1
|
Li R, Colombo M, Wang G, Rodriguez-Romera A, Benlabiod C, Jooss NJ, O’Sullivan J, Brierley CK, Clark SA, Pérez Sáez JM, Aragón Fernández P, Schoof EM, Porse B, Meng Y, Khan AO, Wen S, Dong P, Zhou W, Sousos N, Murphy L, Clarke M, Olijnik AA, C. Wong Z, Karali CS, Sirinukunwattana K, Ryou H, Norfo R, Cheng Q, Carrelha J, Ren Z, Thongjuea S, Rathinam VA, Krishnan A, Royston D, Rabinovich GA, Mead AJ, Psaila B. A proinflammatory stem cell niche drives myelofibrosis through a targetable galectin-1 axis. Sci Transl Med 2024; 16:eadj7552. [PMID: 39383242 PMCID: PMC7616771 DOI: 10.1126/scitranslmed.adj7552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/01/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024]
Abstract
Myeloproliferative neoplasms are stem cell-driven cancers associated with a large burden of morbidity and mortality. Most patients present with early-stage disease, but a substantial proportion progress to myelofibrosis or secondary leukemia, advanced cancers with a poor prognosis and high symptom burden. Currently, it remains difficult to predict progression, and therapies that reliably prevent or reverse fibrosis are lacking. A major bottleneck to the discovery of disease-modifying therapies has been an incomplete understanding of the interplay between perturbed cellular and molecular states. Several cell types have individually been implicated, but a comprehensive analysis of myelofibrotic bone marrow is lacking. We therefore mapped the cross-talk between bone marrow cell types in myelofibrotic bone marrow. We found that inflammation and fibrosis are orchestrated by a "quartet" of immune and stromal cell lineages, with basophils and mast cells creating a TNF signaling hub, communicating with megakaryocytes, mesenchymal stromal cells, and proinflammatory fibroblasts. We identified the β-galactoside-binding protein galectin-1 as a biomarker of progression to myelofibrosis and poor survival in multiple patient cohorts and as a promising therapeutic target, with reduced myeloproliferation and fibrosis in vitro and in vivo and improved survival after galectin-1 inhibition. In human bone marrow organoids, TNF increased galectin-1 expression, suggesting a feedback loop wherein the proinflammatory myeloproliferative neoplasm clone creates a self-reinforcing niche, fueling progression to advanced disease. This study provides a resource for studying hematopoietic cell-niche interactions, with relevance for cancer-associated inflammation and disorders of tissue fibrosis.
Collapse
Affiliation(s)
- Rong Li
- CAMS Oxford Institute; University of Oxford; Oxford, United Kingdom (UK)
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Michela Colombo
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Human Technopole; Milan, Italy
| | - Guanlin Wang
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- MRC WIMM Centre for Computational Biology, University of Oxford; Oxford, United Kingdom
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
- Qizhi Institute, Shanghai, China
| | - Antonio Rodriguez-Romera
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Camelia Benlabiod
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Natalie J. Jooss
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Jennifer O’Sullivan
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Charlotte K. Brierley
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Sally-Ann Clark
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Juan M. Pérez Sáez
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | | | - Erwin M. Schoof
- Department of Biotechnology and Biomedicine, Technical University of Denmark; Denmark
| | - Bo Porse
- The Finsen Laboratory, Copenhagen University Hospital; Copenhagen, Denmark
- Biotech Research and Innovation Centre, Faculty of Health Sciences, University of Copenhagen; Denmark
- Department of Clinical Medicine, University of Copenhagen; Copenhagen, Denmark
| | - Yiran Meng
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Abdullah O. Khan
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences; University of Birmingham; Birmingham, UK
| | - Sean Wen
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Pengwei Dong
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
| | - Wenjiang Zhou
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
| | - Nikolaos Sousos
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Lauren Murphy
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Matthew Clarke
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Aude-Anais Olijnik
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Zoë C. Wong
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Christina Simoglou Karali
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Korsuk Sirinukunwattana
- Oxford Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford; Oxford, UK
| | - Hosuk Ryou
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford; Oxford, UK
| | - Ruggiero Norfo
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Qian Cheng
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Joana Carrelha
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford; Oxford, UK
| | - Zemin Ren
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Supat Thongjuea
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Vijay A Rathinam
- Department of Immunology, University of Connecticut Health School of Medicine; Farmington, ConnecticutUSA
| | - Anandi Krishnan
- Stanford Cancer Institute, Stanford University School of Medicine; Stanford, California, USA
| | - Daniel Royston
- Biotech Research and Innovation Centre, Faculty of Health Sciences, University of Copenhagen; Denmark
- Oxford University Hospitals NHS Trust; Oxford, UK
| | - Gabriel A. Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adam J Mead
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Oxford University Hospitals NHS Trust; Oxford, UK
| | - Bethan Psaila
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Oxford University Hospitals NHS Trust; Oxford, UK
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| |
Collapse
|
2
|
Chen F, Tang H, Cai X, Lin J, Kang R, Tang D, Liu J. DAMPs in immunosenescence and cancer. Semin Cancer Biol 2024; 106-107:123-142. [PMID: 39349230 DOI: 10.1016/j.semcancer.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/02/2024]
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous molecules released by cells in response to injury or stress, recognized by host pattern recognition receptors that assess the immunological significance of cellular damage. The interaction between DAMPs and innate immune receptors triggers sterile inflammation, which serves a dual purpose: promoting tissue repair and contributing to pathological conditions, including age-related diseases. Chronic inflammation mediated by DAMPs accelerates immunosenescence and influences both tumor progression and anti-tumor immunity, underscoring the critical role of DAMPs in the nexus between aging and cancer. This review explores the characteristics of immunosenescence and its impact on age-related cancers, investigates the various types of DAMPs, their release mechanisms during cell death, and the immune activation pathways they initiate. Additionally, we examine the therapeutic potential of targeting DAMPs in age-related diseases. A detailed understanding of DAMP-induced signal transduction could provide critical insights into immune regulation and support the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Fangquan Chen
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Hu Tang
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Xiutao Cai
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Junhao Lin
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China.
