, often associated with greater clinical severity (e.g. decreased responsiveness to granulocyte colony-stimulating factor and increased leukemogenesis). Using an inducible expression system, we observed that this ELANE mutation diminishes enzymatic activity and granulocytic differentiation without significantly affecting cell proliferation, cell death, or UPR induction in murine myeloblast 32D and human promyelocytic NB4 cells. Impaired differentiation was associated with decreased expression of genes encoding critical hematopoietic transcription factors (Gfi1, Cebpd, Cebpe, and Spi1), cell surface proteins (Csf3r and Gr1), and neutrophil granule proteins (Mpo and Elane). Together, these findings challenge the currently prevailing model that SCN results from mutant ELANE, which triggers endoplasmic reticulum stress, UPR, and apoptosis.
Collapse
Affiliation(s)
- Bhavuk Garg
- Department of Pediatrics, Cleveland Clinic, Cleveland, Ohio 44195; Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio 44195; Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio 44195
| | - Hrishikesh M Mehta
- Department of Pediatrics, Cleveland Clinic, Cleveland, Ohio 44195; Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio 44195; Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio 44195
| | - Borwyn Wang
- Department of Pediatrics, Cleveland Clinic, Cleveland, Ohio 44195; Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio 44195; Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio 44195
| | - Ralph Kamel
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Marshall S Horwitz
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington 98195
| | - Seth J Corey
- Department of Pediatrics, Cleveland Clinic, Cleveland, Ohio 44195; Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio 44195; Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio 44195.
| |
Collapse
|
15
|
Pei L, Shen X, Qu K, Tan C, Zou J, Wang Y, Ping F. Exploration of the Two-Way Adjustment Mechanism of Rhei Radix et Rhizoma for Cardiovascular Diseases. Comb Chem High Throughput Screen 2020; 23:1100-1112. [PMID: 32436824 DOI: 10.2174/1386207323666200521120308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/22/2020] [Accepted: 02/08/2020] [Indexed: 11/22/2022]
Abstract
AIM AND OBJECTIVE Myocardial infarction, cerebral infarction, and other diseases caused by vascular obstruction have always jeopardized human life and health. Several reports indicate that Rhei Radix et Rhizoma has a good clinical effect in the prevention and treatment of cardiovascular diseases. Owing to the complexity of herbal medicine, the pharmacodynamic mechanism of Rhei Radix et Rhizoma is still unclear. The objectives of this study were to explore the two-way adjustment mechanism of Rhei Radix et Rhizoma and provide a new solution for the prevention and treatment of cardiovascular disease. MATERIALS AND METHODS This study used data mining, reverse pharmacophore matching, network construction, GO and KEGG Analysis, and molecular docking to investigate the two-way adjustment mechanism of Rhei Radix et Rhizoma. The methods used were based on systems pharmacology and big data analysis technology. RESULTS The results suggest that Rhei Radix et Rhizoma uses a two-way adjustment of activating blood circulation, as well as blood coagulation in the prevention and treatment of cardiovascular diseases. The components involved in activating blood circulation are mainly anthraquinone components. The corresponding targets are NOS2, NOS3, CALM1, and the corresponding pathways are calcium signaling pathway, VEGF signaling pathway, platelet activation, and the PI3K-Akt signaling pathway. For blood coagulation, the components are mainly tannin components; the corresponding targets are F2, F10, ELANE, and the corresponding pathways are the neuroactive ligand-receptor interaction, complement and coagulation cascades. CONCLUSION This study indicated that Rhei Radix et Rhizoma exerts the two-way adjustment of activating blood circulation and blood coagulation in the prevention and treatment of cardiovascular diseases. It can make up for the side effects of the existing blood circulation drugs for cardiovascular disease, only activating blood circulation, and the uncontrollable large-area bleeding due to the long-term use of the drugs. This study provides a material basis for the development of new blood-activating drugs based on natural medicine.
