1
|
Iribarren C, Levedahl KH, Atanasoai I, Mattsson M, Höglund M, Söderlund S, Hägglund H, Eriksson N, Carlson M, Nilsson GP. Plasma Protein Profiling to Discern Indolent from Advanced Systemic Mastocytosis. J Mol Diagn 2024; 26:792-804. [PMID: 38925457 DOI: 10.1016/j.jmoldx.2024.05.010] [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: 02/13/2024] [Revised: 05/06/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Mastocytosis is a heterogeneous disorder characterized by abnormal mast cell accumulation, in which the clinical severity may be explained by distinct molecular mechanisms. This study aimed to explore plasma protein biomarkers associated with systemic mastocytosis subtypes, as well as the cellular origin of the identified proteins. Plasma samples from patients with mastocytosis, including cutaneous mastocytosis (CM), indolent systemic mastocytosis (ISM), and advanced systemic mastocytosis (AdvSM), and a reference group of patients with polycythemia vera, were analyzed by Proximity Extension Assay technology targeting 275 proteins. Furthermore, potential cellular origin was explored using an available single-cell RNA-sequencing data set generated from patients with ISM. The study cohort included 16 patients with CM, 92 patients with systemic mastocytosis (ISM, n = 80; AdvSM, n = 12), and 60 patients with polycythemia vera. A principal component analysis based on 275 plasma proteins revealed one cluster of patients with CM and ISM that was separated from patients with AdvSM. Up to 29 proteins were associated with distinct severe activity in patients with systemic mastocytosis (ISM versus AdvSM), including IL-1 receptor type 1 (IL-1RT1) and tumor necrosis factor ligand superfamily member 13B (TNFSF13B) (q < 0.01). Furthermore, single-cell RNA-sequencing analysis from ISM-derived bone marrow cells revealed that the mRNA for the identified proteins was not exclusive of mast cells. Distinct plasma protein profiles show potential to refine ISM and AdvSM diagnoses, possibly reflecting differences in pathogenic mechanisms and diverse clinical manifestations.
Collapse
Affiliation(s)
- Cristina Iribarren
- Division of Immunology and Allergy, Department of Medicine Solna, and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Kerstin H Levedahl
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Ionut Atanasoai
- Division of Immunology and Allergy, Department of Medicine Solna, and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Section of Hematology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Mattias Mattsson
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Martin Höglund
- Section of Hematology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Stina Söderlund
- Section of Hematology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Hans Hägglund
- Section of Hematology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Marie Carlson
- Gastroenterology Research Group, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gunnar P Nilsson
- Division of Immunology and Allergy, Department of Medicine Solna, and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Section of Hematology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
2
|
Sato M, Kurokawa R, Okimoto N, Tokushige J, Ikemura M, Abe O. CT and Fluorine-18-Fluorodeoxyglucose (18F-FDG) PET/CT Imaging Findings of Aggressive Systemic Mastocytosis: A Case Report. Cureus 2024; 16:e64879. [PMID: 39156319 PMCID: PMC11330563 DOI: 10.7759/cureus.64879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2024] [Indexed: 08/20/2024] Open
Abstract
Aggressive systemic mastocytosis (ASM) is an advanced subtype of systemic mastocytosis characterized by organ involvement. In this article, we report a case with ASM in a 54-year-old woman with characteristic findings on computed tomography (CT) and fluorine-18-fluorodeoxyglucose positron emission tomography (18F-FDG PET)/CT. Contrast-enhanced CT on admission revealed hepatosplenomegaly, generalized osteosclerosis, colonic edema, edematous thickening of the wall in the ascending colon and edema in the surrounding regions of these organs and mesentery, ileus, subcutaneous edema, periportal collar sign, and multiple mesenteric lymphadenopathies. There was no 18F-FDG uptake in the lesions other than mild 18F-FDG uptake in the vertebrae, making the possibility of differential diagnoses such as metastasis, lymphoma, and extramedullary leukemia lower. Based on bone marrow biopsy results and clinical findings, the diagnosis of ASM was established. ASM can be a potentially fatal disease with a poor prognosis, and understanding its distinctive clinical course and imaging findings is crucial for early therapeutic intervention.
Collapse
Affiliation(s)
- Mai Sato
- Radiology, The University of Tokyo, Tokyo, JPN
| | | | | | - Junji Tokushige
- Hematology and Oncology, The University of Tokyo, Tokyo, JPN
| | | | - Osamu Abe
- Radiology, The University of Tokyo, Tokyo, JPN
| |
Collapse
|
3
|
Wu Z, Sun L, Xu Y, Huang H, Wu Z, Qiu B, Yan J, Yin X. The Value of Chemokine and Chemokine Receptors in Diagnosis, Prognosis, and Immunotherapy of Hepatocellular Carcinoma. Cancer Manag Res 2024; 16:403-420. [PMID: 38736589 PMCID: PMC11086648 DOI: 10.2147/cmar.s450959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/20/2024] [Indexed: 05/14/2024] Open
Abstract
Background Chemokines and chemokine receptors (CCRs) are involved in a variety of anti-tumour and pro-tumour immune processes in vivo, such as angiogenesis, metastasis, proliferation and invasiveness, and influence patient prognosis and response to therapy. Methods CCRs differentially expressed in HCC and associated with prognosis were extracted from TCGA and GEO databases, and the obtained CCRs were then used to construct signature genes, and the signature gene were selected for expression validation as well as functional experiments to explore the role of CCRs in the treatment and prognosis of HCC. Results We constructed a prognostic model including five CCRs (CCL20, CCL23, CCR3, CCR10, and CXCR3) and validated the expression of signature genes. The model's risk score is an independent prognostic factor for HCC. We have also developed prognostic model nomograms for clinical use. In addition, we validated that CCR3 expression is associated with poor prognosis in HCC, and the proliferation and migration ability of HCC cells was significantly inhibited after interfering with the expression of CCR3 in MHCC-LM3. We also looked at differences in pathway enrichment, immune infiltration and immune checkpoints. Finally, we found that risk scores were also correlated with drug sensitivity, the high-risk group had a better sensitivity to sorafenib. Conclusion The CCRs-related gene signature may better assess HCC prognosis and response to immunotherapy and tyrosine kinase inhibitors such as sorafenib in HCC, providing prospective solutions for diagnosis and treatment.
Collapse
Affiliation(s)
- Zhengyi Wu
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Liang Sun
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Yongkang Xu
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - He Huang
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Zhipeng Wu
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Bingbing Qiu
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Jinlong Yan
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Xiangbao Yin
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| |
Collapse
|
4
|
Degboé Y, Severino-Freire M, Couture G, Apoil PA, Gaudenzio N, Hermine O, Ruyssen-Witrand A, Paul C, Laroche M, Constantin A, Livideanu CB. The Prevalence Of Osteoporosis Is Low in Adult Cutaneous Mastocytosis Patients. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:1306-1312. [PMID: 38423295 DOI: 10.1016/j.jaip.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Systemic mastocytosis (SM) is a clonal disorder of mast cells (MCs) frequently associated with vertebral osteoporosis (OP) and subsequent vertebral fractures (VFs). The natural history of this OP remains unclear. Importantly, we do not know whether OP represents an early event triggered alongside MC abnormalities, and whether MC clonality is sufficient to trigger osteoporosis. OBJECTIVE To describe OP in patients with medullar clonality in cutaneous mastocytosis (CM) and monoclonal mast cell activation syndrome (MMAS) and to compare their osteoporosis characteristics with those of nonadvanced SM patients (bone marrow mastocytosis and indolent systemic mastocytosis). METHODS We retrospectively analyzed clinical, biological, and densitometric data of 27 CM, 13 MMAS, and 135 SM patients from the Mastocytosis Expert Center (CEREMAST) in Toulouse, France. RESULTS The OP (respectively 3.7, 30.8, and 34.1%) and VFs (0.0%, 15.4%, and 20%) were less frequent in CM than in MMAS and SM, despite the presence of clonal MCs in the bone marrow. Most patients with OP and VFs in the non-SM groups had the usual risk factors for OP. Interestingly, the only non-SM patient with a typical SM-like OP had high bone marrow tryptase, developed bone marrow KIT mutation during follow-up, and had a family history of SM. Our data show that OP is not a common clinical finding in CM but is frequent in MMAS. When OP and VFs occur in CM and MMAS patients, they differ from the usual phenotype of SM bone fragility. CONCLUSIONS Our findings suggest that, in most CM patients, the meaning and management of OP differs from that of OP in MMAS and nonadvanced SM. Prospective longitudinal studies and the validation of predictors are needed to identify CM and MMAS patients developing SM-related OP.
Collapse
Affiliation(s)
- Yannick Degboé
- Rheumatology Centre, Toulouse University Hospital and University Toulouse III, Toulouse, France; INFINITY-Toulouse Institute for Infectious and Inflammatory Diseases, INSERM UMR1291- CNRS UMR5051- University Toulouse III, Toulouse, France; Department of Dermatology and Mastocytosis Expert Centre (CEREMAST), Toulouse University Hospital and University Toulouse III, Toulouse, France.
| | - Maella Severino-Freire
- Department of Dermatology and Mastocytosis Expert Centre (CEREMAST), Toulouse University Hospital and University Toulouse III, Toulouse, France
| | - Guillaume Couture
- Rheumatology Centre, Toulouse University Hospital and University Toulouse III, Toulouse, France
| | - Pol-André Apoil
- Department of Dermatology and Mastocytosis Expert Centre (CEREMAST), Toulouse University Hospital and University Toulouse III, Toulouse, France; Department of Immunology, Toulouse University Hospital and University Toulouse III, Toulouse, France
| | - Nicolas Gaudenzio
- INFINITY-Toulouse Institute for Infectious and Inflammatory Diseases, INSERM UMR1291- CNRS UMR5051- University Toulouse III, Toulouse, France
| | - Olivier Hermine
- Department of Hematology and Mastocytosis Expert Centre (CEREMAST), Necker Children's Hospital and Paris Descartes University, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Adeline Ruyssen-Witrand
- Rheumatology Centre, Toulouse University Hospital and University Toulouse III, Toulouse, France; Centre d'Investigation Clinique de Toulouse CIC1436, Inserm, Team PEPSS, Toulouse, France
| | - Carle Paul
- INFINITY-Toulouse Institute for Infectious and Inflammatory Diseases, INSERM UMR1291- CNRS UMR5051- University Toulouse III, Toulouse, France; Department of Dermatology and Mastocytosis Expert Centre (CEREMAST), Toulouse University Hospital and University Toulouse III, Toulouse, France
| | - Michel Laroche
- Rheumatology Centre, Toulouse University Hospital and University Toulouse III, Toulouse, France
| | - Arnaud Constantin
- Rheumatology Centre, Toulouse University Hospital and University Toulouse III, Toulouse, France; INFINITY-Toulouse Institute for Infectious and Inflammatory Diseases, INSERM UMR1291- CNRS UMR5051- University Toulouse III, Toulouse, France
| | - Cristina Bulai Livideanu
- INFINITY-Toulouse Institute for Infectious and Inflammatory Diseases, INSERM UMR1291- CNRS UMR5051- University Toulouse III, Toulouse, France; Department of Dermatology and Mastocytosis Expert Centre (CEREMAST), Toulouse University Hospital and University Toulouse III, Toulouse, France
| |
Collapse
|
5
|
Theoharides TC, Twahir A, Kempuraj D. Mast cells in the autonomic nervous system and potential role in disorders with dysautonomia and neuroinflammation. Ann Allergy Asthma Immunol 2024; 132:440-454. [PMID: 37951572 DOI: 10.1016/j.anai.2023.10.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/16/2023] [Accepted: 10/06/2023] [Indexed: 11/14/2023]
Abstract
Mast cells (MC) are ubiquitous in the body, and they are critical for not only in allergic diseases but also in immunity and inflammation, including having potential involvement in the pathophysiology of dysautonomias and neuroinflammatory disorders. MC are located perivascularly close to nerve endings and sites such as the carotid bodies, heart, hypothalamus, the pineal gland, and the adrenal gland that would allow them not only to regulate but also to be affected by the autonomic nervous system (ANS). MC are stimulated not only by allergens but also many other triggers including some from the ANS that can affect MC release of neurosensitizing, proinflammatory, and vasoactive mediators. Hence, MC may be able to regulate homeostatic functions that seem to be dysfunctional in many conditions, such as postural orthostatic tachycardia syndrome, autism spectrum disorder, myalgic encephalomyelitis/chronic fatigue syndrome, and Long-COVID syndrome. The evidence indicates that there is a possible association between these conditions and diseases associated with MC activation. There is no effective treatment for any form of these conditions other than minimizing symptoms. Given the many ways MC could be activated and the numerous mediators released, it would be important to develop ways to inhibit stimulation of MC and the release of ANS-relevant mediators.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida; Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts.
| | - Assma Twahir
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida
| |
Collapse
|
6
|
Dahlin JS, Nilsson G. Systemic mastocytosis: dying or survivin. Blood 2024; 143:945-947. [PMID: 38483409 DOI: 10.1182/blood.2023023532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2024] Open
Affiliation(s)
| | - Gunnar Nilsson
- Karolinska Institutet
- Karolinska University Hospital
- Uppsala University
| |
Collapse
|
7
|
Greiner G, Witzeneder N, Klein K, Tangermann S, Kodajova P, Jaeger E, Ratzinger F, Gerner MC, Jawhar M, Baumgartner S, Fruehwirth K, Schmetterer KG, Zuber J, Gleixner KV, Mayerhofer M, Schwarzinger I, Simonitsch-Klupp I, Esterbauer H, Baer C, Walter W, Meggendorfer M, Strassl R, Haferlach T, Hartmann K, Kenner L, Sperr WR, Reiter A, Sexl V, Arock M, Valent P, Hoermann G. Tumor necrosis factor α promotes clonal dominance of KIT D816V+ cells in mastocytosis: role of survivin and impact on prognosis. Blood 2024; 143:1006-1017. [PMID: 38142424 DOI: 10.1182/blood.2023020515] [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: 03/20/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/26/2023] Open
Abstract
ABSTRACT Systemic mastocytosis (SM) is defined by the expansion and accumulation of neoplastic mast cells (MCs) in the bone marrow (BM) and extracutaneous organs. Most patients harbor a somatic KIT D816V mutation, which leads to growth factor-independent KIT activation and accumulation of MC. Tumor necrosis factor α (TNF) is a proapoptotic and inflammatory cytokine that has been implicated in the clonal selection of neoplastic cells. We found that KIT D816V increases the expression and secretion of TNF. TNF expression in neoplastic MCs is reduced by KIT-targeting drugs. Similarly, knockdown of KIT or targeting the downstream signaling cascade of MAPK and NF-κB signaling reduced TNF expression levels. TNF reduces colony formation in human BM cells, whereas KIT D816V+ cells are less susceptible to the cytokine, potentially contributing to clonal selection. In line, knockout of TNF in neoplastic MC prolonged survival and reduced myelosuppression in a murine xenotransplantation model. Mechanistic studies revealed that the relative resistance of KIT D816V+ cells to TNF is mediated by the apoptosis-regulator BIRC5 (survivin). Expression of BIRC5 in neoplastic MC was confirmed by immunohistochemistry of samples from patients with SM. TNF serum levels are significantly elevated in patients with SM and high TNF levels were identified as a biomarker associated with inferior survival. We here characterized TNF as a KIT D816V-dependent cytokine that promotes clonal dominance. We propose TNF and apoptosis-associated proteins as potential therapeutic targets in SM.