| |
Collapse
|
3
|
De Luca G, Goette NP, Lev PR, Baroni Pietto MC, Marin Oyarzún CP, Castro Ríos MA, Moiraghi B, Sackmann F, Kamiya LJ, Verri V, Caula V, Fernandez V, Vicente A, Pose Cabarcos J, Caruso V, Camacho MF, Larripa IB, Khoury M, Marta RF, Glembotsky AC, Heller PG. Elevated levels of damage-associated molecular patterns HMGB1 and S100A8/A9 coupled with toll-like receptor-triggered monocyte activation are associated with inflammation in patients with myelofibrosis. Front Immunol 2024; 15:1365015. [PMID: 39391311 PMCID: PMC11465240 DOI: 10.3389/fimmu.2024.1365015] [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: 01/03/2024] [Accepted: 08/26/2024] [Indexed: 10/12/2024] Open
Abstract
Inflammation plays a pivotal role in the pathogenesis of primary and post-essential thrombocythemia or post-polycythemia vera myelofibrosis (MF) in close cooperation with the underlying molecular drivers. This inflammatory state is induced by a dynamic spectrum of inflammatory cytokines, although recent evidence points to the participation of additional soluble inflammatory mediators. Damage-associated molecular patterns (DAMPs) represent endogenous signals released upon cell death or damage which trigger a potent innate immune response. We assessed the contribution of two prototypical DAMPs, HMGB1 and S100A8/A9, to MF inflammation. Circulating HMGB1 and S100A8/A9 were elevated in MF patients in parallel to the degree of systemic inflammation and levels increased progressively during advanced disease stages. Patients with elevated DAMPs had higher frequency of adverse clinical features, such as anemia, and inferior survival, suggesting their contribution to disease progression. Monocytes, which are key players in MF inflammation, were identified as a source of S100A8/A9 but not HMGB1 release, while both DAMPs correlated with cell death parameters, such as serum LDH and cell-free DNA, indicating that passive release is an additional mechanism leading to increased DAMPs. HMGB1 and S100A8/A9 promote inflammation through binding to Toll-like receptor (TLR) 4, whereas the former also binds TLR2. Monocytes from MF patients were shown to be hyperactivated at baseline, as reflected by higher CD11b and tissue factor exposure and increased expression levels of proinflammatory cytokines IL-1β and IL-6. Patient monocytes showed preserved TLR4 and TLR2 expression and were able to mount normal or even exacerbated functional responses and cytokine upregulation following stimulation of TLR4 and TLR2. Elevated levels of endogenous TLR ligands HMGB1 and S100A8/A9 coupled to the finding of preserved or hyperreactive TLR-triggered responses indicate that DAMPs may promote monocyte activation and cytokine production in MF, fueling inflammation. Plasma IL-1β and IL-6 were elevated in MF and correlated with DAMPs levels, raising the possibility that DAMPs could contribute to cytokine generation in vivo. In conclusion, this study highlights that, in cooperation with classic proinflammatory cytokines, DAMPs represent additional inflammatory mediators that may participate in the generation of MF inflammatory state, potentially providing novel biomarkers of disease progression and new therapeutic targets.
Collapse
Affiliation(s)
- Geraldine De Luca
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Nora P. Goette
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Paola R. Lev
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Maria C. Baroni Pietto
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cecilia P. Marin Oyarzún
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | | | | | | - Laureano J. Kamiya
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Veronica Verri
- División Hematología Clínica, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Victoria Caula
- División Hematología Clínica, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Vanina Fernandez
- Departamento de Hematología, Hospital Posadas, Buenos Aires, Argentina
| | - Angeles Vicente
- Departamento de Hematología, Hospital Alemán, Buenos Aires, Argentina
| | - Julio Pose Cabarcos
- Departamento de Hematología, Sanatorio Otamendi Miroli, Buenos Aires, Argentina
| | - Vanesa Caruso
- Departamento de Hematología, Hospital Piñero, Buenos Aires, Argentina
| | - Maria F. Camacho
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Irene B. Larripa
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Marina Khoury
- Departamento de Docencia e Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rosana F. Marta
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ana C. Glembotsky
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paula G. Heller
- División Hematología Investigación, Instituto de Investigaciones Médicas Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
4
|
Capitanio D, Calledda FR, Abbonante V, Cattaneo D, Moriggi M, Niccolò B, Bucelli C, Tosi D, Gianelli U, Vannucchi AM, Iurlo A, Gelfi C, Balduini A, Malara A. Proteomic screening identifies PF4/Cxcl4 as a critical driver of myelofibrosis. Leukemia 2024; 38:1971-1984. [PMID: 39025985 DOI: 10.1038/s41375-024-02354-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Despite increased understanding of the genomic landscape of Myeloproliferative Neoplasms (MPNs), the pathological mechanisms underlying abnormal megakaryocyte (Mk)-stromal crosstalk and fibrotic progression in MPNs remain unclear. We conducted mass spectrometry-based proteomics on mice with Romiplostim-dependent myelofibrosis to reveal alterations in signaling pathways and protein changes in Mks, platelets, and bone marrow (BM) cells. The chemokine Platelet Factor 4 (PF4)/Cxcl4 was up-regulated in all proteomes and increased in plasma and BM fluids of fibrotic mice. High TPO concentrations sustained in vitro PF4 synthesis and secretion in cultured Mks, while Ruxolitinib restrains the abnormal PF4 expression in vivo. We discovered that PF4 is rapidly internalized by stromal cells through surface glycosaminoglycans (GAGs) to promote myofibroblast differentiation. Cxcl4 gene silencing in Mks mitigated the profibrotic phenotype of stromal cells in TPO-saturated co-culture conditions. Consistently, extensive stromal PF4 uptake and altered GAGs deposition were detected in Romiplostim-treated, JAK2V617F mice and BM biopsies of MPN patients. BM PF4 levels and Mk/platelet CXCL4 expression were elevated in patients, exclusively in overt fibrosis. Finally, pharmacological inhibition of GAGs ameliorated in vivo fibrosis in Romiplostim-treated mice. Thus, our findings highlight the critical role of PF4 in the fibrosis progression of MPNs and substantiate the potential therapeutic strategy of neutralizing PF4-GAGs interaction.
Collapse
Affiliation(s)
- Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | | | - Vittorio Abbonante
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Bartalucci Niccolò
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Azienda Ospedaliera-Universitaria Careggi, Florence, Italy
| | - Cristina Bucelli
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Delfina Tosi
- Department of Health Sciences, University of Milan, S.C. di Anatomia Patologica, ASST-Santi Paolo e Carlo, Milan, Italy
| | - Umberto Gianelli
- Department of Health Sciences, University of Milan, S.C. di Anatomia Patologica, ASST-Santi Paolo e Carlo, Milan, Italy
| | - Alessandro Maria Vannucchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Azienda Ospedaliera-Universitaria Careggi, Florence, Italy
| | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | | | - Alessandro Malara
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
| |
Collapse
|
5
|
Mahmoodi M, Mirzarazi Dahagi E, Nabavi M, Penalva YCM, Gosaine A, Murshed M, Couldwell S, Munter LM, Kaartinen MT. Circulating plasma fibronectin affects tissue insulin sensitivity, adipocyte differentiation, and transcriptional landscape of adipose tissue in mice. Physiol Rep 2024; 12:e16152. [PMID: 39054559 PMCID: PMC11272447 DOI: 10.14814/phy2.16152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
Plasma fibronectin (pFN) is a hepatocyte-derived circulating extracellular matrix protein that affects cell morphology, adipogenesis, and insulin signaling of adipocytes in vitro. In this study, we show pFN accrual to adipose tissue and its contribution to tissue homeostasis in mice. Hepatocyte-specific conditional Fn1 knockout mice (Fn1-/-ALB) show a decrease in adipose tissue FN levels and enhanced insulin sensitivity of subcutaneous (inguinal), visceral (epididymal) adipose tissue on a normal diet. Diet-induced obesity model of the Fn1-/-ALB mouse showed normal weight gain and whole-body fat mass, and normal adipose tissue depot volumes and unaltered circulating leptin and adiponectin levels. However, Fn1-/-ALB adipose depots showed significant alterations in adipocyte size and gene expression profiles. The inguinal adipose tissue on a normal diet, which had alterations in fatty acid metabolism and thermogenesis suggesting browning. The presence of increased beige adipocyte markers Ucp1 and Prdm16 supported this. In the inguinal fat, the obesogenic diet resulted in downregulation of the browning markers and changes in gene expression reflecting development, morphogenesis, and mesenchymal stem cell maintenance. Epididymal adipose tissue showed alterations in developmental and stem cell gene expression on both diets. The data suggests a role for pFN in adipose tissue insulin sensitivity and cell profiles.