Collapse
Affiliation(s)
- Lishan Pei
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Xia Shen
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Kai Qu
- Shaanxi Hospital of Chinese Medicine, Xi' an, Shaanxi, China
| | - Conge Tan
- Basic Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Junbo Zou
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yanxia Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Fan Ping
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| |
Collapse
|
16
|
Mir P, Klimiankou M, Findik B, Hähnel K, Mellor-Heineke S, Zeidler C, Skokowa J, Welte K. New insights into the pathomechanism of cyclic neutropenia. Ann N Y Acad Sci 2020; 1466:83-92. [PMID: 32083314 DOI: 10.1111/nyas.14309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 12/27/2019] [Accepted: 01/14/2020] [Indexed: 01/10/2023]
Abstract
Cyclic neutropenia (CyN) is a hematologic disorder in which peripheral blood absolute neutrophil counts (ANCs) show cycles of approximately 21-day intervals. The majority of CyN patients harbor ELANE mutations, but the mechanism of ANC cycling is unclear. We performed analysis of bone marrow (BM) subpopulations in CyN patients at the peak and the nadir of the ANC cycle and detected high proportions of BM hematopoietic stem cells (HSCs) and hematopoietic stem and progenitor cells (HSPCs) at the nadir of the ANC cycle, as compared with the peak. BM HSPCs produced fewer granulocyte colony-forming unit colonies at the ANC peak. To investigate the mechanism of cycling, we found that mRNA expression levels of ELANE and unfolded protein response (UPR)-related genes (ATF6, BiP (HSPA5), CHOP (DDIT3), and PERK (EIF2AK3)) were elevated, but antiapoptotic genes (Bcl-2 (BCL2) and bcl-xL (BCL2L1)) were reduced in CD34+ cells tested at the ANC nadir. Moreover, HSPCs revealed increased levels of reactive oxygen species and gH2AX at the ANC nadir. We suggest that in CyN patients, some HSPCs escape the UPR-induced endoplasmic reticulum (ER) stress and proliferate in response to granulocyte colony-stimulating factor (G-CSF) to a certain threshold at which UPR again affects the majority of HSPCs. There is a cyclic balance between ER stress-induced apoptosis of HSPCs and compensatory G-CSF-stimulated HSPC proliferation followed by granulocytic differentiation.
Collapse
Affiliation(s)
- Perihan Mir
- Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Maksim Klimiankou
- Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Betuel Findik
- Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Karin Hähnel
- Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Sabine Mellor-Heineke
- Department of Oncology, Hematology and Bone Marrow Transplantation, Hannover Medical School, Hannover, Germany
| | - Cornelia Zeidler
- Department of Oncology, Hematology and Bone Marrow Transplantation, Hannover Medical School, Hannover, Germany
| | - Julia Skokowa
- Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, University Hospital Tübingen, Tübingen, Germany
| | - Karl Welte
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, University Children's Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
17
|
Jia Y, Yue C, Bradford K, Qing X, Panosyan EH, Gotesman M. Novel ELANE Gene Mutation in a Newborn with Severe Congenital Neutropenia: Case Report and Literature Review. J Pediatr Genet 2019; 9:203-206. [PMID: 32714623 DOI: 10.1055/s-0039-3399523] [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: 08/03/2019] [Accepted: 09/30/2019] [Indexed: 10/25/2022]
Abstract
Severe neutropenia is defined as an absolute neutrophil count (ANC) of less than 0.5 × 10 9 /L. Severe congenital neutropenia (SCN) is an inborn disorder with maturation arrest of granulocytes due to various genetic abnormalities, which may lead to immunodeficiency. Among several associated genetic mutations, the variants or heterozygous mutations of the ELANE gene coding neutrophil elastase comprise approximately 50% of the genetic causes of SCN. We present a newborn (male) with severe neutropenia due to a novel ELANE gene mutation. The newborn was born at 38 6/7 weeks gestation to a 25-year-old mother with hypertension and morbid obesity. Pregnancy and delivery were uncomplicated but the baby obtained a complete blood count (CBC) on day of life 2 for a work up of hyperbilirubinemia. He was noted to initially have an ANC of 0.2 × 10 9 /L and 0 on subsequent blood counts. A bone marrow biopsy showed a left shift and consistent with myeloid maturation arrest. In direct DNA sequencing analysis, we found an ELANE gene mutation (Val119Glu, V119E), which may be a new gene mutation to cause SCN. The diagnosis of SCN in newborns is usually based on neutropenia identified on a routine CBC. Sufficient awareness and high suspicion of this rare disease can prevent missed or delayed diagnosis of SCN. Our analysis also suggests a new pathological mutation in the ELANE gene and supports the important role of molecular testing in SCN.