Collapse
Affiliation(s)
- Georg Greiner
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Ihr Labor, Medical Diagnostic Laboratories, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Nadine Witzeneder
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Klara Klein
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Simone Tangermann
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Petra Kodajova
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eva Jaeger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Marlene C Gerner
- Division of Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Mohamad Jawhar
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
- Department of Hematology and Oncology, Helios Pforzheim, Pforzheim, Germany
| | - Sigrid Baumgartner
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Medical University of Vienna, Vienna, Austria
| | - Karin Fruehwirth
- Medical Central Laboratory, State Hospital Feldkirch, Feldkirch, Austria
| | - Klaus G Schmetterer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Johannes Zuber
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Karoline V Gleixner
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | | | - Ilse Schwarzinger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | | | | | - Robert Strassl
- Division of Clinical Virology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Karin Hartmann
- Division of Allergy, Department of Dermatology, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Lukas Kenner
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Applied Metabolomics, Vienna, Austria
- Center for Biomarker Research in Medicine, Graz, Austria
| | - Wolfgang R Sperr
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Andreas Reiter
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | | | - Michel Arock
- Department of Hematological Biology and French National Reference Center for Mastocytosis (CEREMAST), Pitié-Salpêtrière Hospital, Paris Sorbonne University, Paris, France
| | - Peter Valent
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Gregor Hoermann
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Munich Leukemia Laboratory, Munich, Germany
| |
Collapse
|
8
|
Nicoloro-SantaBarbara J, Majd M, Burdick KE, Dixon V, Giannetti MP. Cognitive Impairment and Depression in Mastocytosis: A Synthesis of the Literature. Curr Allergy Asthma Rep 2024; 24:53-62. [PMID: 38294589 DOI: 10.1007/s11882-024-01127-z] [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] [Accepted: 01/05/2024] [Indexed: 02/01/2024]
Abstract
PURPOSE OF REVIEW Symptoms of depression and cognitive dysfunction are commonly reported in mastocytosis. The aims of this review paper are to summarize the current literature on cognitive dysfunction and depressive symptoms, elucidate some of the mechanistic pathways underlying depressive symptoms in mastocytosis, identify gaps in the literature, and offer guidance for future research in this area. RECENT FINDINGS The study of cognition and depression in mastocytosis is in its infancy and the methodological flaws of the current literature limit interpretability. There is preliminary evidence that some individuals with mastocytosis might experience mild deficits in memory. On average, depression symptom scores fell within the mild to moderate or sub-syndromal range. Regrettably, only one study utilized a standardized diagnostic instrument to assess major depressive disorder. The authors' tendency to inaccurately equate depressive symptoms with a diagnosis of major depressive disorder presents a notable issue. The prevalence of cognitive deficits and depression appears to be similar to other chronic illnesses. Future work needs to better characterize cognition and characterize "depression" in this population.
Collapse
Affiliation(s)
- Jennifer Nicoloro-SantaBarbara
- Department of Psychiatry, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Marzieh Majd
- Department of Psychiatry, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Katherine E Burdick
- Department of Psychiatry, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Victoria Dixon
- Department of Psychology, Northeastern University, Boston, MA, USA
| | - Matthew P Giannetti
- Harvard Medical School, Boston, MA, USA
- Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
9
|
Rosell A, Karlström C, Dahlin JS, Boey D, Klimkowska M, Ax K, Thålin C, Ungerstedt J. No indication of aberrant neutrophil extracellular trap release in indolent or advanced systemic mastocytosis. Scand J Immunol 2024; 99:e13333. [PMID: 38112220 DOI: 10.1111/sji.13333] [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: 05/28/2023] [Revised: 09/13/2023] [Accepted: 09/16/2023] [Indexed: 12/21/2023]
Abstract
In disease states with chronic inflammation, there is a crosstalk between mast cells and neutrophil granulocytes in the inflamed microenvironment, which may be potentiated by tryptase. In systemic mastocytosis (SM), mast cells are constitutively active and tryptase is elevated in blood. Mast cell activation in SM leads to symptoms from various organs depending on where the active mast cells reside, for example, palpitations, flush, allergic symptoms including anaphylactic reactions, and osteoporosis. Whether neutrophil function is altered in SM is not well understood. In the current study, we assessed nucleosomal citrullinated histone H3 (H3Cit-DNA) as a proxy for neutrophil extracellular trap release in plasma from 55 patients with indolent and advanced SM. We observed a strong trend towards a correlation between leukocyte count, eosinophil count and neutrophil count and H3Cit-DNA levels in patients with advanced SM but not in indolent SM; however, no differences in H3Cit-DNA levels in SM patients compared with healthy controls. H3Cit-DNA levels did not correlate with SM disease burden, tryptase levels, history of anaphylaxis or presence of cutaneous mastocytosis; thus, there is no evidence of a general neutrophil extracellular trap release in SM. Interestingly, H3Cit-DNA levels and leukocyte counts were elevated in a subgroup of SM patients with aberrant mast cell CD2 expression, which warrants further investigation. In conclusion, we found no evidence of global increase in neutrophil extracellular trap release in SM.
Collapse
Affiliation(s)
- Axel Rosell
- Center for Hematology and Regenerative Medicine (HERM), NEO, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- ME Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Cecilia Karlström
- Center for Hematology and Regenerative Medicine (HERM), NEO, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
- ME Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Joakim S Dahlin
- Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Daryl Boey
- Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Monika Klimkowska
- Pathology Unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Kajsa Ax
- Center for Hematology and Regenerative Medicine (HERM), NEO, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte Thålin
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Johanna Ungerstedt
- Center for Hematology and Regenerative Medicine (HERM), NEO, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
- ME Hematology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
10
|
Wang T, Yang S, Long Y, Li Y, Wang T, Hou Z. Olink proteomics analysis uncovers the landscape of inflammation-related proteins in patients with acute compartment syndrome. Front Immunol 2023; 14:1293826. [PMID: 38045696 PMCID: PMC10691257 DOI: 10.3389/fimmu.2023.1293826] [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: 09/13/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023] Open
Abstract
Purpose Our primary purpose was to explore the landscape of inflammation-related proteins, and our second goal was to investigate these proteins as potential biomarkers of acute compartment syndrome (ACS), which is a serious complication of tibial fractures. Methods We collected sera from 15 healthy subjects (control group, CG) and 30 patients with tibial fractures on admission day, comprising 15 patients with ACS (ACS group, AG) and 15 patients without ACS (fracture group, FG). Ten samples in each group were analyzed by the inflammation panel of Olink Proteomics Analysis, and all samples were verified by an ELISA. Receiver-operating characteristic (ROC) curve analysis was performed to identify the diagnostic ability and cutoff values of potential biomarkers. Results Our findings showed that the levels of IL6, CSF-1, and HGF in the FG were significantly higher than those in the CG. Similar results were found between the AG and CG, and their cutoff values for predicting ACS compared with the CG were 9.225 pg/ml, 81.04 pg/ml, and 0.3301 ng/ml, respectively. Furthermore, their combination had the highest diagnostic accuracy. Notably, compared with FG, we only found a higher expression of CCL23 in the AG. Additionally, we identified 35.75 pg/ml as the cutoff value of CCL23 for predicting ACS in patients with tibial fractures. Conclusion We identified CCL23 as a potential biomarker of ACS in comparison with tibial fracture patients and the significance of the combined diagnosis of IL6, CSF-1, and HGF for predicting ACS compared with healthy individuals. Furthermore, we also found their cutoff values, providing clinicians with a new method for rapidly diagnosing ACS. However, we need larger samples to verify our results.
Collapse
Affiliation(s)
- Tao Wang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Shijiazhuang, Hebei, China
| | - Shuo Yang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Shijiazhuang, Hebei, China
| | - Yubin Long
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Shijiazhuang, Hebei, China
- Department of Orthopedics, The First Central Hospital of Baoding, Baoding, China
| | - Yiran Li
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Shijiazhuang, Hebei, China
| | - Ting Wang
- Department of Nursing, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhiyong Hou
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Orthopaedic Research Institute of Hebei Province, Shijiazhuang, Hebei, China
| |
Collapse
|
11
|
Lyons JJ, Farkas H, Germenis AE, Rijavec M, Smith TD, Valent P. Genetic Variants Leading to Urticaria and Angioedema and Associated Biomarkers. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:2286-2301. [PMID: 37263349 DOI: 10.1016/j.jaip.2023.05.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/20/2023] [Accepted: 05/15/2023] [Indexed: 06/03/2023]
Abstract
Advances in next generation sequencing technologies, as well as their expanded accessibility and clinical use over the past 2 decades, have led to an exponential increase in the number of identified single gene disorders. Among these are primary atopic disorders-inborn errors of immunity resulting in severe allergic phenotypes as a primary presenting feature. Two cardinal aspects of type I immediate hypersensitivity allergic reactions are hives and angioedema. Mast cells (MCs) are frequent primary drivers of these symptoms, but other cells have also been implicated. Even where MC degranulation is believed to be the cause, mediator-induced symptoms may greatly vary among individuals. Angioedema-particularly in the absence of hives-may also be caused by hereditary angioedema conditions resulting from aberrant regulation of contact system activation and excessive bradykinin generation or impairment of vascular integrity. In these patients, swelling can affect unpredictable locations and fail to respond to MC-directed therapies. Genetic variants have helped delineate key pathways in the etiology of urticaria and nonatopic angioedema and led to the development of targeted therapies. Herein, we describe the currently known inherited and acquired genetic causes for these conditions, highlight specific features in their clinical presentations, and discuss the benefits and limitations of biomarkers that can help distinguish them.
Collapse
Affiliation(s)
- Jonathan J Lyons
- Translational Allergic Immunopathology Unit, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Henriette Farkas
- Department of Internal Medicine and Haematology, Hungarian Angioedema Center of Reference and Excellence, Semmelweis University, Budapest, Hungary
| | - Anastasios E Germenis
- Department of Immunology and Histocompatibility, School of Medicine, University of Thessaly, Larissa, Greece
| | - Matija Rijavec
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik, Slovenia; Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Tukisa D Smith
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, Calif
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Theoharides TC, Kempuraj D. Potential Role of Moesin in Regulating Mast Cell Secretion. Int J Mol Sci 2023; 24:12081. [PMID: 37569454 PMCID: PMC10418457 DOI: 10.3390/ijms241512081] [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: 06/27/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Mast cells have existed for millions of years in species that never suffer from allergic reactions. Hence, in addition to allergies, mast cells can play a critical role in homeostasis and inflammation via secretion of numerous vasoactive, pro-inflammatory and neuro-sensitizing mediators. Secretion may utilize different modes that involve the cytoskeleton, but our understanding of the molecular mechanisms regulating secretion is still not well understood. The Ezrin/Radixin/Moesin (ERM) family of proteins is involved in linking cell surface-initiated signaling to the actin cytoskeleton. However, how ERMs may regulate secretion from mast cells is still poorly understood. ERMs contain two functional domains connected through a long α-helix region, the N-terminal FERM (band 4.1 protein-ERM) domain and the C-terminal ERM association domain (C-ERMAD). The FERM domain and the C-ERMAD can bind to each other in a head-to-tail manner, leading to a closed/inactive conformation. Typically, phosphorylation on the C-terminus Thr has been associated with the activation of ERMs, including secretion from macrophages and platelets. It has previously been shown that the ability of the so-called mast cell "stabilizer" disodium cromoglycate (cromolyn) to inhibit secretion from rat mast cells closely paralleled the phosphorylation of a 78 kDa protein, which was subsequently shown to be moesin, a member of ERMs. Interestingly, the phosphorylation of moesin during the inhibition of mast cell secretion was on the N-terminal Ser56/74 and Thr66 residues. This phosphorylation pattern could lock moesin in its inactive state and render it inaccessible to binding to the Soluble NSF attachment protein receptors (SNAREs) and synaptosomal-associated proteins (SNAPs) critical for exocytosis. Using confocal microscopic imaging, we showed moesin was found to colocalize with actin and cluster around secretory granules during inhibition of secretion. In conclusion, the phosphorylation pattern and localization of moesin may be important in the regulation of mast cell secretion and could be targeted for the development of effective inhibitors of secretion of allergic and inflammatory mediators from mast cells.