Collapse
Affiliation(s)
- Mahdokht Mahmoodi
- Faculty of Dental Medicine and Oral Health Sciences (Biomedical Sciences)McGill UniversityMontrealQuebecCanada
| | - Elahe Mirzarazi Dahagi
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Mir‐Hamed Nabavi
- Faculty of Dental Medicine and Oral Health Sciences (Biomedical Sciences)McGill UniversityMontrealQuebecCanada
| | - Ylauna C. M. Penalva
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Centre de Recherche en Biologie Structurale (CRBS)McGill UniversityMontrealQuebecCanada
| | - Amrita Gosaine
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Monzur Murshed
- Faculty of Dental Medicine and Oral Health Sciences (Biomedical Sciences)McGill UniversityMontrealQuebecCanada
- Shriners Hospital for ChildrenMontrealQuebecCanada
| | - Sandrine Couldwell
- Faculty of Dental Medicine and Oral Health Sciences (Biomedical Sciences)McGill UniversityMontrealQuebecCanada
| | - Lisa M. Munter
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Centre de Recherche en Biologie Structurale (CRBS)McGill UniversityMontrealQuebecCanada
| | - Mari T. Kaartinen
- Faculty of Dental Medicine and Oral Health Sciences (Biomedical Sciences)McGill UniversityMontrealQuebecCanada
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Department of Medicine (Division of Experimental Medicine), Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
6
|
Liu Q, Han M, Wu Z, Fu W, Ji J, Liang Q, Tan M, Zhai L, Gao J, Shi D, Jiang Q, Sun Z, Lai Y, Xu Q, Sun Y. DDX5 inhibits hyaline cartilage fibrosis and degradation in osteoarthritis via alternative splicing and G-quadruplex unwinding. NATURE AGING 2024; 4:664-680. [PMID: 38760576 PMCID: PMC11108786 DOI: 10.1038/s43587-024-00624-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 04/04/2024] [Indexed: 05/19/2024]
Abstract
Hyaline cartilage fibrosis is typically considered an end-stage pathology of osteoarthritis (OA), which results in changes to the extracellular matrix. However, the mechanism behind this is largely unclear. Here, we found that the RNA helicase DDX5 was dramatically downregulated during the progression of OA. DDX5 deficiency increased fibrosis phenotype by upregulating COL1 expression and downregulating COL2 expression. In addition, loss of DDX5 aggravated cartilage degradation by inducing the production of cartilage-degrading enzymes. Chondrocyte-specific deletion of Ddx5 led to more severe cartilage lesions in the mouse OA model. Mechanistically, weakened DDX5 resulted in abundance of the Fn1-AS-WT and Plod2-AS-WT transcripts, which promoted expression of fibrosis-related genes (Col1, Acta2) and extracellular matrix degradation genes (Mmp13, Nos2 and so on), respectively. Additionally, loss of DDX5 prevented the unfolding Col2 promoter G-quadruplex, thereby reducing COL2 production. Together, our data suggest that strategies aimed at the upregulation of DDX5 hold significant potential for the treatment of cartilage fibrosis and degradation in OA.
Collapse
Affiliation(s)
- Qianqian Liu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Mingrui Han
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zhigui Wu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Wenqiang Fu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, China
| | - Jun Ji
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qingqing Liang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Linhui Zhai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jian Gao
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Dongquan Shi
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ziying Sun
- Department of Orthopaedics, Jinling Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yuping Lai
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
7
|
Ma M, Jiang W, Zhou R. DAMPs and DAMP-sensing receptors in inflammation and diseases. Immunity 2024; 57:752-771. [PMID: 38599169 DOI: 10.1016/j.immuni.2024.03.002] [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: 12/18/2023] [Revised: 02/17/2024] [Accepted: 03/01/2024] [Indexed: 04/12/2024]
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous danger molecules produced in cellular damage or stress, and they can activate the innate immune system. DAMPs contain multiple types of molecules, including nucleic acids, proteins, ions, glycans, and metabolites. Although these endogenous molecules do not trigger immune response under steady-state condition, they may undergo changes in distribution, physical or chemical property, or concentration upon cellular damage or stress, and then they become DAMPs that can be sensed by innate immune receptors to induce inflammatory response. Thus, DAMPs play an important role in inflammation and inflammatory diseases. In this review, we summarize the conversion of homeostatic molecules into DAMPs; the diverse nature and classification, cellular origin, and sensing of DAMPs; and their role in inflammation and related diseases. Furthermore, we discuss the clinical strategies to treat DAMP-associated diseases via targeting DAMP-sensing receptors.
Collapse
Affiliation(s)
- Ming Ma
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Wei Jiang
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Rongbin Zhou
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China; Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
8
|
Duparc H, Muller D, Gilles L, Chédeville AL, El Khoury M, Guignard R, Debili N, Wittner M, Kauskot A, Pasquier F, Antony-Debré I, Marty C, Vainchenker W, Plo I, Raslova H. Deregulation of the p19/CDK4/CDK6 axis in Jak2 V617F megakaryocytes accelerates the development of myelofibrosis. Leukemia 2024; 38:898-902. [PMID: 38378843 DOI: 10.1038/s41375-024-02170-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/22/2024]
Affiliation(s)
- Hélène Duparc
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris Cité, Paris, France
| | - Delphine Muller
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Laure Gilles
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Agathe L Chédeville
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris Cité, Paris, France
| | - Mira El Khoury
- INSERM UMRS 938, PARIS, Sorbonne Université, Centre de Recherche Saint- Antoine, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Rose Guignard
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris Cité, Paris, France
| | - Najet Debili
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Monika Wittner
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Alexandre Kauskot
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Florence Pasquier
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Département d'Hématologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Iléana Antony-Debré
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Caroline Marty
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - William Vainchenker
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Hana Raslova
- INSERM, UMR1287, Gustave Roussy, Equipe labellisée Ligue Nationale Contre le Cancer, Villejuif, France.
- Université Paris-Saclay, Gif-sur-Yvette, Villejuif, France.
- Gustave Roussy, Villejuif, France.
| |
Collapse
|
9
|
de Jong MME, Chen L, Raaijmakers MHGP, Cupedo T. Bone marrow inflammation in haematological malignancies. Nat Rev Immunol 2024:10.1038/s41577-024-01003-x. [PMID: 38491073 DOI: 10.1038/s41577-024-01003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2024] [Indexed: 03/18/2024]
Abstract
Tissue inflammation is a hallmark of tumour microenvironments. In the bone marrow, tumour-associated inflammation impacts normal niches for haematopoietic progenitor cells and mature immune cells and supports the outgrowth and survival of malignant cells residing in these niche compartments. This Review provides an overview of our current understanding of inflammatory changes in the bone marrow microenvironment of myeloid and lymphoid malignancies, using acute myeloid leukaemia and multiple myeloma as examples and highlights unique and shared features of inflammation in niches for progenitor cells and plasma cells. Importantly, inflammation exerts profoundly different effects on normal bone marrow niches in these malignancies, and we provide context for possible drivers of these divergent effects. We explore the role of tumour cells in inflammatory changes, as well as the role of cellular constituents of normal bone marrow niches, including myeloid cells and stromal cells. Integrating knowledge of disease-specific dynamics of malignancy-associated bone marrow inflammation will provide a necessary framework for future targeting of these processes to improve patient outcome.