Collapse
Affiliation(s)
- Yue Jia
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, California, United States
| | - Changjun Yue
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, California, United States
| | - Kathryn Bradford
- Division of Hematology/Oncology, Department of Pediatrics, Mattel Children's Hospital, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Xin Qing
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, California, United States
| | - Eduard H Panosyan
- Division of Hematology/Oncology, Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, United States
| | - Moran Gotesman
- Division of Hematology/Oncology, Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, United States
| |
Collapse
|
18
|
Needham PG, Guerriero CJ, Brodsky JL. Chaperoning Endoplasmic Reticulum-Associated Degradation (ERAD) and Protein Conformational Diseases. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a033928. [PMID: 30670468 DOI: 10.1101/cshperspect.a033928] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Misfolded proteins compromise cellular homeostasis. This is especially problematic in the endoplasmic reticulum (ER), which is a high-capacity protein-folding compartment and whose function requires stringent protein quality-control systems. Multiprotein complexes in the ER are able to identify, remove, ubiquitinate, and deliver misfolded proteins to the 26S proteasome for degradation in the cytosol, and these events are collectively termed ER-associated degradation, or ERAD. Several steps in the ERAD pathway are facilitated by molecular chaperone networks, and the importance of ERAD is highlighted by the fact that this pathway is linked to numerous protein conformational diseases. In this review, we discuss the factors that constitute the ERAD machinery and detail how each step in the pathway occurs. We then highlight the underlying pathophysiology of protein conformational diseases associated with ERAD.
Collapse
Affiliation(s)
- Patrick G Needham
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | | | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| |
Collapse
|
19
|
Nasri M, Ritter M, Mir P, Dannenmann B, Aghaallaei N, Amend D, Makaryan V, Xu Y, Fletcher B, Bernhard R, Steiert I, Hahnel K, Berger J, Koch I, Sailer B, Hipp K, Zeidler C, Klimiankou M, Bajoghli B, Dale DC, Welte K, Skokowa J. CRISPR/Cas9-mediated ELANE knockout enables neutrophilic maturation of primary hematopoietic stem and progenitor cells and induced pluripotent stem cells of severe congenital neutropenia patients. Haematologica 2019; 105:598-609. [PMID: 31248972 PMCID: PMC7049355 DOI: 10.3324/haematol.2019.221804] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/21/2019] [Indexed: 11/09/2022] Open
Abstract
A Autosomal-dominant ELANE mutations are the most common cause of severe congenital neutropenia. Although the majority of congenital neutropenia patients respond to daily granulocyte colony stimulating factor, approximately 15 % do not respond to this cytokine at doses up to 50 μg/kg/day and approximately 15 % of patients will develop myelodysplasia or acute myeloid leukemia. “Maturation arrest,” the failure of the marrow myeloid progenitors to form mature neutrophils, is a consistent feature of ELANE associated congenital neutropenia. As mutant neutrophil elastase is the cause of this abnormality, we hypothesized that ELANE associated neutropenia could be treated and “maturation arrest” corrected by a CRISPR/Cas9-sgRNA ribonucleoprotein mediated ELANE knockout. To examine this hypothesis, we used induced pluripotent stem cells from two congenital neutropenia patients and primary hematopoietic stem and progenitor cells from four congenital neutropenia patients harboring ELANE mutations as well as HL60 cells expressing mutant ELANE. We observed that granulocytic differentiation of ELANE knockout induced pluripotent stem cells and primary hematopoietic stem and progenitor cells were comparable to healthy individuals. Phagocytic functions, ROS production, and chemotaxis of the ELANE KO (knockout) neutrophils were also normal. Knockdown of ELANE in the mutant ELANE expressing HL60 cells also allowed full maturation and formation of abundant neutrophils. These observations suggest that ex vivo CRISPR/Cas9 RNP based ELANE knockout of patients’ primary hematopoietic stem and progenitor cells followed by autologous transplantation may be an alternative therapy for congenital neutropenia.