Collapse
Affiliation(s)
- Theoharis C. Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| |
Collapse
|
13
|
Söderlund S, Boey D, van Midden W, Kjellander M, Ax K, Qian H, Dahlin JS, Ungerstedt J. Proteomic and transcriptomic screening demonstrates increased mast cell-derived CCL23 in systemic mastocytosis. J Allergy Clin Immunol 2023; 152:205-213. [PMID: 36813186 DOI: 10.1016/j.jaci.2023.01.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/16/2022] [Accepted: 01/11/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND Systemic mastocytosis (SM) is a heterogeneous group of mast cell-driven diseases diagnosed by bone marrow sampling. However, there are a limited number of available blood disease biomarkers. OBJECTIVE Our aim was to identify mast cell-derived proteins that could potentially serve as blood biomarkers for indolent and advanced forms of SM. METHODS We performed a plasma proteomics screening coupled with single-cell transcriptomic analysis in SM patients and healthy subjects. RESULTS Plasma proteomics screening identified 19 proteins upregulated in indolent disease compared to healthy, and 16 proteins in advanced disease compared to indolent. Among these, 5 proteins, CCL19, CCL23, CXCL13, IL-10, and IL-12Rβ1, were higher in indolent relative to healthy and in advanced disease compared to indolent. Single-cell RNA sequencing demonstrated that CCL23, IL-10, and IL-6 were selectively produced by mast cells. Notably, plasma CCL23 levels correlated positively with known markers of SM disease severity, namely tryptase levels, percentage bone marrow mast cell infiltration, and IL-6. CONCLUSION CCL23 is produced predominantly by mast cells in SM, and CCL23 plasma levels are associated with disease severity, correlating positively with established markers of disease burden, thus suggesting that CCL23 is a specific SM biomarker. In addition, the combination of CCL19, CCL23, CXCL13, IL-10, and IL-12Rβ1 may be useful for defining disease stage.
Collapse
Affiliation(s)
- Stina Söderlund
- Section of Hematology, Uppsala University Hospital, Uppsala, Sweden; Center for Hematology and Regenerative Medicine (HERM), NEO, Department for Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Daryl Boey
- Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Wouter van Midden
- Center for Hematology and Regenerative Medicine (HERM), NEO, Department for Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Matilda Kjellander
- Center for Hematology and Regenerative Medicine (HERM), NEO, Department for Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Kajsa Ax
- Center for Hematology and Regenerative Medicine (HERM), NEO, Department for Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Hong Qian
- Center for Hematology and Regenerative Medicine (HERM), NEO, Department for Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Joakim S Dahlin
- Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Johanna Ungerstedt
- Center for Hematology and Regenerative Medicine (HERM), NEO, Department for Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden; ME Hematology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
14
|
Rama TA, Henriques AF, Matito A, Jara-Acevedo M, Caldas C, Mayado A, Muñoz-González JI, Moreira A, Cavaleiro-Rufo J, García-Montero A, Órfão A, Sanchez-Muñoz L, Álvarez-Twose I. Bone and Cytokine Markers Associated With Bone Disease in Systemic Mastocytosis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1536-1547. [PMID: 36801493 DOI: 10.1016/j.jaip.2023.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/05/2023] [Accepted: 02/03/2023] [Indexed: 02/19/2023]
Abstract
BACKGROUND Mastocytosis encompasses a heterogeneous group of diseases characterized by tissue accumulation of clonal mast cells, which frequently includes bone involvement. Several cytokines have been shown to play a role in the pathogenesis of bone mass loss in systemic mastocytosis (SM), but their role in SM-associated osteosclerosis remains unknown. OBJECTIVE To investigate the potential association between cytokine and bone remodeling markers with bone disease in SM, aiming at identifying biomarker profiles associated with bone loss and/or osteosclerosis. METHODS A total of 120 adult patients with SM, divided into 3 age and sex-matched groups according to their bone status were studied: (1) healthy bone (n = 46), (2) significant bone loss (n = 47), and (3) diffuse bone sclerosis (n = 27). Plasma levels of cytokines and serum baseline tryptase and bone turnover marker levels were measured at diagnosis. RESULTS Bone loss was associated with significantly higher levels of serum baseline tryptase (P = .01), IFN-γ (P = .05), IL-1β (P = .05), and IL-6 (P = .05) versus those found in patients with healthy bone. In contrast, patients with diffuse bone sclerosis showed significantly higher levels of serum baseline tryptase (P < .001), C-terminal telopeptide (P < .001), amino-terminal propeptide of type I procollagen (P < .001), osteocalcin (P < .001), bone alkaline phosphatase (P < .001), osteopontin (P < .01), and the C-C Motif Chemokine Ligand 5/RANTES chemokine (P = .01), together with lower IFN-γ (P = .03) and RANK-ligand (P = .04) plasma levels versus healthy bone cases. CONCLUSIONS SM with bone mass loss is associated with a proinflammatory cytokine profile in plasma, whereas diffuse bone sclerosis shows increased serum/plasma levels of biomarkers related to bone formation and turnover, in association with an immunosuppressive cytokine secretion profile.
Collapse
Affiliation(s)
- Tiago Azenha Rama
- Serviço de Imunoalergologia, Centro Hospitalar Universitário São João, Porto, Portugal; Serviço de Imunologia Básica e Clínica, Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, Porto, Portugal; EPIUnit - Institute of Public Health, University of Porto, Porto, Portugal; Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal.
| | - Ana Filipa Henriques
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) - Reference Center (CSUR) for Mastocytosis, Hospital Virgen del Valle, Complejo Hospitalario Universitario de Toledo, Toledo, Spain; Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Almudena Matito
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) - Reference Center (CSUR) for Mastocytosis, Hospital Virgen del Valle, Complejo Hospitalario Universitario de Toledo, Toledo, Spain; Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Maria Jara-Acevedo
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; DNA Sequencing Service (NUCLEUS), Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Caldas
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; DNA Sequencing Service (NUCLEUS), Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Andrea Mayado
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain; Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS) Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Javier I Muñoz-González
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain; Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS) Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - André Moreira
- Serviço de Imunoalergologia, Centro Hospitalar Universitário São João, Porto, Portugal; Serviço de Imunologia Básica e Clínica, Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, Porto, Portugal; EPIUnit - Institute of Public Health, University of Porto, Porto, Portugal; Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
| | - João Cavaleiro-Rufo
- EPIUnit - Institute of Public Health, University of Porto, Porto, Portugal; Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
| | - Andrés García-Montero
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain; Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS) Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Alberto Órfão
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain; Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine, Cytometry Service (NUCLEUS) Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Laura Sanchez-Muñoz
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) - Reference Center (CSUR) for Mastocytosis, Hospital Virgen del Valle, Complejo Hospitalario Universitario de Toledo, Toledo, Spain; Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Iván Álvarez-Twose
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) - Reference Center (CSUR) for Mastocytosis, Hospital Virgen del Valle, Complejo Hospitalario Universitario de Toledo, Toledo, Spain; Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
15
|
Parente R, Giudice V, Cardamone C, Serio B, Selleri C, Triggiani M. Secretory and Membrane-Associated Biomarkers of Mast Cell Activation and Proliferation. Int J Mol Sci 2023; 24:ijms24087071. [PMID: 37108232 PMCID: PMC10139107 DOI: 10.3390/ijms24087071] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Mast cells (MCs) are immune cells distributed in many organs and tissues and involved in the pathogenesis of allergic and inflammatory diseases as a major source of pro-inflammatory and vasoactive mediators. MC-related disorders are heterogeneous conditions characterized by the proliferation of MC within tissues and/or MC hyper-reactivity that leads to the uncontrolled release of mediators. MC disorders include mastocytosis, a clonal disease characterized by tissue MC proliferation, and MC activation syndromes that can be primary (clonal), secondary (related to allergic disorders), or idiopathic. Diagnosis of MC disorders is difficult because symptoms are transient, unpredictable, and unspecific, and because these conditions mimic many other diseases. Validation of markers of MC activation in vivo will be useful to allow faster diagnosis and better management of MC disorders. Tryptase, being the most specific MC product, is a widely used biomarker of proliferation and activation. Other mediators, such as histamine, cysteinyl leukotrienes, and prostaglandin D2, are unstable molecules and have limitations in their assays. Surface MC markers, detected by flow cytometry, are useful for the identification of neoplastic MC in mastocytosis but, so far, none of them has been validated as a biomarker of MC activation. Further studies are needed to identify useful biomarkers of MC activation in vivo.
Collapse
Affiliation(s)
- Roberta Parente
- Division of Allergy and Clinical Immunology, University of Salerno, 84081 Baronissi, Italy
| | - Valentina Giudice
- Division of Hematology and Transplant Center, University of Salerno, 84081 Baronissi, Italy
| | - Chiara Cardamone
- Division of Allergy and Clinical Immunology, University of Salerno, 84081 Baronissi, Italy
| | - Bianca Serio
- Division of Hematology and Transplant Center, University of Salerno, 84081 Baronissi, Italy
| | - Carmine Selleri
- Division of Hematology and Transplant Center, University of Salerno, 84081 Baronissi, Italy
| | - Massimo Triggiani
- Division of Allergy and Clinical Immunology, University of Salerno, 84081 Baronissi, Italy
| |
Collapse
|
16
|
Theoharides TC, Kempuraj D. Role of SARS-CoV-2 Spike-Protein-Induced Activation of Microglia and Mast Cells in the Pathogenesis of Neuro-COVID. Cells 2023; 12:688. [PMID: 36899824 PMCID: PMC10001285 DOI: 10.3390/cells12050688] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/07/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19). About 45% of COVID-19 patients experience several symptoms a few months after the initial infection and develop post-acute sequelae of SARS-CoV-2 (PASC), referred to as "Long-COVID," characterized by persistent physical and mental fatigue. However, the exact pathogenetic mechanisms affecting the brain are still not well-understood. There is increasing evidence of neurovascular inflammation in the brain. However, the precise role of the neuroinflammatory response that contributes to the disease severity of COVID-19 and long COVID pathogenesis is not clearly understood. Here, we review the reports that the SARS-CoV-2 spike protein can cause blood-brain barrier (BBB) dysfunction and damage neurons either directly, or via activation of brain mast cells and microglia and the release of various neuroinflammatory molecules. Moreover, we provide recent evidence that the novel flavanol eriodictyol is particularly suited for development as an effective treatment alone or together with oleuropein and sulforaphane (ViralProtek®), all of which have potent anti-viral and anti-inflammatory actions.
Collapse
Affiliation(s)
- Theoharis C. Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
17
|
Valent P, Akin C, Sperr WR, Horny HP, Arock M, Metcalfe DD, Galli SJ. New Insights into the Pathogenesis of Mastocytosis: Emerging Concepts in Diagnosis and Therapy. ANNUAL REVIEW OF PATHOLOGY 2023; 18:361-386. [PMID: 36270293 DOI: 10.1146/annurev-pathmechdis-031521-042618] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Mastocytosis is a heterogeneous group of neoplasms defined by a numerical increase and accumulation of clonal mast cells (MCs) in various organ systems. The disease may present as cutaneous mastocytosis or systemic mastocytosis (SM). On the basis of histopathological and molecular features, clinical variables, and organ involvement, SM is divided into indolent SM, smoldering SM, SM with an associated hematologic neoplasm, aggressive SM, and MC leukemia. Each variant is defined by unique diagnostic criteria and a unique spectrum of clinical presentations. A key driver of MC expansion and disease evolution is the oncogenic machinery triggered by mutant forms of KIT. The genetic background, additional somatic mutations, and comorbidities also contribute to the course and prognosis. Patients with SM may also suffer from mediator-related symptoms or even an MC activation syndrome. This article provides an update of concepts on the genetics, etiology, and pathology of mastocytosis, with emphasis on diagnostic criteria and new treatment concepts.
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria; .,Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Cem Akin
- Division of Allergy and Clinical Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Wolfgang R Sperr
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria; .,Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Hans-Peter Horny
- Institute of Pathology, Ludwig-Maximilian University, Munich, Germany
| | - Michel Arock
- Department of Hematological Biology, Pitié-Salpêtrière Charles-Foix Hospital, AP-HP Sorbonne University, Paris, France
| | - Dean D Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephen J Galli
- Department of Pathology, Department of Microbiology and Immunology, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
18
|
Lin H, Shen J, Zhu Y, Zhou L, Zhang S, Liu Z, Wu F, Zhan R. Serum CCL23 emerges as a biomarker for poor prognosis in patients with intracerebral hemorrhage. Clin Chim Acta 2022; 537:188-193. [DOI: 10.1016/j.cca.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 11/03/2022]
|
19
|
Nontryptase Urinary and Hematologic Biomarkers of Mast Cell Expansion and Mast Cell Activation: Status 2022. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:1974-1984. [PMID: 35346887 DOI: 10.1016/j.jaip.2022.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/17/2022]
Abstract
Quantitation of urinary metabolites of histamine, prostaglandin D2, and leukotriene E4 can fill the gap in our current efforts to improve diagnosis and management of symptomatic patients with systemic mastocytosis, and/or mast cell activation syndrome, In addition, patients symptomatic due to mast cell activation but who do not meet all the criteria for mast cell activation syndrome can have elevated baseline mediator metabolites. Serum tryptase levels have been the workhorse in diagnosing these disorders, but it has several drawbacks including the need to obtain acute and baseline samples, which require 2 visits to health care facilities and 2 venipunctures. Recently, increased baseline tryptase level has been reported in hereditary alpha tryptasemia, complicating diagnostic possibilities of an increased baseline tryptase level. Furthermore, no treatment can specifically be targeted at tryptase itself. In contrast, the finding of 1 or more elevated urinary levels of histamine, prostaglandin D2, and/or leukotriene E4 metabolites (1) greatly narrows diagnostic possibilities for causes of symptoms; (2) informs the practitioner what specific metabolic pathways are involved; and (3) targets the treatment in a specific, direct fashion. As a bonus, baseline spot/random urine samples can be obtained by the patients themselves and repeated at exactly the correct time when symptoms occur.