Collapse
Affiliation(s)
- Madelon M E de Jong
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Lanpeng Chen
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Tom Cupedo
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| |
Collapse
|
10
|
Abbonante V, Karkempetzaki AI, Leon C, Krishnan A, Huang N, Di Buduo CA, Cattaneo D, Ward CMT, Matsuura S, Guinard I, Weber J, De Acutis A, Vozzi G, Iurlo A, Ravid K, Balduini A. Newly identified roles for PIEZO1 mechanosensor in controlling normal megakaryocyte development and in primary myelofibrosis. Am J Hematol 2024; 99:336-349. [PMID: 38165047 PMCID: PMC10922533 DOI: 10.1002/ajh.27184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024]
Abstract
Mechanisms through which mature megakaryocytes (Mks) and their progenitors sense the bone marrow extracellular matrix to promote lineage differentiation in health and disease are still partially understood. We found PIEZO1, a mechanosensitive cation channel, to be expressed in mouse and human Mks. Human mutations in PIEZO1 have been described to be associated with blood cell disorders. Yet, a role for PIEZO1 in megakaryopoiesis and proplatelet formation has never been investigated. Here, we show that activation of PIEZO1 increases the number of immature Mks in mice, while the number of mature Mks and Mk ploidy level are reduced. Piezo1/2 knockout mice show an increase in Mk size and platelet count, both at basal state and upon marrow regeneration. Similarly, in human samples, PIEZO1 is expressed during megakaryopoiesis. Its activation reduces Mk size, ploidy, maturation, and proplatelet extension. Resulting effects of PIEZO1 activation on Mks resemble the profile in Primary Myelofibrosis (PMF). Intriguingly, Mks derived from Jak2V617F PMF mice show significantly elevated PIEZO1 expression, compared to wild-type controls. Accordingly, Mks isolated from bone marrow aspirates of JAK2V617F PMF patients show increased PIEZO1 expression compared to Essential Thrombocythemia. Most importantly, PIEZO1 expression in bone marrow Mks is inversely correlated with patient platelet count. The ploidy, maturation, and proplatelet formation of Mks from JAK2V617F PMF patients are rescued upon PIEZO1 inhibition. Together, our data suggest that PIEZO1 places a brake on Mk maturation and platelet formation in physiology, and its upregulation in PMF Mks might contribute to aggravating some hallmarks of the disease.
Collapse
Affiliation(s)
- Vittorio Abbonante
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Anastasia Iris Karkempetzaki
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- University of Crete, School of Medicine, Heraklion, Greece
| | - Catherine Leon
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Anandi Krishnan
- Institute of Immunology, Stanford University School of Medicine, Palo Alto, California, United States
| | - Nasi Huang
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | | | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Christina Marie Torres Ward
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Shinobu Matsuura
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ines Guinard
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Josiane Weber
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Aurora De Acutis
- Interdepartmental Research Center "E. Piaggio", University of Pisa, Pisa, Italy
| | - Giovanni Vozzi
- Interdepartmental Research Center "E. Piaggio", University of Pisa, Pisa, Italy
| | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| |
Collapse
|
11
|
De Luca G, Lev PR, Camacho MF, Goette NP, Sackmann F, Castro Ríos MA, Moiraghi B, Cortes Guerrieri V, Bendek G, Carricondo E, Enrico A, Vallejo V, Varela A, Khoury M, Gutierrez M, Larripa IB, Marta RF, Glembotsky AC, Heller PG. High cell-free DNA is associated with disease progression, inflammasome activation and elevated levels of inflammasome-related cytokine IL-18 in patients with myelofibrosis. Front Immunol 2023; 14:1161832. [PMID: 38035089 PMCID: PMC10687201 DOI: 10.3389/fimmu.2023.1161832] [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: 02/08/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
Myelofibrosis (MF) is a clonal hematopoietic stem cell disorder classified among chronic myeloproliferative neoplasms, characterized by exacerbated myeloid and megakaryocytic proliferation and bone marrow fibrosis. It is induced by driver (JAK2/CALR/MPL) and high molecular risk mutations coupled to a sustained inflammatory state that contributes to disease pathogenesis. Patient outcome is determined by stratification into risk groups and refinement of current prognostic systems may help individualize treatment decisions. Circulating cell-free (cf)DNA comprises short fragments of double-stranded DNA, which promotes inflammation by stimulating several pathways, including inflammasome activation, which is responsible for IL-1β and IL-18 maturation and release. In this work, we assessed the contribution of cfDNA as a marker of disease progression and mediator of inflammation in MF. cfDNA was increased in MF patients and higher levels were associated with adverse clinical outcome, a high-risk molecular profile, advanced disease stages and inferior overall survival, indicating its potential value as a prognostic marker. Cell-free DNA levels correlated with tumor burden parameters and markers of systemic inflammation. To mimic the effects of cfDNA, monocytes were stimulated with poly(dA:dT), a synthetic double-stranded DNA. Following stimulation, patient monocytes released higher amounts of inflammasome-processed cytokine, IL-18 to the culture supernatant, reflecting enhanced inflammasome function. Despite overexpression of cytosolic DNA inflammasome sensor AIM2, IL-18 release from MF monocytes was shown to rely mainly on the NLRP3 inflammasome, as it was prevented by NLRP3-specific inhibitor MCC950. Circulating IL-18 levels were increased in MF plasma, reflecting in vivo inflammasome activation, and highlighting the previously unrecognized involvement of this cytokine in MF cytokine network. Monocyte counts were higher in patients and showed a trend towards correlation with IL-18 levels, suggesting monocytes represent a source of circulating IL-18. The close correlation shown between IL-18 and cfDNA levels, together with the finding of enhanced DNA-triggered IL-18 release from monocytes, suggest that cfDNA promotes inflammation, at least in part, through inflammasome activation. This work highlights cfDNA, the inflammasome and IL-18 as additional players in the complex inflammatory circuit that fosters MF progression, potentially providing new therapeutic targets.