Collapse
Affiliation(s)
- Masoud Nasri
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Malte Ritter
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Perihan Mir
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Benjamin Dannenmann
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Narges Aghaallaei
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Diana Amend
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Vahagn Makaryan
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Yun Xu
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Breanna Fletcher
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Regine Bernhard
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Ingeborg Steiert
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Karin Hahnel
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Jürgen Berger
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Iris Koch
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Brigitte Sailer
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Katharina Hipp
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Cornelia Zeidler
- Department of Oncology, Hematology, Immunology and Bone Marrow Transplantation, Hannover Medical School, Hannover, Germany
| | - Maksim Klimiankou
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Baubak Bajoghli
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - David C Dale
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Karl Welte
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany.,University Children's Hospital Tübingen, Tübingen, Germany
| | - Julia Skokowa
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
20
|
Human iPSC-based model of severe congenital neutropenia reveals elevated UPR and DNA damage in CD34+ cells preceding leukemic transformation. Exp Hematol 2019; 71:51-60. [DOI: 10.1016/j.exphem.2018.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/23/2018] [Accepted: 12/30/2018] [Indexed: 11/24/2022]
|
21
|
Congenital neutropenia and primary immunodeficiency diseases. Crit Rev Oncol Hematol 2019; 133:149-162. [DOI: 10.1016/j.critrevonc.2018.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 02/06/2023] Open
|
22
|
Ley K, Hoffman HM, Kubes P, Cassatella MA, Zychlinsky A, Hedrick CC, Catz SD. Neutrophils: New insights and open questions. Sci Immunol 2018; 3:eaat4579. [PMID: 30530726 DOI: 10.1126/sciimmunol.aat4579] [Citation(s) in RCA: 329] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/08/2018] [Indexed: 02/07/2024]
Abstract
Neutrophils are the first line of defense against bacteria and fungi and help combat parasites and viruses. They are necessary for mammalian life, and their failure to recover after myeloablation is fatal. Neutrophils are short-lived, effective killing machines. Their life span is significantly extended under infectious and inflammatory conditions. Neutrophils take their cues directly from the infectious organism, from tissue macrophages and other elements of the immune system. Here, we review how neutrophils traffic to sites of infection or tissue injury, how they trap and kill bacteria, how they shape innate and adaptive immune responses, and the pathophysiology of monogenic neutrophil disorders.