Collapse
|
20
|
Pérez‐Pons A, Jara‐Acevedo M, Henriques A, Navarro‐Navarro P, García‐Montero AC, Álvarez‐Twose I, Pedreira CE, Sánchez‐Muñoz L, Damasceno D, Caldas C, Muñoz‐González JI, Matito A, Flores‐Montero J, González‐López O, Criado I, Mayado A, Orfao A. Altered innate immune profile in blood of systemic mastocytosis patients. Clin Transl Allergy 2022; 12:e12167. [PMID: 35734269 PMCID: PMC9194602 DOI: 10.1002/clt2.12167] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/26/2022] [Accepted: 05/21/2022] [Indexed: 11/09/2022] Open
Abstract
Background Mast cells (MC) from systemic mastocytosis (SM) patients release MC mediators that lead to an altered microenvironment with potential consequences on innate immune cells, such as monocytes and dendritic cells (DC). Here we investigated the distribution and functional behaviour of different populations of blood monocytes and DC among distinct diagnostic subtypes of SM. Methods Overall, we studied 115 SM patients - 45 bone marrow mastocytosis (BMM), 61 indolent SM (ISM), 9 aggressive SM (ASM)- and 32 healthy donors (HD). Spontaneous and in vitro-stimulated cytokine production by blood monocytes, and their plasma levels, together with the distribution of different subsets of blood monocytes and DCs, were investigated. Results SM patients showed increased plasma levels and spontaneous production by blood monocytes of IL1β, IL6, IL8, TNFα and IL10, associated with an exhausted ability of LPS + IFNγ-stimulated blood monocytes to produce IL1β and TGFβ. SM (particularly ISM) patients also showed decreased counts of total monocytes, at the expense of intermediate monocytes and non-classical monocytes. Interestingly, while ISM and ASM patients had decreased numbers of plasmacytoid DC and myeloid DC (and their major subsets) in blood, an expansion of AXL+ DC was specifically encountered in BMM cases. Conclusion These results demonstrate an altered distribution of blood monocytes and DC subsets in SM associated with constitutive activation of functionally impaired blood monocytes and increased plasma levels of a wide variety of inflammatory cytokines, reflecting broad activation of the innate immune response in mastocytosis.
Collapse
Affiliation(s)
- Alba Pérez‐Pons
- Cancer Research Center (IBMCC, USAL‐CSIC)Department of Medicine and Cytometry Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Biomedical Research Institute of SalamancaSalamancaSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
| | - María Jara‐Acevedo
- Cancer Research Center (IBMCC, USAL‐CSIC)Department of Medicine and Cytometry Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Biomedical Research Institute of SalamancaSalamancaSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
- Sequencing Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
| | - Ana Henriques
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast)Virgen del Valle HospitalToledoSpain
| | - Paula Navarro‐Navarro
- Cancer Research Center (IBMCC, USAL‐CSIC)Department of Medicine and Cytometry Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Biomedical Research Institute of SalamancaSalamancaSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
- Sequencing Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
| | - Andrés C. García‐Montero
- Cancer Research Center (IBMCC, USAL‐CSIC)Department of Medicine and Cytometry Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Biomedical Research Institute of SalamancaSalamancaSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
| | - Iván Álvarez‐Twose
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast)Virgen del Valle HospitalToledoSpain
| | - Carlos E. Pedreira
- Systems and Computing Department (PESC)COPPEFederal University of Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
| | - Laura Sánchez‐Muñoz
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast)Virgen del Valle HospitalToledoSpain
| | - Daniela Damasceno
- Cancer Research Center (IBMCC, USAL‐CSIC)Department of Medicine and Cytometry Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Biomedical Research Institute of SalamancaSalamancaSpain
| | - Carolina Caldas
- Cancer Research Center (IBMCC, USAL‐CSIC)Department of Medicine and Cytometry Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Biomedical Research Institute of SalamancaSalamancaSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
| | - Javier I. Muñoz‐González
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast)Virgen del Valle HospitalToledoSpain
| | - Almudena Matito
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast)Virgen del Valle HospitalToledoSpain
| | - Juan Flores‐Montero
- Cancer Research Center (IBMCC, USAL‐CSIC)Department of Medicine and Cytometry Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Biomedical Research Institute of SalamancaSalamancaSpain
| | - Oscar González‐López
- Cancer Research Center (IBMCC, USAL‐CSIC)Department of Medicine and Cytometry Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Biomedical Research Institute of SalamancaSalamancaSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
| | - Ignacio Criado
- Cancer Research Center (IBMCC, USAL‐CSIC)Department of Medicine and Cytometry Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Biomedical Research Institute of SalamancaSalamancaSpain
| | - Andrea Mayado
- Cancer Research Center (IBMCC, USAL‐CSIC)Department of Medicine and Cytometry Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Biomedical Research Institute of SalamancaSalamancaSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
| | - Alberto Orfao
- Cancer Research Center (IBMCC, USAL‐CSIC)Department of Medicine and Cytometry Service (NUCLEUS)Universidad de SalamancaSalamancaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Biomedical Research Institute of SalamancaSalamancaSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
| |
Collapse
|
21
|
Stark KG, Falkowski NR, Brown CA, McDonald RA, Huffnagle GB. Contribution of the Microbiome, Environment, and Genetics to Mucosal Type 2 Immunity and Anaphylaxis in a Murine Food Allergy Model. FRONTIERS IN ALLERGY 2022; 3:851993. [PMID: 35769569 PMCID: PMC9234882 DOI: 10.3389/falgy.2022.851993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
There is heterogeneity inherent in the immune responses of individual mice in murine models of food allergy, including anaphylaxis, similar to the clinical heterogeneity observed in humans with food allergies to a defined food. One major driver of this heterogeneity may be differences in the microbiome between sensitized individuals. Our laboratory and others have reported that disruption of the microbiome (dysbiosis) by broad spectrum antibiotics and/or yeast colonization can alter systemic immunity and favor the development of mucosal Type 2 immunity to aeroallergens. Our objective was to use a well-characterized murine model (Balb/c mice) of food allergies (chicken egg ovalbumin, OVA) and determine if antibiotic-mediated dysbiosis (including C. albicans colonization) could enhance the manifestation of food allergies. Furthermore, we sought to identify elements of the microbiome and host response that were associated with this heterogeneity in the anaphylactic reaction between individual food allergen-sensitized mice. In our dataset, the intensity of the anaphylactic reactions was most strongly associated with a disrupted microbiome that included colonization by C. albicans, loss of a specific Lachnoclostridium species (tentatively, Lachnoclostridium YL32), development of a highly polarized Type 2 response in the intestinal mucosa and underlying tissue, and activation of mucosal mast cells. Serum levels of allergen-specific IgE were not predictive of the response and a complete absence of a microbiome did not fully recapitulate the response. Conventionalization of germ-free mice resulted in Akkermansia muciniphila outgrowth and a higher degree of heterogeneity in the allergic response. C57BL/6 mice remained resistant even under the same dysbiosis-inducing antibiotic regimens, while changes in the microbiome markedly altered the reactivity of Balb/c mice to OVA, as noted above. Strikingly, we also observed that genetically identical mice from different rooms in our vivarium develop different levels of a Type 2 response, as well as anaphylactic reactions. The intestinal microbiome in these mice also differed between rooms. Thus, our data recapitulate the heterogeneity in anaphylactic reactions, ranging from severe to none, seen in patients that have circulating levels of food allergen-reactive IgE and support the concept that alterations in the microbiome can be one factor underlying this heterogeneity.
Collapse
Affiliation(s)
- Kelsey G. Stark
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Nicole R. Falkowski
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Christopher A. Brown
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
- Institute for Research on Innovation and Science (IRIS), Institute for Social Research (ISR), University of Michigan, Ann Arbor, MI, United States
| | - Roderick A. McDonald
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Gary B. Huffnagle
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Gary B. Huffnagle
| |
Collapse
|
22
|
Valent P, Hartmann K, Schwaab J, Alvarez-Twose I, Brockow K, Bonadonna P, Hermine O, Niedoszytko M, Carter MC, Hoermann G, Sperr WR, Butterfield JH, Ustun C, Zanotti R, Radia DH, Castells M, Triggiani M, Schwartz LB, Orfao A, George TI, Sotlar K, Gotlib J, Reiter A, Horny HP, Arock M, Akin C, Metcalfe DD. Personalized Management Strategies in Mast Cell Disorders: ECNM-AIM User's Guide for Daily Clinical Practice. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:1999-2012.e6. [PMID: 35342031 DOI: 10.1016/j.jaip.2022.03.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 12/18/2022]
Abstract
Mastocytosis is a myeloid neoplasm defined by expansion and focal accumulation of clonal mast cells (MCs) in one or more organs. The disease exhibits a complex pathology and may be complicated by MC activation, bone abnormalities, neurological problems, gastrointestinal symptoms, and/or hematologic progression. The World Health Organization divides mastocytosis into cutaneous forms, systemic mastocytosis (SM) and MC sarcoma. In most patients with SM, somatic mutations in KIT are detected. Patients with indolent SM have a normal to near-normal life expectancy, whereas patients with advanced SM, including aggressive SM and MC leukemia, have a poor prognosis. In those with advanced SM, multiple somatic mutations and an associated hematologic neoplasm may be detected. Mediator-related symptoms can occur in any type of mastocytosis. Symptoms may be mild, severe, or even life-threatening. In patients with severe acute symptoms, an MC activation syndrome may be diagnosed. In these patients, relevant comorbidities include IgE-dependent and IgE-independent allergies. Management of patients with SM is an emerging challenge in daily practice and requires in-depth knowledge and a multidisciplinary and personalized approach with selection of appropriate procedures and interventions. In this article, we review the current knowledge on SM and MC activation syndrome, with emphasis on multidisciplinary aspects in diagnosis and patient-specific management. In addition, we provide a user's guide for application of markers, algorithms, prognostic scores, and treatments for use in daily practice.
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria.
| | - Karin Hartmann
- Division of Allergy, Department of Dermatology, University Hospital Basel, Basel, Switzerland; University of Basel, Basel, Switzerland; Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Juliana Schwaab
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Ivan Alvarez-Twose
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast), Toledo, Spain; CIBERONC, Hospital Virgen del Valle, Toledo, Spain
| | - Knut Brockow
- Department of Dermatology and Allergy Biederstein, Technical University of Munich, Munich, Germany
| | | | - Olivier Hermine
- Imagine Institute Université de Paris, Sorbonne, INSERM U1163, Centre national de référence des mastocytoses, Hôpital Necker, Assistance publique hôpitaux de Paris, Paris, France
| | - Marek Niedoszytko
- Department of Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Melody C Carter
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Gregor Hoermann
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria; MLL Munich Leukemia Laboratory, Munich, Germany
| | - Wolfgang R Sperr
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | | | - Celalettin Ustun
- Department of Medicine, Division of Hematology, Oncology and Cell Therapy, The Coleman Foundation Blood and Marrow Transplant Center at Rush University Medical Center, Chicago, Ill
| | - Roberta Zanotti
- Section of Hematology, Multidisciplinary Outpatients Clinics for Mastocytosis, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Deepti H Radia
- Department of Clinical Haematology, Guys and St Thomas' NHS Hospitals, London, United Kingdom
| | - Mariana Castells
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Massimo Triggiani
- Division of Allergy and Clinical Immunology, University of Salerno, Salerno, Italy
| | - Lawrence B Schwartz
- Department of Internal Medicine, Division of Rheumatology, Allergy & Immunology, Virginia Commonwealth University (VCU), Richmond, Va
| | - Alberto Orfao
- Servicio Central de Citometria (NUCLEUS), Centro de Investigacion del Cancer (IBMCC; CSIC/USAL), Instituto Biosanitario de Salamanca (IBSAL), Salamanca, Spain; Department of Medicine, University of Salamanca, Spain
| | - Tracy I George
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Karl Sotlar
- Institute of Pathology, University Hospital Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Jason Gotlib
- Stanford Cancer Institute/Stanford University School of Medicine, Stanford, Calif
| | - Andreas Reiter
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Hans-Peter Horny
- Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Michel Arock
- Department of Hematological Biology, Pitié-Salpêtrière Hospital, Pierre et Marie Curie University (UPMC), Paris, France
| | - Cem Akin
- Division of Allergy and Clinical Immunology, University of Michigan, Ann Arbor, Mich
| | - Dean D Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| |
Collapse
|
23
|
Graham RLJ, McMullen AA, Moore G, Dempsey-Hibbert NC, Myers B, Graham C. SWATH-MS identification of CXCL7, LBP, TGFβ1 and PDGFRβ as novel biomarkers in human systemic mastocytosis. Sci Rep 2022; 12:5087. [PMID: 35332176 PMCID: PMC8948255 DOI: 10.1038/s41598-022-08345-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
Mastocytosis is a rare myeloproliferative disease, characterised by accumulation of neoplastic mast cells in one or several organs. It presents as cutaneous or systemic. Patients with advanced systemic mastocytosis have a median survival of 3.5 years. The aetiology of mastocytosis is poorly understood, patients present with a broad spectrum of varying clinical symptoms that lack specificity to point clearly to a definitive diagnosis. Discovery of novel blood borne biomarkers would provide a tractable method for rapid identification of mastocytosis and its sub-types. Moving towards this goal, we carried out a clinical biomarker study on blood from twenty individuals (systemic mastocytosis: n = 12, controls: n = 8), which were subjected to global proteome investigation using the novel technology SWATH-MS. This identified several putative biomarkers for systemic mastocytosis. Orthogonal validation of these putative biomarkers was achieved using ELISAs. Utilising this workflow, we identified and validated CXCL7, LBP, TGFβ1 and PDGF receptor-β as novel biomarkers for systemic mastocytosis. We demonstrate that CXCL7 correlates with neutrophil count offering a new insight into the increased prevalence of anaphylaxis in mastocytosis patients. Additionally, demonstrating the utility of SWATH-MS for the discovery of novel biomarkers in the systemic mastocytosis diagnostic sphere.