Collapse
Affiliation(s)
- Geraldine De Luca
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paola R. Lev
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Maria F. Camacho
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Nora P. Goette
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | | | | | - Beatriz Moiraghi
- Departamento de Hematología, Hospital Ramos Mejía, Buenos Aires, Argentina
| | - Veronica Cortes Guerrieri
- División Hematología Clínica, IDIM Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Georgina Bendek
- Departamento de Hematología, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Emiliano Carricondo
- Departamento de Hematología, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Alicia Enrico
- Departamento de Hematología, Hospital Italiano de La Plata, Buenos Aires, Argentina
| | - Veronica Vallejo
- Departamento de Hematología, Instituto Cardiovascular de Buenos Aires, Buenos Aires, Argentina
| | - Ana Varela
- Departamento de Hematología, Hospital Ramos Mejía, Buenos Aires, Argentina
| | - Marina Khoury
- Departamento de Docencia e Investigación, IDIM Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marina Gutierrez
- Unidad Genómica, Laboratorio Stamboulian, Buenos Aires, Argentina
| | - Irene B. Larripa
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, IMEX-CONICET/Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Rosana F. Marta
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ana C. Glembotsky
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paula G. Heller
- División Hematología Investigación, Instituto de Investigaciones Médicas Dr. Alfredo Lanari, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Investigaciones Médicas (IDIM), UBA-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
12
|
Calledda FR, Malara A, Balduini A. Inflammation and bone marrow fibrosis: novel immunotherapeutic targets. Curr Opin Hematol 2023; 30:237-244. [PMID: 37548363 DOI: 10.1097/moh.0000000000000778] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
PURPOSE OF REVIEW Myelofibrosis (MF) is primarily driven by constitutive activation of the Janus kinase/signal transducer of activators of transcription (JAK/STAT) pathway. While JAK inhibitors have shown to alleviate disease symptoms, their disease-modifying effects in MF are limited. The only curative treatment remains allogeneic stem cell transplantation, which can be applied to a minority of patients. As a result, there is a need to explore novel targets in MF to facilitate appropriate drug development and therapeutic pathways. RECENT FINDINGS Recent research has focused on identifying novel signals that contribute to the abnormal cross-talk between hematopoietic and stromal cells, which promotes MF and disease progression. Inflammation and immune dysregulation have emerged as key drivers of both the initiation and progression of MF. A growing number of actionable targets has been identified, including cytokines, transcription factors, signalling networks and cell surface-associated molecules. These targets exhibit dysfunctions in malignant and nonmalignant hematopoietic cells, but also in nonhematopoietic cells of the bone marrow. The study of these inflammation-related molecules, in preclinical models and MF patient's samples, is providing novel therapeutic targets. SUMMARY The identification of immunotherapeutic targets is expanding the therapeutic landscape of MF. This review provides a summary of the most recent advancements in the study of immunotherapeutic targets in MF.
Collapse
|
13
|
Bale S, Verma P, Varga J, Bhattacharyya S. Extracellular Matrix-Derived Damage-Associated Molecular Patterns (DAMP): Implications in Systemic Sclerosis and Fibrosis. J Invest Dermatol 2023; 143:1877-1885. [PMID: 37452808 DOI: 10.1016/j.jid.2023.04.030] [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: 01/19/2023] [Revised: 03/24/2023] [Accepted: 04/07/2023] [Indexed: 07/18/2023]
Abstract
Damage-associated molecular patterns (DAMPs) are intracellular molecules released under cellular stress or recurring tissue injury, which serve as endogenous ligands for toll-like receptors (TLRs). Such DAMPs are either actively secreted by immune cells or passively released into the extracellular environment from damaged cells or generated as alternatively spliced mRNA variants of extracellular matrix (ECM) glycoproteins. When recognized by pattern recognition receptors (PRRs) such as TLRs, DAMPs trigger innate immune responses. Currently, the best-characterized PRRs include, in addition to TLRs, nucleotide-binding oligomerization domain-like receptors, RIG-I-like RNA helicases, C-type lectin receptors, and many more. Systemic sclerosis (SSc) is a chronic autoimmune condition characterized by inflammation and progressive fibrosis in multiple organs. Using an unbiased survey for SSc-associated DAMPs, we have identified the ECM glycoproteins fibronectin-containing extra domain A and tenascin C as the most highly upregulated in SSc skin and lung biopsies. These DAMPs activate TLR4 on resident stromal cells to elicit profibrotic responses and sustained myofibroblasts activation resulting in progressive fibrosis. This review summarizes the current understanding of the complex functional roles of DAMPs in the progression and failure of resolution of fibrosis in general, with a particular focus on SSc, and considers viable therapeutic approaches targeting DAMPs.
Collapse
Affiliation(s)
- Swarna Bale
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Priyanka Verma
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - John Varga
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Swati Bhattacharyya
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
14
|
Vainchenker W, Yahmi N, Havelange V, Marty C, Plo I, Constantinescu SN. Recent advances in therapies for primary myelofibrosis. Fac Rev 2023; 12:23. [PMID: 37771602 PMCID: PMC10523375 DOI: 10.12703/r/12-23] [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] [Indexed: 09/30/2023] Open
Abstract
Primary myelofibrosis (PMF), polycythemia vera (PV) and essential thrombocythemia (ET) form the classical BCR-ABL1-negative myeloproliferative neoplasms (MPNs) that are driven by a constitutive activation of JAK2 signaling. PMF as well as secondary MF (post-ET and post-PV MF) are the most aggressive MPNs. Presently, there is no curative treatment, except allogenic hematopoietic stem cell transplantation. JAK inhibitors, essentially ruxolitinib, are the therapy of reference for intermediate and high-risk MF. However, presently the current JAK inhibitors behave mainly as anti-inflammatory drugs, improving general symptoms and spleen size without major impact on disease progression. A better understanding of the genetics of MF, the biology of its leukemic stem cells (LSCs), the mechanisms of fibrosis and of cytopenia and the role of inflammatory cytokines has led to new approaches with the development of numerous therapeutic agents that target epigenetic regulation, telomerase, apoptosis, cell cycle, cytokines and signaling. Furthermore, the use of a new less toxic form of interferon-α has been revived, as it is presently one of the only molecules that targets the mutated clone. These new approaches have different aims: (a) to provide alternative therapy to JAK inhibition; (b) to correct cytopenia; and (c) to inhibit fibrosis development. However, the main important goal is to find new disease modifier treatments, which will profoundly modify the progression of the disease without major toxicity. Presently the most promising approaches consist of the inhibition of telomerase and the combination of JAK2 inhibitors (ruxolitinib) with either a BCL2/BCL-xL or BET inhibitor. Yet, the most straightforward future approaches can be considered to be the development of and/or selective inhibition of JAK2V617F and the targeting MPL and calreticulin mutants by immunotherapy. It can be expected that the therapy of MF will be significantly improved in the coming years.