Collapse
Affiliation(s)
- Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Circle Drive, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego,9500 Gilman Drive, La Jolla, CA, USA
| | - Hal M Hoffman
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of California, San Diego and Rady Children's Hospital, San Diego, CA, USA
| | - Paul Kubes
- Immunology Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Marco A Cassatella
- Department of Medicine, Section of General Pathology, University of Verona, Strada Le Grazie 4, 37134 Verona, Italy
| | - Arturo Zychlinsky
- Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Circle Drive, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego,9500 Gilman Drive, La Jolla, CA, USA
| | - Sergio D Catz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
23
|
Skokowa J, Zeidler C, Welte K. Chronische Neutropenien im Kindesalter. Monatsschr Kinderheilkd 2018. [DOI: 10.1007/s00112-018-0545-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
24
|
Corey SJ, Oyarbide U. New monogenic disorders identify more pathways to neutropenia: from the clinic to next-generation sequencing. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:172-180. [PMID: 29222253 PMCID: PMC5912212 DOI: 10.1182/asheducation-2017.1.172] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Neutrophils are the most common type of leukocyte in human circulating blood and constitute one of the chief mediators for innate immunity. Defined as a reduction from a normal distribution of values, neutropenia results from a number of congenital and acquired conditions. Neutropenia may be insignificant, temporary, or associated with a chronic condition with or without a vulnerability to life-threatening infections. As an inherited bone marrow failure syndrome, neutropenia may be associated with transformation to myeloid malignancy. Recognition of an inherited bone marrow failure syndrome may be delayed into adulthood. The list of monogenic neutropenia disorders is growing, heterogeneous, and bewildering. Furthermore, greater knowledge of immune-mediated and drug-related causes makes the diagnosis and management of neutropenia challenging. Recognition of syndromic presentations and especially the introduction of next-generation sequencing are improving the accuracy and expediency of diagnosis as well as their clinical management. Furthermore, identification of monogenic neutropenia disorders is shedding light on the molecular mechanisms of granulopoiesis and myeloid malignancies.
Collapse
Affiliation(s)
- Seth J Corey
- Department of Pediatrics, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA
| | - Usua Oyarbide
- Department of Pediatrics, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
25
|
Makaryan V, Kelley ML, Fletcher B, Bolyard AA, Aprikyan AA, Dale DC. Elastase inhibitors as potential therapies for ELANE-associated neutropenia. J Leukoc Biol 2017; 102:1143-1151. [PMID: 28754797 DOI: 10.1189/jlb.5a1016-445r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 06/23/2017] [Accepted: 07/12/2017] [Indexed: 02/03/2023] Open
Abstract
Mutations in ELANE, the gene for neutrophil elastase (NE), a protease expressed early in neutrophil development, are the most frequent cause of cyclic (CyN) and severe congenital neutropenia (SCN). We hypothesized that inhibitors of NE, acting either by directly inhibiting enzymatic activity or as chaperones for the mutant protein, might be effective as therapy for CyN and SCN. We investigated β-lactam-based inhibitors of human NE (Merck Research Laboratories, Kenilworth, NJ, USA), focusing on 1 inhibitor called MK0339, a potent, orally absorbed agent that had been tested in clinical trials and shown to have a favorable safety profile. Because fresh, primary bone marrow cells are rarely available in sufficient quantities for research studies, we used 3 cellular models: patient-derived, induced pluripotent stem cells (iPSCs); HL60 cells transiently expressing mutant NE; and HL60 cells with regulated expression of the mutant enzyme. In all 3 models, the cells expressing the mutant enzyme had reduced survival as measured with annexin V and FACS. Coincubation with the inhibitors, particularly MK0339, promoted cell survival and increased formation of mature neutrophils. These studies suggest that cell-permeable inhibitors of neutrophil elastase show promise as novel therapies for ELANE-associated neutropenia.
Collapse
Affiliation(s)
- Vahagn Makaryan
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Merideth L Kelley
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Breanna Fletcher
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Audrey Anna Bolyard
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Severe Chronic Neutropenia International Registry, University of Washington, Seattle, Washington, USA; and
| | | | - David C Dale
- Department of Medicine, University of Washington, Seattle, Washington, USA;
| |
Collapse
|
26
|
Abstract
Severe congenital neutropenias are a heterogeneous group of rare haematological diseases characterized by impaired maturation of neutrophil granulocytes. Patients with severe congenital neutropenia are prone to recurrent, often life-threatening infections beginning in their first months of life. The most frequent pathogenic defects are autosomal dominant mutations in ELANE, which encodes neutrophil elastase, and autosomal recessive mutations in HAX1, whose product contributes to the activation of the granulocyte colony-stimulating factor (G-CSF) signalling pathway. The pathophysiological mechanisms of these conditions are the object of extensive research and are not fully understood. Furthermore, severe congenital neutropenias may predispose to myelodysplastic syndromes or acute myeloid leukaemia. Molecular events in the malignant progression include acquired mutations in CSF3R (encoding G-CSF receptor) and subsequently in other leukaemia-associated genes (such as RUNX1) in a majority of patients. Diagnosis is based on clinical manifestations, blood neutrophil count, bone marrow examination and genetic and immunological analyses. Daily subcutaneous G-CSF administration is the treatment of choice and leads to a substantial increase in blood neutrophil count, reduction of infections and drastic improvement of quality of life. Haematopoietic stem cell transplantation is the alternative treatment. Regular clinical assessments (including yearly bone marrow examinations) to monitor treatment course and detect chromosomal abnormalities (for example, monosomy 7 and trisomy 21) as well as somatic pre-leukaemic mutations are recommended.