Collapse
Affiliation(s)
- R L J Graham
- School of Biological Sciences, Queens University Belfast, Chlorine Gardens, Belfast, BT9 5DL, UK
| | - A A McMullen
- Department of Life Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK
| | - G Moore
- School of Biological Sciences, Queens University Belfast, Chlorine Gardens, Belfast, BT9 5DL, UK
| | - N C Dempsey-Hibbert
- Department of Life Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK
| | - B Myers
- University Hospitals of Leicester NHS Trust, Leicester, LE3 9QP, UK
| | - C Graham
- School of Biological Sciences, Queens University Belfast, Chlorine Gardens, Belfast, BT9 5DL, UK.
| |
Collapse
|
24
|
Theoharides TC. Could SARS-CoV-2 Spike Protein Be Responsible for Long-COVID Syndrome? Mol Neurobiol 2022; 59:1850-1861. [PMID: 35028901 PMCID: PMC8757925 DOI: 10.1007/s12035-021-02696-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023]
Abstract
SARS-CoV-2 infects cells via its spike protein binding to its surface receptor on target cells and results in acute symptoms involving especially the lungs known as COVID-19. However, increasing evidence indicates that many patients develop a chronic condition characterized by fatigue and neuropsychiatric symptoms, termed long-COVID. Most of the vaccines produced so far for COVID-19 direct mammalian cells via either mRNA or an adenovirus vector to express the spike protein, or administer recombinant spike protein, which is recognized by the immune system leading to the production of neutralizing antibodies. Recent publications provide new findings that may help decipher the pathogenesis of long-COVID. One paper reported perivascular inflammation in brains of deceased patients with COVID-19, while others showed that the spike protein could damage the endothelium in an animal model, that it could disrupt an in vitro model of the blood-brain barrier (BBB), and that it can cross the BBB resulting in perivascular inflammation. Moreover, the spike protein appears to share antigenic epitopes with human molecular chaperons resulting in autoimmunity and can activate toll-like receptors (TLRs), leading to release of inflammatory cytokines. Moreover, some antibodies produced against the spike protein may not be neutralizing, but may change its conformation rendering it more likely to bind to its receptor. As a result, one wonders whether the spike protein entering the brain or being expressed by brain cells could activate microglia, alone or together with inflammatory cytokines, since protective antibodies could not cross the BBB, leading to neuro-inflammation and contributing to long-COVID. Hence, there is urgent need to better understand the neurotoxic effects of the spike protein and to consider possible interventions to mitigate spike protein-related detrimental effects to the brain, possibly via use of small natural molecules, especially the flavonoids luteolin and quercetin.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, 136 Harrison Avenue, Suite 304, Boston, MA, 02111, USA.
- School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA.
- Departments of Internal Medicine and Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, 02111, USA.
- Institute of Neuro-Immune Medicine, Nova Southeastern University, Clearwater, FL, 33759, USA.
| |
Collapse
|
25
|
Agopian J, Da Costa Q, Nguyen QV, Scorrano G, Kousteridou P, Yuan M, Chelbi R, Goubard A, Castellano R, Maurizio J, Teodosio C, De Sepulveda P, Asara JM, Orfao A, Hermine O, Dubreuil P, Brenet F. GlcNAc is a mast-cell chromatin-remodeling oncometabolite that promotes systemic mastocytosis aggressiveness. Blood 2021; 138:1590-1602. [PMID: 33974006 DOI: 10.1182/blood.2020008948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 05/03/2021] [Indexed: 11/20/2022] Open
Abstract
Systemic mastocytosis (SM) is a KIT-driven hematopoietic neoplasm characterized by the excessive accumulation of neoplastic mast cells (MCs) in various organs and, mainly, the bone marrow (BM). Multiple genetic and epigenetic mechanisms contribute to the onset and severity of SM. However, little is known to date about the metabolic underpinnings underlying SM aggressiveness, which has thus far impeded the development of strategies to leverage metabolic dependencies when existing KIT-targeted treatments fail. Here, we show that plasma metabolomic profiles were able to discriminate indolent from advanced forms of the disease. We identified N-acetyl-d-glucosamine (GlcNAc) as the most predictive metabolite of SM severity. High plasma levels of GlcNAc in patients with advanced SM correlated with the activation of the GlcNAc-fed hexosamine biosynthesis pathway in patients BM aspirates and purified BM MCs. At the functional level, GlcNAc enhanced human neoplastic MCs proliferation and promoted rapid health deterioration in a humanized mouse model of SM. In addition, in the presence of GlcNAc, immunoglobulin E-stimulated MCs triggered enhanced release of proinflammatory cytokines and a stronger acute response in a mouse model of passive cutaneous anaphylaxis. Mechanistically, elevated GlcNAc levels promoted the transcriptional accessibility of chromatin regions that contain genes encoding mediators of receptor tyrosine kinases cascades and inflammatory responses, thus leading to a more aggressive phenotype. Therefore, GlcNAc is an oncometabolite driver of SM aggressiveness. This study suggests the therapeutic potential for targeting metabolic pathways in MC-related diseases to manipulate MCs effector functions.
Collapse
Affiliation(s)
- Julie Agopian
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Unité Mixte de Recherche (UMR) 258 Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Institut Paoli-Calmettes, Equipe Labellisée Ligue Contre le Cancer, Marseille, France
- French Reference Center for Mastocytosis (CEREMAST), Paris, France
| | - Quentin Da Costa
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Unité Mixte de Recherche (UMR) 258 Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Institut Paoli-Calmettes, Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - Quang Vo Nguyen
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Unité Mixte de Recherche (UMR) 258 Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Institut Paoli-Calmettes, Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - Giulia Scorrano
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Unité Mixte de Recherche (UMR) 258 Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Institut Paoli-Calmettes, Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - Paraskevi Kousteridou
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Unité Mixte de Recherche (UMR) 258 Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Institut Paoli-Calmettes, Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - Min Yuan
- Division of Signal Transduction, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Rabie Chelbi
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Unité Mixte de Recherche (UMR) 258 Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Institut Paoli-Calmettes, Equipe Labellisée Ligue Contre le Cancer, Marseille, France
- Inovarion, Paris, France
| | - Armelle Goubard
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Unité Mixte de Recherche (UMR) 258 Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Institut Paoli-Calmettes, Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - Remy Castellano
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Unité Mixte de Recherche (UMR) 258 Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Institut Paoli-Calmettes, Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - Julien Maurizio
- Centre d'Immunologie de Marseille-Luminy (CIML), INSERM U631, CNRS UMR 6102, Aix-Marseille Université, Marseille, France
| | - Cristina Teodosio
- Department of Immunohematology, Leiden University Medical Center, ZC Leiden, The Netherlands
| | - Paulo De Sepulveda
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Unité Mixte de Recherche (UMR) 258 Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Institut Paoli-Calmettes, Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - John M Asara
- Division of Signal Transduction, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Alberto Orfao
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), Centro de Investigación Biomédica en Red Cáncer (CIBERONC), University of Salamanca, Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Spanish Network on Mastocytosis (REMA), Toledo, Spain; and
| | - Olivier Hermine
- French Reference Center for Mastocytosis (CEREMAST), Paris, France
- Institut Imagine, INSERM U1163, CNRS Equipe de Recherche Labelisée (ERL) 8654, Paris Université, Service d'Hématologie Clinique, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Patrice Dubreuil
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Unité Mixte de Recherche (UMR) 258 Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Institut Paoli-Calmettes, Equipe Labellisée Ligue Contre le Cancer, Marseille, France
- French Reference Center for Mastocytosis (CEREMAST), Paris, France
| | - Fabienne Brenet
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Unité Mixte de Recherche (UMR) 258 Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Institut Paoli-Calmettes, Equipe Labellisée Ligue Contre le Cancer, Marseille, France
- French Reference Center for Mastocytosis (CEREMAST), Paris, France
| |
Collapse
|
26
|
Karan D. CCL23 in Balancing the Act of Endoplasmic Reticulum Stress and Antitumor Immunity in Hepatocellular Carcinoma. Front Oncol 2021; 11:727583. [PMID: 34671553 PMCID: PMC8522494 DOI: 10.3389/fonc.2021.727583] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/17/2021] [Indexed: 11/15/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is a cellular process in response to stress stimuli in protecting functional activities. However, sustained hyperactive ER stress influences tumor growth and development. Hepatocytes are enriched with ER and highly susceptible to ER perturbations and stress, which contribute to immunosuppression and the development of aggressive and drug-resistant hepatocellular carcinoma (HCC). ER stress-induced inflammation and tumor-derived chemokines influence the immune cell composition at the tumor site. Consequently, a decrease in the CCL23 chemokine in hepatic tumors is associated with poor survival of HCC patients and could be a mechanism hepatic tumor cells use to evade the immune system. This article describes the prospective role of CCL23 in alleviating ER stress and its impact on the HCC tumor microenvironment in promoting antitumor immunity. Moreover, approaches to reactivate CCL23 combined with immune checkpoint blockade or chemotherapy drugs may provide novel opportunities to target hepatocellular carcinoma.
Collapse
Affiliation(s)
- Dev Karan
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
27
|
Damasceno D, Almeida J, Teodosio C, Sanoja-Flores L, Mayado A, Pérez-Pons A, Puig N, Arana P, Paiva B, Solano F, Romero A, Matarraz S, van den Bossche WBL, Flores-Montero J, Durie B, van Dongen JJM, Orfao A. Monocyte Subsets and Serum Inflammatory and Bone-Associated Markers in Monoclonal Gammopathy of Undetermined Significance and Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13061454. [PMID: 33810169 PMCID: PMC8004952 DOI: 10.3390/cancers13061454] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Monocyte/macrophages have been shown to be altered in monoclonal gammopathy of undetermined significance (MGUS), smoldering (SMM) and active multiple myeloma (MM), with an impact on the disruption of the homeostasis of the normal bone marrow (BM) microenvironment. METHODS We investigated the distribution of different subsets of monocytes (Mo) in blood and BM of newly-diagnosed untreated MGUS (n = 23), SMM (n = 14) and MM (n = 99) patients vs. healthy donors (HD; n = 107), in parallel to a large panel of cytokines and bone-associated serum biomarkers. RESULTS Our results showed normal production of monocyte precursors and classical Mo (cMo) in MGUS, while decreased in SMM and MM (p ≤ 0.02), in association with lower blood counts of recently-produced CD62L+ cMo in SMM (p = 0.004) and of all subsets of (CD62L+, CD62L- and FcεRI+) cMo in MM (p ≤ 0.02). In contrast, intermediate and end-stage non-classical Mo were increased in BM of MGUS (p ≤ 0.03), SMM (p ≤ 0.03) and MM (p ≤ 0.002), while normal (MGUS and SMM) or decreased (MM; p = 0.01) in blood. In parallel, increased serum levels of interleukin (IL)1β were observed in MGUS (p = 0.007) and SMM (p = 0.01), higher concentrations of serum IL8 were found in SMM (p = 0.01) and MM (p = 0.002), and higher serum IL6 (p = 0.002), RANKL (p = 0.01) and bone alkaline phosphatase (BALP) levels (p = 0.01) with decreased counts of FcεRI+ cMo, were restricted to MM presenting with osteolytic lesions. This translated into three distinct immune/bone profiles: (1) normal (typical of HD and most MGUS cases); (2) senescent-like (increased IL1β and/or IL8, found in a minority of MGUS, most SMM and few MM cases with no bone lesions); and (3) pro-inflammatory-high serum IL6, RANKL and BALP with significantly (p = 0.01) decreased blood counts of immunomodulatory FcεRI+ cMo-, typical of MM presenting with bone lesions. CONCLUSIONS These results provide new insight into the pathogenesis of plasma cell neoplasms and the potential role of FcεRI+ cMo in normal bone homeostasis.
Collapse
Affiliation(s)
- Daniela Damasceno
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (D.D.); (J.A.); (A.M.); (A.P.-P.); (S.M.); (J.F.-M.)
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (B.P.)
| | - Julia Almeida
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (D.D.); (J.A.); (A.M.); (A.P.-P.); (S.M.); (J.F.-M.)
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (B.P.)
| | - Cristina Teodosio
- Leiden University Medical Center, Department of Immunology, 2333 ZA Leiden, The Netherlands; (C.T.); (W.B.L.v.d.B.); (J.J.M.v.D.)
| | - Luzalba Sanoja-Flores
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (B.P.)
- Institute of Biomedicine of Seville, Department of Hematology, University Hospital Virgen del Rocío of the Consejo Superior de Investigaciones Científicas (CSIC), University of Seville, 41013 Seville, Spain
| | - Andrea Mayado
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (D.D.); (J.A.); (A.M.); (A.P.-P.); (S.M.); (J.F.-M.)
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (B.P.)
| | - Alba Pérez-Pons
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (D.D.); (J.A.); (A.M.); (A.P.-P.); (S.M.); (J.F.-M.)
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (B.P.)
| | - Noemi Puig
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (B.P.)
- Service of Hematology, University Hospital of Salamanca (CAUSA) and IBSAL, 37007 Salamanca, Spain
| | - Paula Arana
- Regulation of the Immune System Group, Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, Plaza de Cruces 12, 48903 Barakaldo, Spain;
| | - Bruno Paiva
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (B.P.)
- Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Fernando Solano
- Hematology Service, Hospital Nuestra Señora del Prado, Talavera de la Reina, 45600 Toledo, Spain;
| | - Alfonso Romero
- Primary Health Care Center “Miguel Armijo”, Primary Health Care of Salamanca, Conserjería de Sanidad de Castilla y León (SACYL), 37007 Salamanca, Spain;
| | - Sergio Matarraz
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (D.D.); (J.A.); (A.M.); (A.P.-P.); (S.M.); (J.F.-M.)
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (B.P.)
| | - Wouter B. L. van den Bossche
- Leiden University Medical Center, Department of Immunology, 2333 ZA Leiden, The Netherlands; (C.T.); (W.B.L.v.d.B.); (J.J.M.v.D.)
- Department of Immunology, Erasmus University Medical Center, 3015 GA Rotterdam, The Netherlands
| | - Juan Flores-Montero
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (D.D.); (J.A.); (A.M.); (A.P.-P.); (S.M.); (J.F.-M.)
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (B.P.)
| | - Brian Durie
- Centro del Cáncer Cedars-Sinai Samuel Oschin, Los Angeles, CA 90048, USA;
| | - Jacques J. M. van Dongen
- Leiden University Medical Center, Department of Immunology, 2333 ZA Leiden, The Netherlands; (C.T.); (W.B.L.v.d.B.); (J.J.M.v.D.)
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca and Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (D.D.); (J.A.); (A.M.); (A.P.-P.); (S.M.); (J.F.-M.)
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (B.P.)
- Correspondence:
| |
Collapse
|
28
|
Meszaros M, Horvath P, Kis A, Kunos L, Tarnoki AD, Tarnoki DL, Lazar Z, Bikov A. Circulating levels of clusterin and complement factor H in patients with obstructive sleep apnea. Biomark Med 2021; 15:323-330. [PMID: 33666516 DOI: 10.2217/bmm-2020-0533] [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: 11/21/2022] Open
Abstract
Aim: Obstructive sleep apnea (OSA) activates the complement system; however, the levels of membrane attack complex (MAC) are unaltered suggesting regulatory mechanisms. Our aim was to investigate complement factor H (CFH) and clusterin, two important complement regulators in OSA. Materials & methods: We analyzed clusterin and CFH levels in plasma of 86 patients with OSA and 33 control subjects. Results: There was no difference in CFH levels between patients (1099.4/784.6-1570.5/μg/ml) and controls (1051.4/652.0-1615.1/μg/ml, p = 0.72). Clusterin levels were higher in patients with OSA (309.7/217.2-763.2/μg/ml vs 276.1/131.0-424.3/μg/ml, p = 0.048) with a trend for a positive correlation with disease severity (p = 0.073). Conclusion: Increase in clusterin levels may be protective in OSA by blocking the MAC formation.
Collapse
Affiliation(s)
- Martina Meszaros
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Peter Horvath
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Adrian Kis
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Laszlo Kunos
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Adam D Tarnoki
- Department of Radiology, Oncologic Imaging Diagnostic Center, National Institute of Oncology, Budapest, Hungary.,Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - David L Tarnoki
- Department of Radiology, Oncologic Imaging Diagnostic Center, National Institute of Oncology, Budapest, Hungary.,Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Zsofia Lazar
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Andras Bikov
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK.,Division of Infection, Immunity & Respiratory Medicine, University of Manchester, UK
| |
Collapse
|
29
|
Theoharides TC, Cholevas C, Polyzoidis K, Politis A. Long-COVID syndrome-associated brain fog and chemofog: Luteolin to the rescue. Biofactors 2021; 47:232-241. [PMID: 33847020 PMCID: PMC8250989 DOI: 10.1002/biof.1726] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/01/2021] [Indexed: 01/08/2023]
Abstract
COVID-19 leads to severe respiratory problems, but also to long-COVID syndrome associated primarily with cognitive dysfunction and fatigue. Long-COVID syndrome symptoms, especially brain fog, are similar to those experienced by patients undertaking or following chemotherapy for cancer (chemofog or chemobrain), as well in patients with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) or mast cell activation syndrome (MCAS). The pathogenesis of brain fog in these illnesses is presently unknown but may involve neuroinflammation via mast cells stimulated by pathogenic and stress stimuli to release mediators that activate microglia and lead to inflammation in the hypothalamus. These processes could be mitigated by phytosomal formulation (in olive pomace oil) of the natural flavonoid luteolin.
Collapse
Affiliation(s)
- Theoharis C. Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of ImmunologyTufts University School of MedicineBostonMassachusettsUSA
- School of Graduate Biomedical SciencesTufts University School of MedicineBostonMassachusettsUSA
- Department of Internal MedicineTufts University School of Medicine and Tufts Medical CenterBostonMassachusettsUSA
- Department of PsychiatryTufts University School of Medicine and Tufts Medical CenterBostonMassachusettsUSA
- BrainGateThessalonikiGreece
| | | | | | - Antonios Politis
- First Department of PsychiatryEginition Hospital, National and Kapodistrian UniversityAthensGreece
| |
Collapse
|
30
|
Nedoszytko B, Arock M, Lyons JJ, Bachelot G, Schwartz LB, Reiter A, Jawhar M, Schwaab J, Lange M, Greiner G, Hoermann G, Niedoszytko M, Metcalfe DD, Valent P. Clinical Impact of Inherited and Acquired Genetic Variants in Mastocytosis. Int J Mol Sci 2021; 22:ijms22010411. [PMID: 33401724 PMCID: PMC7795405 DOI: 10.3390/ijms22010411] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 02/08/2023] Open
Abstract
Mastocytosis is a rare and complex disease characterized by expansion of clonal mast cells (MC) in skin and/or various internal organ systems. Involvement of internal organs leads to the diagnosis of systemic mastocytosis (SM). The WHO classification divides SM into indolent SM, smoldering SM and advanced SM variants, including SM with an associated hematologic neoplasm, aggressive SM, and MC leukemia. Historically, genetic analysis of individuals with pure cutaneous mastocytosis (CM) and SM have focused primarily on cohort studies of inherited single nucleotide variants and acquired pathogenic variants. The most prevalent pathogenic variant (mutation) in patients with SM is KIT p.D816V, which is detectable in most adult patients. Other somatic mutations have also been identified-especially in advanced SM-in TET2, SRSF2, ASXL1, RUNX1, CBL and JAK2, and shown to impact clinical and cellular phenotypes. Although only small patient cohorts have been analyzed, disease associations have also been identified in several germline variants within genes encoding certain cytokines or their receptors (IL13, IL6, IL6R, IL31, IL4R) and toll-like receptors. More recently, an increased prevalence of hereditary alpha-tryptasemia (HαT) caused by increased TPSAB1 copy number encoding alpha-tryptase has been described in patients with SM. Whereas HαT is found in 3-6% of general Western populations, it is identified in up to 17% of patients with SM. In the current manuscript we review the prevalence, functional role and clinical impact of various germline and somatic genetic variants in patients with mastocytosis.
Collapse
Affiliation(s)
- Boguslaw Nedoszytko
- Department of Dermatology, Allergology and Venereology, Medical University of Gdansk, 80-211 Gdansk, Poland;
- Correspondence:
| | - Michel Arock
- Department of Hematology, APHP, Hôpital Pitié-Salpêtrière and Sorbonne University, 75013 Paris, France; (M.A.); (G.B.)
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Cell Death and Drug Resistance in Hematological Disorders Team, 75006 Paris, France
| | - Jonathan J. Lyons
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-188, USA; (J.J.L.); (D.D.M.)
| | - Guillaume Bachelot
- Department of Hematology, APHP, Hôpital Pitié-Salpêtrière and Sorbonne University, 75013 Paris, France; (M.A.); (G.B.)
| | - Lawrence B. Schwartz
- Department of Internal Medicine, Division of Rheumatology, Allergy & Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Andreas Reiter
- University Hospital Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.R.); (M.J.); (J.S.)
| | - Mohamad Jawhar
- University Hospital Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.R.); (M.J.); (J.S.)
| | - Juliana Schwaab
- University Hospital Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.R.); (M.J.); (J.S.)
| | - Magdalena Lange
- Department of Dermatology, Allergology and Venereology, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Georg Greiner
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria;
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria; (G.H.); (P.V.)
- Ihr Labor, Medical Diagnostic Laboratories, 1220 Vienna, Austria
| | - Gregor Hoermann
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria; (G.H.); (P.V.)
- MLL Munich Leukemia Laboratory, 81377 Munich, Germany
| | - Marek Niedoszytko
- Department of Allergology, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Dean D. Metcalfe
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-188, USA; (J.J.L.); (D.D.M.)
| | - Peter Valent
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria; (G.H.); (P.V.)
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
31
|
|
32
|
Valent P. KIT D816V and the cytokine storm in mastocytosis: production and role of interleukin-6. Haematologica 2020; 105:5-6. [PMID: 31894094 DOI: 10.3324/haematol.2019.234864] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology .,Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
33
|
Abstract
Bone pathology can be challenging because the skeleton is a living tissue prone to developing a diverse array of inflammatory, metabolic, genetic, reactive, circulatory, and neoplastic abnormalities. Several areas of bone pathology are particularly difficult or problematic for hematopathologists given the close resemblance of some hematologic entities to primary/metastatic bone lesions; examples include plasmacytic disorders versus osteoblastic tumors and lymphoma/leukemia versus round cell tumors of bone. This article provides a conceptual and practical overview of selective bone disorders commonly encountered in the differential diagnosis of hematologic diseases.
Collapse
Affiliation(s)
- Deniz Peker
- Department of Pathology, University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL 35249, USA
| | - Shi Wei
- Department of Pathology, University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL 35249, USA
| | - Gene P Siegal
- Department of Pathology, University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL 35249, USA.
| |
Collapse
|
34
|
Frequency and prognostic impact of KIT and other genetic variants in indolent systemic mastocytosis. Blood 2019; 134:456-468. [DOI: 10.1182/blood.2018886507] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Abstract
Indolent systemic mastocytosis (ISM) patients have a normal life expectancy, except in the 5% to 10% of cases that progress to more advanced SM (advSM), which has a significantly poorer outcome. Mutations in genes other than KIT frequently found in myeloid neoplasms have been associated with a poorer outcome among advSM, whereas limited information exists about their frequency and prognostic impact in ISM. We investigated the frequency and prognostic impact of variants in 18 genes, found to be altered in advSM, in 322 ISM patients (median follow-up, 5.7 years) divided into discovery (n = 200) and validation (n = 122) cohorts. Overall, 71 genetic variants were detected in 55 of 322 (17%) patients. Mutated ISM cases, particularly those carrying ASXL1, RUNX1, and/or DNMT3A (A/R/D) pathogenic variant allele frequencies (VAFs) ≥ 30%, exhibited significantly shortened (P < .001) progression-free survival (PFS) and overall survival (OS). Multivariate analysis showed that serum β2-microglobulin (sβ2M) levels > 2.5 µg/mL (hazard ratio [HR], 9.8; P = .001), together with a KIT D816V VAF ≥ 1% in bone marrow (BM) (HR, 10.1; P = .02) and pathogenic variants of A/R/D VAFs ≥ 30% (HR, 4.2; P = .02), were the best combination of independent predictors for PFS. In turn, A/R/D gene pathogenic VAF ≥ 30% was the only independent predictor for OS (HR, 51.8; P < .001). Based on these variables, 2 scoring systems were constructed for risk stratification of ISM at diagnosis with significantly different 10-year PFS (100%, 91%, 0% for scores of 0, 1, ≥2, respectively) and OS (100% and 50% for scores of 0 and 1) rates.
Collapse
|
35
|
Jannat A, Khan M, Shabbir M, Badshah Y. Expression of suppressor of cytokine signaling 3 (SOCS3) and interleukin-6 (-174-G/C) polymorphism in atopic conditions. PLoS One 2019; 14:e0219084. [PMID: 31251775 PMCID: PMC6599118 DOI: 10.1371/journal.pone.0219084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/15/2019] [Indexed: 11/24/2022] Open
Abstract
Hypersensitivity of the immune system is caused by elevated immunoglobulin E (IgE) levels in the serum, in response to a discrete allergen leading to allergic reactions. IgE-mediated inflammation is regulated by the cascade of defense related signaling molecules including interleukin-6 (IL-6) that plays pivotal role in the survival and maturation of mast cells during an allergic reaction. IL-6 mediated defense responses are tightly regulated by Suppressor of Cytokine Signaling 3 (SOCS3), an inhibitory molecules of Janus Kinase-Signal Transducers and Activators of Transcription (JAK-STAT) signaling, in a negative feedback mechanism. The given study focuses on the assessment of crosstalk between SOCS3 and IL-6 to unravel the molecular significance of SOCS3 and IL-6 in the diagnosis and prognosis of allergy. The expression study of SOCS3 through real-time PCR analysis revealed, a 5.9 mean fold increase in SOCS3 expression in atopic cases in comparison to control cases. Moreover, IL-6 has, also, been found significantly enhanced in the serum level of atopic cases (26.4 pg/ml) as compared to control cases (3.686 pg/ml). Female population was found to be at a higher risk to develop atopic condition than male population as females exhibited higher expression of both SOCS3 and IL-6 than males. Furthermore, the polymorphic study of IL-6 promoter region (IL-6 174-G/C) in atopic population has reasserted the importance of SOCS3 and IL-6 in the diagnosis and prognosis of allergy. Expression of SOCS3 and IL-6 serum levels were found to be highly correlated. Therefore establishing the role of IL-6 (-174-G/C) polymorphism on the expression of SOCS3 and IL-6 in atopic cases. Notably, the study established SOCS3 and IL-6 as potential targets for the diagnosis/prognosis of allergy and for the development of reliable therapeutic strategies to control atopic conditions in the near future.