Collapse
Affiliation(s)
- William Vainchenker
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Nasrine Yahmi
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Violaine Havelange
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Cliniques universitaires Saint Luc, Department of Hematology, Université Catholique de Louvain, Brussels, Belgium
| | - Caroline Marty
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Stefan N Constantinescu
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, Brussels, Belgium
- WEL Research Institute, WELBIO Department, Wavre, Belgium
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| |
Collapse
|
15
|
Hasselbalch HC, Junker P, Skov V, Kjær L, Knudsen TA, Larsen MK, Holmström MO, Andersen MH, Jensen C, Karsdal MA, Willumsen N. Revisiting Circulating Extracellular Matrix Fragments as Disease Markers in Myelofibrosis and Related Neoplasms. Cancers (Basel) 2023; 15:4323. [PMID: 37686599 PMCID: PMC10486581 DOI: 10.3390/cancers15174323] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 09/10/2023] Open
Abstract
Philadelphia chromosome-negative chronic myeloproliferative neoplasms (MPNs) arise due to acquired somatic driver mutations in stem cells and develop over 10-30 years from the earliest cancer stages (essential thrombocythemia, polycythemia vera) towards the advanced myelofibrosis stage with bone marrow failure. The JAK2V617F mutation is the most prevalent driver mutation. Chronic inflammation is considered to be a major pathogenetic player, both as a trigger of MPN development and as a driver of disease progression. Chronic inflammation in MPNs is characterized by persistent connective tissue remodeling, which leads to organ dysfunction and ultimately, organ failure, due to excessive accumulation of extracellular matrix (ECM). Considering that MPNs are acquired clonal stem cell diseases developing in an inflammatory microenvironment in which the hematopoietic cell populations are progressively replaced by stromal proliferation-"a wound that never heals"-we herein aim to provide a comprehensive review of previous promising research in the field of circulating ECM fragments in the diagnosis, treatment and monitoring of MPNs. We address the rationales and highlight new perspectives for the use of circulating ECM protein fragments as biologically plausible, noninvasive disease markers in the management of MPNs.
Collapse
Affiliation(s)
- Hans Carl Hasselbalch
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Peter Junker
- Department of Rheumatology, Odense University Hospital, 5000 Odense, Denmark;
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Trine A. Knudsen
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Morten Kranker Larsen
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Morten Orebo Holmström
- National Center for Cancer Immune Therapy, Herlev Hospital, 2730 Herlev, Denmark; (M.O.H.); (M.H.A.)
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Herlev Hospital, 2730 Herlev, Denmark; (M.O.H.); (M.H.A.)
| | - Christina Jensen
- Nordic Bioscience A/S, 2730 Herlev, Denmark; (C.J.); (M.A.K.); (N.W.)
| | - Morten A. Karsdal
- Nordic Bioscience A/S, 2730 Herlev, Denmark; (C.J.); (M.A.K.); (N.W.)
| | | |
Collapse
|
16
|
Guinard I, Nguyen T, Brassard-Jollive N, Weber J, Ruch L, Reininger L, Brouard N, Eckly A, Collin D, Lanza F, Léon C. Matrix stiffness controls megakaryocyte adhesion, fibronectin fibrillogenesis, and proplatelet formation through Itgβ3. Blood Adv 2023; 7:4003-4018. [PMID: 37171626 PMCID: PMC10410137 DOI: 10.1182/bloodadvances.2022008680] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/13/2023] Open
Abstract
Megakaryocytes (MKs) are the precursor cells of platelets, located in the bone marrow (BM). Once mature, they extend elongated projections named proplatelets through sinusoid vessels, emerging from the marrow stroma into the circulating blood. Not all signals from the microenvironment that regulate proplatelet formation are understood, particularly those from the BM biomechanics. We sought to investigate how MKs perceive and adapt to modifications of the stiffness of their environment. Although the BM is one of the softest tissue of the body, its rigidification results from excess fibronectin (FN), and other matrix protein deposition occur upon myelofibrosis. Here, we have shown that mouse MKs are able to detect the stiffness of a FN-coated substrate and adapt their morphology accordingly. Using a polydimethylsiloxane substrate with stiffness varying from physiological to pathological marrow, we found that a stiff matrix favors spreading, intracellular contractility, and FN fibrils assembly at the expense of proplatelet formation. Itgb3, but not Itgb1, is required for stiffness sensing, whereas both integrins are involved in fibrils assembly. In contrast, soft substrates promote proplatelet formation in an Itgb3-dependent manner, consistent with the ex vivo decrease in proplatelet formation and the in vivo decrease in platelet number in Itgb3-deficient mice. Our findings demonstrate the importance of environmental stiffness for MK functions with potential pathophysiological implications during pathologies that deregulate FN deposition and modulate stiffness in the marrow.
Collapse
Affiliation(s)
- Ines Guinard
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Thao Nguyen
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Noémie Brassard-Jollive
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Josiane Weber
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Laurie Ruch
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Laura Reininger
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Nathalie Brouard
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Anita Eckly
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | | | - François Lanza
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Catherine Léon
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
17
|
Panzer B, Kopp CW, Neumayer C, Koppensteiner R, Jozkowicz A, Poledniczek M, Gremmel T, Jilma B, Wadowski PP. Toll-like Receptors as Pro-Thrombotic Drivers in Viral Infections: A Narrative Review. Cells 2023; 12:1865. [PMID: 37508529 PMCID: PMC10377790 DOI: 10.3390/cells12141865] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Toll-like receptors (TLRs) have a critical role in the pathogenesis and disease course of viral infections. The induced pro-inflammatory responses result in the disturbance of the endovascular surface layer and impair vascular homeostasis. The injury of the vessel wall further promotes pro-thrombotic and pro-coagulatory processes, eventually leading to micro-vessel plugging and tissue necrosis. Moreover, TLRs have a direct role in the sensing of viruses and platelet activation. TLR-mediated upregulation of von Willebrand factor release and neutrophil, as well as macrophage extra-cellular trap formation, further contribute to (micro-) thrombotic processes during inflammation. The following review focuses on TLR signaling pathways of TLRs expressed in humans provoking pro-thrombotic responses, which determine patient outcome during viral infections, especially in those with cardiovascular diseases.
Collapse
Affiliation(s)
- Benjamin Panzer
- Department of Cardiology, Wilhelminenspital, 1090 Vienna, Austria
| | - Christoph W Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Renate Koppensteiner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Alicja Jozkowicz
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Medical Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Michael Poledniczek
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Gremmel
- Institute of Cardiovascular Pharmacotherapy and Interventional Cardiology, Karl Landsteiner Society, 3100 St. Pölten, Austria
- Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Patricia P Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
18
|
Bianchi E, Rontauroli S, Tavernari L, Mirabile M, Pedrazzi F, Genovese E, Sartini S, Dall'Ora M, Grisendi G, Fabbiani L, Maccaferri M, Carretta C, Parenti S, Fantini S, Bartalucci N, Calabresi L, Balliu M, Guglielmelli P, Potenza L, Tagliafico E, Losi L, Dominici M, Luppi M, Vannucchi AM, Manfredini R. Inhibition of ERK1/2 signaling prevents bone marrow fibrosis by reducing osteopontin plasma levels in a myelofibrosis mouse model. Leukemia 2023; 37:1068-1079. [PMID: 36928007 PMCID: PMC10169646 DOI: 10.1038/s41375-023-01867-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/20/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Clonal myeloproliferation and development of bone marrow (BM) fibrosis are the major pathogenetic events in myelofibrosis (MF). The identification of novel antifibrotic strategies is of utmost importance since the effectiveness of current therapies in reverting BM fibrosis is debated. We previously demonstrated that osteopontin (OPN) has a profibrotic role in MF by promoting mesenchymal stromal cells proliferation and collagen production. Moreover, increased plasma OPN correlated with higher BM fibrosis grade and inferior overall survival in MF patients. To understand whether OPN is a druggable target in MF, we assessed putative inhibitors of OPN expression in vitro and identified ERK1/2 as a major regulator of OPN production. Increased OPN plasma levels were associated with BM fibrosis development in the Romiplostim-induced MF mouse model. Moreover, ERK1/2 inhibition led to a remarkable reduction of OPN production and BM fibrosis in Romiplostim-treated mice. Strikingly, the antifibrotic effect of ERK1/2 inhibition can be mainly ascribed to the reduced OPN production since it could be recapitulated through the administration of anti-OPN neutralizing antibody. Our results demonstrate that OPN is a novel druggable target in MF and pave the way to antifibrotic therapies based on the inhibition of ERK1/2-driven OPN production or the neutralization of OPN activity.