Collapse
Affiliation(s)
- Julia Skokowa
- Department of Hematology, Oncology, Clinical Immunology, University of Tübingen, Tübingen, Germany
| | - David C Dale
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Ivo P Touw
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Cornelia Zeidler
- Department of Hematology and Oncology, Medical School Hannover, Hannover, Germany
| | - Karl Welte
- University Children's Hospital, Department of General Pediatrics and Pediatric Hematology and Oncology, Hoppe-Seyler-Str. 1, Tübingen 72076, Germany
| |
Collapse
|
27
|
Abstract
Neutropenia, usually defined as a blood neutrophil count <1·5 × 109 /l, is a common medical problem for children and adults. There are many causes for neutropenia, and at each stage in life the clinical pattern of causes and consequences differs significantly. I recommend utilizing the age of the child and clinical observations for the preliminary diagnosis and primary management. In premature infants, neutropenia is quite common and contributes to the risk of sepsis with necrotizing enterocolitis. At birth and for the first few months of life, neutropenia is often attributable to isoimmune or alloimmune mechanisms and predisposes to the risk of severe bacterial infections. Thereafter when a child is discovered to have neutropenia, often associated with relatively minor symptoms, it is usually attributed to autoimmune disorder or viral infection. The congenital neutropenia syndromes are usually recognized when there are recurrent infections, the neutropenia is severe and there are congenital anomalies suggesting a genetic disorder. This review focuses on the key clinical finding and laboratory tests for diagnosis with commentaries on treatment, particularly the use of granulocyte colony-stimulating factor to treat childhood neutropenia.
Collapse
Affiliation(s)
- David C Dale
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
28
|
Invasive Fungal Infection in Primary Immunodeficiencies Other Than Chronic Granulomatous Disease. CURRENT FUNGAL INFECTION REPORTS 2017. [DOI: 10.1007/s12281-017-0273-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
29
|
Role of granule proteases in the life and death of neutrophils. Biochem Biophys Res Commun 2017; 482:473-481. [PMID: 28212734 DOI: 10.1016/j.bbrc.2016.11.086] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/08/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023]
Abstract
Neutrophils constitute a crucial component of the innate immune defenses against microbes. Produced in the bone marrow and patrolling in blood vessels, neutrophils are recruited to injured tissues and are immediately active to contain pathogen invasion. Neutrophils undergo programmed cell death by multiple, context-specific pathways, which have consequences on immunopathology and disease outcome. Studies in the last decade indicate additional functions for neutrophils - or a subset of neutrophils - in modulating adaptive responses and tumor progression. Neutrophil granules contain abundant amounts of various proteases, which are directly implicated in protective and pathogenic functions of neutrophils. It now emerges that neutral serine proteases such as cathepsin G and proteinase-3 also contribute to the neutrophil life cycle, but do so via different pathways than that of the aspartate protease cathepsin D and that of mutants of the serine protease elastase. The aim of this review is to appraise the present knowledge of the function of neutrophil granule proteases and their inhibitors in neutrophil cell death, and to integrate these findings in the current understandings of neutrophil life cycle and programmed cell death pathways.
Collapse
|