Collapse
Affiliation(s)
- Arooma Jannat
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Maryam Khan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Maria Shabbir
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Yasmin Badshah
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
- * E-mail:
| |
Collapse
|
36
|
Valent P, Akin C, Gleixner KV, Sperr WR, Reiter A, Arock M, Triggiani M. Multidisciplinary Challenges in Mastocytosis and How to Address with Personalized Medicine Approaches. Int J Mol Sci 2019; 20:E2976. [PMID: 31216696 PMCID: PMC6627900 DOI: 10.3390/ijms20122976] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/05/2019] [Accepted: 06/16/2019] [Indexed: 12/27/2022] Open
Abstract
Mastocytosis is a hematopoietic neoplasm defined by abnormal expansion and focal accumulation of clonal tissue mast cells in various organ-systems. The disease exhibits a complex pathology and an equally complex clinical behavior. The classification of the World Health Organization (WHO) divides mastocytosis into cutaneous forms, systemic variants, and localized mast cell tumors. In >80% of patients with systemic mastocytosis (SM), a somatic point mutation in KIT at codon 816 is found. Whereas patients with indolent forms of the disease have a normal or near-normal life expectancy, patients with advanced mast cell neoplasms, including aggressive SM and mast cell leukemia, have a poor prognosis with short survival times. In a majority of these patients, multiple somatic mutations and/or an associated hematologic neoplasm, such as a myeloid leukemia, may be detected. Independent of the category of mastocytosis and the serum tryptase level, patients may suffer from mediator-related symptoms and/or osteopathy. Depending on the presence of co-morbidities, the symptomatology in such patients may be mild, severe or even life-threatening. Most relevant co-morbidities in such patients are IgE-dependent allergies, psychiatric, psychological or mental problems, and vitamin D deficiency. The diagnosis and management of mastocytosis is an emerging challenge in clinical practice and requires vast knowledge, a multidisciplinary approach, and personalized medicine procedures. In this article, the current knowledge about mastocytosis is reviewed with special emphasis on the multidisciplinary aspects of the disease and related challenges in daily practice.
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria.
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Cem Akin
- Division of Allergy and Clinical Immunology, University of Michigan, Ann Arbor, MI 48106, USA.
| | - Karoline V Gleixner
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria.
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Wolfgang R Sperr
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria.
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Andreas Reiter
- III. Medizinische Klinik, Universitätsmedizin Mannheim, 68167 Mannheim, Germany.
| | - Michel Arock
- Department of Hematological Biology, Pitié-Salpêtrière Hospital, Pierre et Marie Curie University (UPMC), 75005 Paris, France.
| | - Massimo Triggiani
- Division of Allergy and Clinical Immunology, University of Salerno, 84131 Salerno, Italy.
| |
Collapse
|
37
|
Theoharides TC, Tsilioni I, Ren H. Recent advances in our understanding of mast cell activation - or should it be mast cell mediator disorders? Expert Rev Clin Immunol 2019; 15:639-656. [PMID: 30884251 PMCID: PMC7003574 DOI: 10.1080/1744666x.2019.1596800] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 03/14/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION An increasing number of patients present with multiple symptoms affecting many organs including the brain due to multiple mediators released by mast cells. These unique tissue immune cells are critical for allergic reactions triggered by immunoglobulin E (IgE), but are also stimulated (not activated) by immune, drug, environmental, food, infectious, and stress triggers, leading to secretion of multiple mediators often without histamine and tryptase. The presentation, diagnosis, and management of the spectrum of mast cell disorders are very confusing. As a result, neuropsychiatric symptoms have been left out, and diagnostic criteria made stricter excluding most patients. Areas covered: A literature search was performed on papers published between January 1990 and November 2018 using MEDLINE. Terms used were activation, antihistamines, atopy, autism, brain fog, heparin, KIT mutation, IgE, inflammation, IL-6, IL-31, IL-37, luteolin, mast cells, mastocytosis, mediators, mycotoxins, release, secretion, tetramethoxyluteolin, and tryptase. Expert opinion: Conditions associated with elevated serum or urine levels of any mast cell mediator, in the absence of comorbidities that could explain elevated levels, should be considered 'Mast Cell Mediator Disorders (MCMD).' Emphasis should be placed on the identification of unique mast cell mediators, and development of drugs or supplements that inhibit their release.
Collapse
Affiliation(s)
- Theoharis C. Theoharides
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Department of Internal Medicine, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, USA
- Department of Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, USA
| | - Irene Tsilioni
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Huali Ren
- Department of Otolaryngology, Beijing Electric Power Hospital, Beijing, China
| |
Collapse
|
38
|
Tobío A, Bandara G, Morris DA, Kim DK, O'Connell MP, Komarow HD, Carter MC, Smrz D, Metcalfe DD, Olivera A. Oncogenic D816V-KIT signaling in mast cells causes persistent IL-6 production. Haematologica 2019; 105:124-135. [PMID: 30948489 PMCID: PMC6939509 DOI: 10.3324/haematol.2018.212126] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/02/2019] [Indexed: 12/19/2022] Open
Abstract
Persistent dysregulation of IL-6 production and signaling have been implicated in the pathology of various cancers. In systemic mastocytosis, increased serum levels of IL-6 associate with disease severity and progression, although the mechanisms involved are not well understood. Since systemic mastocytosis often associates with the presence in hematopoietic cells of a somatic gain-of-function variant in KIT, D816V-KIT, we examined its potential role in IL-6 upregulation. Bone marrow mononuclear cultures from patients with greater D816V allelic burden released increased amounts of IL-6 which correlated with the percentage of mast cells in the cultures. Intracellular IL-6 staining by flow cytometry and immunofluorescence was primarily associated with mast cells and suggested a higher percentage of IL-6 positive mast cells in patients with higher D816V allelic burden. Furthermore, mast cell lines expressing D816V-KIT, but not those expressing normal KIT or other KIT variants, produced constitutively high IL-6 amounts at the message and protein levels. We further demonstrate that aberrant KIT activity and signaling are critical for the induction of IL-6 and involve STAT5 and PI3K pathways but not STAT3 or STAT4. Activation of STAT5A and STAT5B downstream of D816V-KIT was mediated by JAK2 but also by MEK/ERK1/2, which not only promoted STAT5 phosphorylation but also its long-term transcription. Our study thus supports a role for mast cells and D816V-KIT activity in IL-6 dysregulation in mastocytosis and provides insights into the intracellular mechanisms. The findings contribute to a better understanding of the physiopathology of mastocytosis and suggest the importance of therapeutic targeting of these pathways.
Collapse
Affiliation(s)
- Araceli Tobío
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Geethani Bandara
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Denise A Morris
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Do-Kyun Kim
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael P O'Connell
- Genetics and Pathogenesis of Allergy Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hirsh D Komarow
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Melody C Carter
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniel Smrz
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dean D Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ana Olivera
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
39
|
Castells M, Butterfield J. Mast Cell Activation Syndrome and Mastocytosis: Initial Treatment Options and Long-Term Management. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2019; 7:1097-1106. [DOI: 10.1016/j.jaip.2019.02.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 02/06/2023]
|
40
|
Orsolini G, Viapiana O, Rossini M, Bonifacio M, Zanotti R. Bone Disease in Mastocytosis. Immunol Allergy Clin North Am 2019; 38:443-454. [PMID: 30007462 DOI: 10.1016/j.iac.2018.04.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Systemic mastocytosis can give very different bone pictures: from osteosclerosis to osteoporosis. Osteoporosis is one of the most frequent manifestations particularly in adults and the most clinical relevant. It is often complicated by a high recurrence of mainly vertebral fragility fractures. The main factor of bone loss is the osteoclast with a relative or absolute predominance of bone resorption. The RANK-RANKL pathway seems of key importance, but histamine and other cytokines also play a significant role in the process. The predominance of resorption made bisphosphonates, as anti-resorptive drugs, the most rational treatment of bone involvement in systemic mastocytosis.
Collapse
|
41
|
Mastocytosis-derived extracellular vesicles exhibit a mast cell signature, transfer KIT to stellate cells, and promote their activation. Proc Natl Acad Sci U S A 2018; 115:E10692-E10701. [PMID: 30352845 DOI: 10.1073/pnas.1809938115] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) have been implicated in the development and progression of hematological malignancies. We thus examined serum samples from patients with systemic mastocytosis (SM) and found EVs with a mast cell signature including the presence of tryptase, FcεRI, MRGX2, and KIT. The concentration of these EVs correlated with parameters of disease including levels of serum tryptase, IL-6, and alkaline phosphatase and physical findings including hepatosplenomegaly. Given reports that EVs from one cell type may influence another cell's behavior, we asked whether SM-EVs might affect hepatic stellate cells (HSCs), based on the abnormal liver pathology associated with mastocytosis. We found that KIT was transferred from SM-EVs into an HSC line eliciting proliferation, cytokine production, and differentiation, processes that have been associated with liver pathology. These effects were reduced by KIT inhibition or neutralization and recapitulated by enforced expression of KIT or constitutively active D816V-KIT, a gain-of-function variant associated with SM. Furthermore, HSCs in liver from mice injected with SM-EVs had increased expression of α-SMA and human KIT, particularly around portal areas, compared with mice injected with EVs from normal individuals, suggesting that SM-EVs can also initiate HSC activation in vivo. Our data are thus consistent with the conclusion that SM-EVs have the potential to influence cells outside the hematological compartment and that therapeutic approaches for treatment of SM may be effective in part through inhibition of effects of EVs on target tissues, findings important both to understanding complex disease pathology and in developing interventional agents for the treatment of hematologic diseases.
Collapse
|
42
|
Hermans MAW, Schrijver B, van Holten-Neelen CCPA, Gerth van Wijk R, van Hagen PM, van Daele PLA, Dik WA. The JAK1/JAK2- inhibitor ruxolitinib inhibits mast cell degranulation and cytokine release. Clin Exp Allergy 2018; 48:1412-1420. [PMID: 29939445 DOI: 10.1111/cea.13217] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND Mastocytosis is characterized by the accumulation of aberrant mast cells (MC). Patients suffering from mastocytosis suffer from a wide range of symptoms due to increased levels of MC mediators. It would therefore be of great benefit to inhibit MC mediator release. However, to date there are few drugs available that are known to effectively lower MC mediator levels. The evidence for the involvement of the janus kinase 2 (JAK2)-signal transducer and activation of transcription 5 (STAT5) signalling pathway in MC activation is slowly accumulating. Interference with the JAK2-STAT5 pathway might inhibit MC mediator release. Ruxolitinib, a JAK1/JAK2 inhibitor, indeed decreases symptoms like pruritus and fatigue in patients with myeloproliferative neoplasms. Yet, detailed studies on how ruxolitinib affects human mast cell activity are lacking. OBJECTIVE To investigate the effect of JAK1/2-inhibition with ruxolitinib in the human mast cell lines LAD2 and HMC1. METHODS LAD2 and HMC1 were stimulated with substance P, codeine or the calcium ionophore A23817. The effect of ruxolitinib on mast cell degranulation (via measurement of β-hexosaminidase, histamine release and CD63 membrane expression) and IL-6, IL-13, MCP-1 and TNF-α production was investigated. The involvement of STAT5 activation was explored using the selective STAT5 inhibitor pimozide. RESULTS Ruxolitinib effectively inhibited codeine- and substance P-induced degranulation in a concentration-dependent manner. Ruxolitinib also significantly inhibited the production of IL-6, TNF-α and MCP-1 as induced by A23817 and substance P. Selective STAT5 inhibition with pimozide resulted in diminished degranulation and inhibition of cytokine production as induced by A23817 and substance P. CONCLUSIONS & CLINICAL RELEVANCE This study demonstrates that the JAK1/JAK2 inhibitor ruxolitinib can inhibit MCactivity, possibly through prevention of STAT5 activation. This renders the JAK-STAT pathway as an interesting target for therapy to release symptom burden in mastocytosis and many other MC mediator-related diseases.
Collapse
Affiliation(s)
- Maud A W Hermans
- Department of Internal Medicine, Sections of Allergy and Clinical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Benjamin Schrijver
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands.,Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Conny C P A van Holten-Neelen
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands.,Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Roy Gerth van Wijk
- Department of Internal Medicine, Sections of Allergy and Clinical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - P Martin van Hagen
- Department of Internal Medicine, Sections of Allergy and Clinical Immunology, Erasmus MC, Rotterdam, The Netherlands.,Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Paul L A van Daele
- Department of Internal Medicine, Sections of Allergy and Clinical Immunology, Erasmus MC, Rotterdam, The Netherlands.,Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Willem A Dik
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands.,Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
43
|
Butterfield JH, Ravi A, Pongdee T. Mast Cell Mediators of Significance in Clinical Practice in Mastocytosis. Immunol Allergy Clin North Am 2018; 38:397-410. [DOI: 10.1016/j.iac.2018.04.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
44
|
McHale C, Mohammed Z, Deppen J, Gomez G. Interleukin-6 potentiates FcεRI-induced PGD 2 biosynthesis and induces VEGF from human in situ-matured skin mast cells. Biochim Biophys Acta Gen Subj 2018; 1862:1069-1078. [PMID: 29410184 DOI: 10.1016/j.bbagen.2018.01.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/18/2018] [Accepted: 01/31/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Interleukin-6 is a gp130 utilizing cytokine that is consistently associated with allergic diseases like asthma and urticaria in humans where mast cells are known to play a critical role. However, the role of IL-6 in allergic disease in not known. IL-6 was reported to enhance degranulation of in vitro-derived mast cells, but the effect of IL-6 on mediator release from human in situ-matured tissue-isolated mast cells had not been reported. METHODS Human mature mast cells were isolated and purified from normal skin tissue from different donors. The expression of surface-expressed IL-6 receptors was demonstrated by flow cytometry. The effect of IL-6 on FcεRI-induced degranulation, PGD2 biosynthesis, and cytokine production was determined with β‑hexosaminidase release assay, Western blotting, quantitative real-time PCR, and ELISA. The small molecule inhibitor of STAT-3, C188-9, was used to demonstrate STAT3 dependency. RESULTS IL-6 significantly potentiated FcεRI-induced PGD2 biosynthesis, but had no effect on degranulation. IL-6 also induced VEGF gene expression and protein secretion, and enhanced FcεRI-induced IL-8 production. Mechanistically, IL-6 enhanced FcεRI-induced COX‑2 expression, PGD2 biosynthesis, and VEGF production in a STAT3 dependent manner. CONCLUSION Here, we demonstrate that IL-6 is a potentiator of FcεRI-induced PGD2 biosynthesis, and can induce or enhance production of pro-angiogenesis factors VEGF and IL-8 from human in situ-matured skin mast cells. GENERAL SIGNIFICANCE These findings from this study indicate that IL-6 contributes to human allergic disease by enhancing the production of inflammatory PGD2 from tissue-resident mast cells. Moreover, the data suggest a novel role for IL-6 in mast cell-mediated angiogenesis.