Collapse
Affiliation(s)
- Elisa Bianchi
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy. .,Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | - Sebastiano Rontauroli
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy.,Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Lara Tavernari
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy.,Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Margherita Mirabile
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy.,Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Pedrazzi
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy.,Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Genovese
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy.,Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefano Sartini
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy.,Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Giulia Grisendi
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences of Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Luca Fabbiani
- Department of Medical and Surgical Sciences of Children & Adults, Pathology Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Monica Maccaferri
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124, Modena, Italy
| | - Chiara Carretta
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy.,Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sandra Parenti
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy.,Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sebastian Fantini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Niccolò Bartalucci
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Laura Calabresi
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Manjola Balliu
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Paola Guglielmelli
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Leonardo Potenza
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124, Modena, Italy
| | - Enrico Tagliafico
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124, Modena, Italy
| | - Lorena Losi
- Department of Life Sciences, Pathology Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences of Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Mario Luppi
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124, Modena, Italy
| | - Alessandro Maria Vannucchi
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy. .,Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
19
|
Tang X, Xu Q, Yang S, Huang X, Wang L, Huang F, Luo J, Zhou X, Wu A, Mei Q, Zhao C, Wu J. Toll-like Receptors and Thrombopoiesis. Int J Mol Sci 2023; 24:ijms24021010. [PMID: 36674552 PMCID: PMC9864288 DOI: 10.3390/ijms24021010] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 12/27/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Platelets are the second most abundant blood component after red blood cells and can participate in a variety of physiological and pathological functions. Beyond its traditional role in hemostasis and thrombosis, it also plays an indispensable role in inflammatory diseases. However, thrombocytopenia is a common hematologic problem in the clinic, and it presents a proportional relationship with the fatality of many diseases. Therefore, the prevention and treatment of thrombocytopenia is of great importance. The expression of Toll-like receptors (TLRs) is one of the most relevant characteristics of thrombopoiesis and the platelet inflammatory function. We know that the TLR family is found on the surface or inside almost all cells, where they perform many immune functions. Of those, TLR2 and TLR4 are the main stress-inducing members and play an integral role in inflammatory diseases and platelet production and function. Therefore, the aim of this review is to present and discuss the relationship between platelets, inflammation and the TLR family and extend recent research on the influence of the TLR2 and TLR4 pathways and the regulation of platelet production and function. Reviewing the interaction between TLRs and platelets in inflammation may be a research direction or program for the treatment of thrombocytopenia-related and inflammatory-related diseases.
Collapse
Affiliation(s)
- Xiaoqin Tang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Qian Xu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Shuo Yang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xinwu Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Long Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Feihong Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Jiesi Luo
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Xiaogang Zhou
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Anguo Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Qibing Mei
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Chunling Zhao
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- Correspondence: (C.Z.); (J.W.); Tel.: +86-186-8307-3667 (C.Z.); +86-139-8241-6641 (J.W.)
| | - Jianming Wu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
- Correspondence: (C.Z.); (J.W.); Tel.: +86-186-8307-3667 (C.Z.); +86-139-8241-6641 (J.W.)
| |
Collapse
|
20
|
Peng Z, Lv X, Huang S. Recent Progress on the Role of Fibronectin in Tumor Stromal Immunity and Immunotherapy. Curr Top Med Chem 2022; 22:2494-2505. [PMID: 35708087 DOI: 10.2174/1568026622666220615152647] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 01/20/2023]
Abstract
As a major component of the stromal microenvironment of various solid tumors, the extracellular matrix (ECM) has attracted increasing attention in cancer-related studies. ECM in the tumor stroma not only provides an external barrier and framework for tumor cell adhesion and movement, but also acts as an active regulator that modulates the tumor microenvironment, including stromal immunity. Fibronectin (Fn), as a core component of the ECM, plays a key role in the assembly and remodeling of the ECM. Hence, understanding the role of Fn in the modulation of tumor stromal immunity is of great importance for cancer immunotherapy. Hence, in-depth studies on the underlying mechanisms of Fn in tumors are urgently needed to clarify the current understanding and issues and to identify new and specific targets for effective diagnosis and treatment purposes. In this review, we summarize the structure and role of Fn, its potent derivatives in tumor stromal immunity, and their biological effects and mechanisms in tumor development. In addition, we discuss the novel applications of Fn in tumor treatment. Therefore, this review can provide prospective insight into Fn immunotherapeutic applications in tumor treatment.
Collapse
Affiliation(s)
- Zheng Peng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xiaolan Lv
- Department of Laboratory Medicine, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Shigao Huang
- Department of Radiation Oncology, The First Affiliated Hospital, Air Force Medical University, Xi an, Shaan Xi, China
| |
Collapse
|
21
|
Malara A, Gruppi C, Massa M, Tira ME, Rosti V, Balduini A, Barosi G. Elevated plasma EDA fibronectin in primary myelofibrosis is determined by high allele burden of JAK2V617F mutation and strongly predicts splenomegaly progression. Front Oncol 2022; 12:987643. [PMID: 36212480 PMCID: PMC9532599 DOI: 10.3389/fonc.2022.987643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/31/2022] [Indexed: 11/18/2022] Open
Abstract
In primary myelofibrosis, extra-domain A fibronectin (EDA-FN), the result of alternative splicing of FN gene, sustains megakaryocyte proliferation and confers a pro-inflammatory phenotype to bone marrow cell niches. In this work we assessed the levels of circulating EDA-FN in plasma samples of 122 patients with primary myelofibrosis. Patients with a homozygous JAK2V617F genotype displayed the higher level of plasma EDA-FN. Increased EDA-FN levels were associated with anemia, elevated high-sensitivity C-reactive protein, bone marrow fibrosis and splanchnic vein thrombosis at diagnosis. While no correlation was observed with CD34+ hematopoietic stem cell mobilization, elevated blood level of EDA-FN at diagnosis was a predictor of large splenomegaly (over 10 cm from the left costal margin) outcome. Thus, EDA-FN expression in primary myelofibrosis may represent the first marker of disease progression, and a novel target to treat splenomegaly.