Collapse
Affiliation(s)
- Cody McHale
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA.
| | - Zahraa Mohammed
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA.
| | - Juline Deppen
- Department of Biomedical Engineering, University of South Carolina School of Medicine, Columbia, SC, USA.
| | - Gregorio Gomez
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA.
| |
Collapse
|
45
|
He Y, Mao C, Wen H, Chen Z, Lai T, Li L, Lu W, Wu H. Influence of ad Libitum Feeding of Piglets With Bacillus Subtilis Fermented Liquid Feed on Gut Flora, Luminal Contents and Health. Sci Rep 2017; 7:44553. [PMID: 28291252 PMCID: PMC5349548 DOI: 10.1038/srep44553] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 02/10/2017] [Indexed: 12/25/2022] Open
Abstract
Some scholars caution that long-term ad libitum feeding with probiotic fermented food poses potential health risks to baby animals. We conducted a feeding experiment to investigate the influence of ad libitum feeding of pre-and post-weaned piglets with a Bacillus subtilis fermented diet on the gut microbiome, gut metabolomic profiles, bile acid metabolism, proinflammatory cytokines and faecal consistency. Compared with piglets fed a Bacillus subtilis-supplemented pellet diet, piglets fed the Bacillus subtilis fermented liquid diet had lower intestinal bacterial diversity (P > 0.05), higher intestinal fungal diversity (P > 0.05), more Firmicutes (P > 0.05), fewer Bacteroidetes, Actinobacteria and Proteobacteria (P > 0.05), higher concentrations of 3-hydroxypropionic acid (P < 0.05), orotic acid (P < 0.05), interleukin-6 (P < 0.01), lactic acid (P < 0.01), deoxycholic acid (P > 0.05) and lithocholic acid (P < 0.01) and a higher incidence of diarrhoea (P > 0.05). The data show that ad libitum feeding of piglets with a Bacillus subtilis fermented liquid diet during the suckling and early post-weaning periods promotes the growth of lactic acid bacteria, bile salt hydrolase-active bacteria and 7a-dehydroxylase-active bacteria in the intestinal lumen; disturbs the normal production of lactic acid, orotic acid and unconjugated bile acids; and increases circulating interleukin-6 levels and diarrhoea incidence.
Collapse
Affiliation(s)
- Yuyong He
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China
| | - Chunxia Mao
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China
| | - Hong Wen
- Jiangxi Provincial Institute of Veterinary Drugs and Feed Control, Nanchang 330096, China
| | - Zhiyu Chen
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China
| | - Tao Lai
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China
| | - Lingyu Li
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China
| | - Wei Lu
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, China
| | - Huadong Wu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
46
|
CCL2 is a KIT D816V-dependent modulator of the bone marrow microenvironment in systemic mastocytosis. Blood 2016; 129:371-382. [PMID: 27856463 DOI: 10.1182/blood-2016-09-739003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/08/2016] [Indexed: 01/08/2023] Open
Abstract
Systemic mastocytosis (SM) is characterized by abnormal accumulation of neoplastic mast cells harboring the activating KIT mutation D816V in the bone marrow and other internal organs. As found in other myeloproliferative neoplasms, increased production of profibrogenic and angiogenic cytokines and related alterations of the bone marrow microenvironment are commonly found in SM. However, little is known about mechanisms and effector molecules triggering fibrosis and angiogenesis in SM. Here we show that KIT D816V promotes expression of the proangiogenic cytokine CCL2 in neoplastic mast cells. Correspondingly, the KIT-targeting drug midostaurin and RNA interference-mediated knockdown of KIT reduced expression of CCL2. We also found that nuclear factor κB contributes to KIT-dependent upregulation of CCL2 in mast cells. In addition, CCL2 secreted by KIT D816V+ mast cells was found to promote the migration of human endothelial cells in vitro. Furthermore, knockdown of CCL2 in neoplastic mast cells resulted in reduced microvessel density and reduced tumor growth in vivo compared with CCL2-expressing cells. Finally, we measured CCL2 serum concentrations in patients with SM and found that CCL2 levels were significantly increased in mastocytosis patients compared with controls. CCL2 serum levels were higher in patients with advanced SM and were found to correlate with poor survival. In summary, we have identified CCL2 as a novel KIT D816V-dependent key regulator of vascular cell migration and angiogenesis in SM. CCL2 expression correlates with disease severity and prognosis. Whether CCL2 may serve as a therapeutic target in advanced SM remains to be determined in forthcoming studies.
Collapse
|
47
|
Kim DK, Beaven MA, Kulinski JM, Desai A, Bandara G, Bai Y, Prussin C, Schwartz LB, Komarow H, Metcalfe DD, Olivera A. Regulation of Reactive Oxygen Species and the Antioxidant Protein DJ-1 in Mastocytosis. PLoS One 2016; 11:e0162831. [PMID: 27611333 PMCID: PMC5017616 DOI: 10.1371/journal.pone.0162831] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/29/2016] [Indexed: 12/20/2022] Open
Abstract
Neoplastic accumulation of mast cells in systemic mastocytosis (SM) associates with activating mutations in the receptor tyrosine kinase KIT. Constitutive activation of tyrosine kinase oncogenes has been linked to imbalances in oxidant/antioxidant mechanisms in other myeloproliferative disorders. However, the impact of KIT mutations on the redox status in SM and the potential therapeutic implications are not well understood. Here, we examined the regulation of reactive oxygen species (ROS) and of the antioxidant protein DJ-1 (PARK-7), which increases with cancer progression and acts to lessen oxidative damage to malignant cells, in relationship with SM severity. ROS levels were increased in both indolent (ISM) and aggressive variants of the disease (ASM). However, while DJ-1 levels were reduced in ISM with lower mast cell burden, they rose in ISM with higher mast cell burden and were significantly elevated in patients with ASM. Studies on mast cell lines revealed that activating KIT mutations induced constant ROS production and consequent DJ-1 oxidation and degradation that could explain the reduced levels of DJ-1 in the ISM population, while IL-6, a cytokine that increases with disease severity, caused a counteracting transcriptional induction of DJ-1 which would protect malignant mast cells from oxidative damage. A mouse model of mastocytosis recapitulated the biphasic changes in DJ-1 and the escalating IL-6, ROS and DJ-1 levels as mast cells accumulate, findings which were reversed with anti-IL-6 receptor blocking antibody. Our findings provide evidence of increased ROS and a biphasic regulation of the antioxidant DJ-1 in variants of SM and implicate IL-6 in DJ-1 induction and expansion of mast cells with KIT mutations. We propose consideration of IL-6 blockade as a potential adjunctive therapy in the treatment of patients with advanced mastocytosis, as it would reduce DJ-1 levels making mutation-positive mast cells vulnerable to oxidative damage.
Collapse
Affiliation(s)
- Do-Kyun Kim
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael A. Beaven
- Laboratory of Molecular Immunology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joseph M. Kulinski
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Avanti Desai
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Geethani Bandara
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yun Bai
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Calman Prussin
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lawrence B. Schwartz
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Hirsh Komarow
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Dean D. Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ana Olivera
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
48
|
Theoharides TC, Tsilioni I, Patel AB, Doyle R. Atopic diseases and inflammation of the brain in the pathogenesis of autism spectrum disorders. Transl Psychiatry 2016; 6:e844. [PMID: 27351598 PMCID: PMC4931610 DOI: 10.1038/tp.2016.77] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 02/23/2016] [Accepted: 03/17/2016] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorders (ASDs) affect as many as 1 in 45 children and are characterized by deficits in sociability and communication, as well as stereotypic movements. Many children also show severe anxiety. The lack of distinct pathogenesis and reliable biomarkers hampers the development of effective treatments. As a result, most children with ASD are prescribed psychopharmacologic agents that do not address the core symptoms of ASD. Autoantibodies against brain epitopes in mothers of children with ASD and many such children strongly correlate with allergic symptoms and indicate an aberrant immune response, as well as disruption of the blood-brain barrier (BBB). Recent epidemiological studies have shown a strong statistical correlation between risk for ASD and either maternal or infantile atopic diseases, such as asthma, eczema, food allergies and food intolerance, all of which involve activation of mast cells (MCs). These unique tissue immune cells are located perivascularly in all tissues, including the thalamus and hypothalamus, which regulate emotions. MC-derived inflammatory and vasoactive mediators increase BBB permeability. Expression of the inflammatory molecules interleukin (IL-1β), IL-6, 1 L-17 and tumor necrosis factor (TNF) is increased in the brain, cerebrospinal fluid and serum of some patients with ASD, while NF-kB is activated in brain samples and stimulated peripheral blood immune cells of other patients; however, these molecules are not specific. Instead the peptide neurotensin is uniquely elevated in the serum of children with ASD, as is corticotropin-releasing hormone, secreted from the hypothalamus under stress. Both peptides trigger MC to release IL-6 and TNF, which in turn, stimulate microglia proliferation and activation, leading to disruption of neuronal connectivity. MC-derived IL-6 and TGFβ induce maturation of Th17 cells and MCs also secrete IL-17, which is increased in ASD. Serum IL-6 and TNF may define an ASD subgroup that benefits most from treatment with the natural flavonoid luteolin. Atopic diseases may create a phenotype susceptible to ASD and formulations targeting focal inflammation of the brain could have great promise in the treatment of ASD.
Collapse
Affiliation(s)
- T C Theoharides
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
- Sackler School of Graduate Biomedical Sciences, Program in Cell, Molecular and Developmental Biology, Tufts University, Boston, MA, USA
- Department of Internal Medicine, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, USA
- Department of Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, USA
| | - I Tsilioni
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| | - A B Patel
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
- Sackler School of Graduate Biomedical Sciences, Program in Cell, Molecular and Developmental Biology, Tufts University, Boston, MA, USA
| | - R Doyle
- Department of Child Psychiatry, Harvard Medical School, Massachusetts General Hospital and McLean Hospital, Boston, MA, USA
| |
Collapse
|
49
|
Dowse R, Ibrahim M, McLornan DP, Moonim MT, Harrison CN, Radia DH. Beneficial effects of JAK inhibitor therapy in Systemic Mastocytosis. Br J Haematol 2016; 176:324-327. [DOI: 10.1111/bjh.13951] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Robin Dowse
- Department of Haematological Medicine; King's College NHS Foundation Trust; London UK
| | - Maria Ibrahim
- Department of Haematological Medicine; King's College NHS Foundation Trust; London UK
| | - Donal P. McLornan
- Department of Haematological Medicine; King's College NHS Foundation Trust; London UK
- Department of Haematology; Guy's and St. Thomas' NHS Foundation Trust; London UK
| | - Mufaddal T. Moonim
- Department of Pathology; Guy's and St. Thomas' NHS Foundation Trust; London UK
| | - Claire N. Harrison
- Department of Haematology; Guy's and St. Thomas' NHS Foundation Trust; London UK
| | - Deepti H. Radia
- Department of Haematology; Guy's and St. Thomas' NHS Foundation Trust; London UK
| |
Collapse
|
50
|
IL-6 promotes an increase in human mast cell numbers and reactivity through suppression of suppressor of cytokine signaling 3. J Allergy Clin Immunol 2016; 137:1863-1871.e6. [PMID: 26774658 DOI: 10.1016/j.jaci.2015.09.059] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/16/2015] [Accepted: 09/30/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND IL-6, levels of which are reported to be increased in association with mastocytosis, asthma, and urticaria, is used in conjunction with stem cell factor to generate CD34(+) cell-derived primary human mast cell (HuMC) cultures. Despite these associations, the effects on and mechanisms by which prolonged exposure to IL-6 alters HuMC numbers and function are not well understood. OBJECTIVES We sought to study the effect of IL-6 on HuMC function, the mechanisms by which IL-6 exerts its effects, and the relationship of these findings to mastocytosis. METHODS HuMCs were cultured in stem cell factor with or without IL-6. Responses to FcεRI aggregation and expression of proteases and receptors, including the soluble IL-6 receptor (sIL-6R), were then quantitated. Epigenetic changes in suppressor of cytokine signaling 3 (SOCS3) were determined by using methylation-specific PCR. Serum samples from healthy control subjects and patients with mastocytosis were assayed for IL-6, tryptase, and sIL-6R. RESULTS IL-6 enhanced mast cell (MC) proliferation, maturation, and reactivity after FcεRI aggregation. IL-6 reduced expression of SOCS3, which correlated with methylation of the SOCS3 promoter and increased expression and activation of signal transducer and activator of transcription 3. IL-6 also suppressed constitutive production of sIL-6R, and serum levels of sIL-6R were similarly reduced in patients with mastocytosis. CONCLUSION IL-6 increases MC proliferation and formation of a more reactive phenotype enabled by suppressing proteolytic cleavage of sIL-6R from IL-6R and downregulation of the SOCS3 autoinhibitory pathway. We suggest IL-6 blockade might ameliorate MC-related symptoms and pathology in patients with MC-related diseases associated with increased IL-6 levels, including mastocytosis.
Collapse
|