Collapse
Affiliation(s)
- Alessandro Malara
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- *Correspondence: Alessandro Malara, ; Alessandra Balduini,
| | - Cristian Gruppi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Margherita Massa
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnostics, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Maria Enrica Tira
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnostics, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- *Correspondence: Alessandro Malara, ; Alessandra Balduini,
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnostics, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| |
Collapse
|
22
|
Khatib-Massalha E, Méndez-Ferrer S. Megakaryocyte Diversity in Ontogeny, Functions and Cell-Cell Interactions. Front Oncol 2022; 12:840044. [PMID: 35186768 PMCID: PMC8854253 DOI: 10.3389/fonc.2022.840044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic stem cells (HSCs) rely on local interactions in the bone marrow (BM) microenvironment with stromal cells and other hematopoietic cells that facilitate their survival and proliferation, and also regulate their functions. HSCs and multipotent progenitor cells differentiate into lineage-specific progenitors that generate all blood and immune cells. Megakaryocytes (Mks) are hematopoietic cells responsible for producing blood platelets, which are essential for normal hemostasis and blood coagulation. Although the most prominent function of Mks is platelet production (thrombopoiesis), other increasingly recognized functions include HSC maintenance and host immune response. However, whether and how these diverse programs are executed by different Mk subpopulations remains poorly understood. This Perspective summarizes our current understanding of diversity in ontogeny, functions and cell-cell interactions. Cumulative evidence suggests that BM microenvironment dysfunction, partly caused by mutated Mks, can induce or alter the progression of a variety of hematologic malignancies, including myeloproliferative neoplasms (MPNs) and other disorders associated with tissue scarring (fibrosis). Therefore, as an example of the heterogeneous functions of Mks in malignant hematopoiesis, we will discuss the role of Mks in the onset and progression of BM fibrosis. In this regard, abnormal interactions between of Mks and other immune cells might directly contribute to fibrotic diseases. Overall, further understanding of megakaryopoiesis and how Mks interact with HSCs and immune cells has potential clinical implications for stem cell transplantation and other therapies for hematologic malignancies, as well as for treatments to stimulate platelet production and prevent thrombocytopenia.
Collapse
Affiliation(s)
- Eman Khatib-Massalha
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Simón Méndez-Ferrer
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Instituto de Biomedicina de Sevilla-IBiS, Hospitales Universitarios Virgen del Rocío y Macarena/Spanish National Research Council (CSIC)/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
23
|
Tang A, Strat AN, Rahman M, Zhang H, Bao W, Liu Y, Shi D, An X, Manwani D, Shi P, Yazdanbakhsh K, Mendelson A. Murine bone marrow mesenchymal stromal cells have reduced hematopoietic maintenance ability in sickle cell disease. Blood 2021; 138:2570-2582. [PMID: 34329381 PMCID: PMC8678997 DOI: 10.1182/blood.2021012663] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/25/2021] [Indexed: 11/20/2022] Open
Abstract
Sickle cell disease (SCD) is characterized by hemolytic anemia, which can trigger oxidative stress, inflammation, and tissue injury that contribute to disease complications. Bone marrow mesenchymal stromal cells (MSCs) tightly regulate hematopoietic stem cell (HSC) homeostasis in health and disease, but their functionality in SCD remains unclear. We identified for the first time that murine SCD MSCs have altered gene signatures, reduced stem cell properties, and increased oxidative stress, due in part to hemolysis. Murine SCD MSCs had lower HSC maintenance ability in vitro and in vivo, as manifested by increased HSC mobilization and decreased HSC engraftment after transplant. Activation of Toll-like receptor-4 through p65 in MSCs further contributed to MSC dysfunction. Transfusions led to an improved MSC and HSC oxidative state in SCD mice. Improving the regulation between MSCs and HSCs has vital implications for enhancing clinical HSC transplantation and gene therapy outcomes and for identification of new molecular targets for alleviating SCD complications.
Collapse
Affiliation(s)
- Alice Tang
- Laboratory of Stem Cell Biology & Engineering
| | | | | | - Helen Zhang
- Laboratory of Stem Cell Biology & Engineering
| | - Weili Bao
- Laboratory of Complement Biology, and
| | | | - David Shi
- Laboratory of Stem Cell Biology & Engineering
| | - Xiuli An
- Laboratory of Membrane BiologyNew York Blood CenterNew YorkNY
| | - Deepa Manwani
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children's Hospital at Montefiore, Bronx, NY; and
| | - Patricia Shi
- Sickle Cell Clinical Research Program, New York Blood Center, New York, NY
| | | | | |
Collapse
|
24
|
Longhitano L, Tibullo D, Vicario N, Giallongo C, La Spina E, Romano A, Lombardo S, Moretti M, Masia F, Coda ARD, Venuto S, Fontana P, Parenti R, Li Volti G, Di Rosa M, Palumbo GA, Liso A. IGFBP-6/sonic hedgehog/TLR4 signalling axis drives bone marrow fibrotic transformation in primary myelofibrosis. Aging (Albany NY) 2021; 13:25055-25071. [PMID: 34905501 PMCID: PMC8714138 DOI: 10.18632/aging.203779] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/03/2021] [Indexed: 12/16/2022]
Abstract
Primary myelofibrosis is a Ph-negative chronic myeloproliferative neoplasm characterized by bone marrow fibrosis and associated with the involvement of several pathways, in addition to bone marrow microenvironment alterations, mostly driven by the activation of the cytokine receptor/JAK2 pathway. Identification of driver mutations has led to the development of targeted therapy for myelofibrosis, contributing to reducing inflammation, although this currently does not translate into bone marrow fibrosis remission. Therefore, understanding the clear molecular cut underlying this pathology is now necessary to improve the clinical outcome of patients. The present study aims to investigate the involvement of IGFBP-6/sonic hedgehog /Toll-like receptor 4 axis in the microenvironment alterations of primary myelofibrosis. We observed a significant increase in IGFBP-6 expression levels in primary myelofibrosis patients, coupled with a reduction to near-normal levels in primary myelofibrosis patients with JAK2V617F mutation. We also found that both IGFBP-6 and purmorphamine, a SHH activator, were able to induce mesenchymal stromal cells differentiation with an up-regulation of cancer-associated fibroblasts markers. Furthermore, TLR4 signaling was also activated after IGFBP-6 and purmorphamine exposure and reverted by cyclopamine exposure, an inhibitor of the SHH pathway, confirming that SHH is involved in TLR4 activation and microenvironment alterations. In conclusion, our results suggest that the IGFBP-6/SHH/TLR4 axis is implicated in alterations of the primary myelofibrosis microenvironment and that IGFBP-6 may play a central role in activating SHH pathway during the fibrotic process.
Collapse
Affiliation(s)
- Lucia Longhitano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Cesarina Giallongo
- Department of Scienze Mediche Chirurgiche e Tecnologie Avanzate "G.F. Ingrassia", University of Catania, Catania 95123, Italy
| | - Enrico La Spina
- Division of Hematology, Department of General Surgery and Medical-Surgical Specialties, A.O.U. "Policlinico-Vittorio Emanuele", University of Catania, Catania 95123, Italy
| | - Alessandra Romano
- Division of Hematology, Department of General Surgery and Medical-Surgical Specialties, A.O.U. "Policlinico-Vittorio Emanuele", University of Catania, Catania 95123, Italy
| | - Sofia Lombardo
- Department of Medical Oncology, The Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Marina Moretti
- Department of Medicine, University of Perugia, Perugia 06129, Italy
| | - Francesco Masia
- Department of Medicine, University of Perugia, Perugia 06129, Italy
| | | | - Santina Venuto
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71100, Italy
| | - Paolo Fontana
- Department of Medical Oncology, The Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Giuseppe A Palumbo
- Department of Scienze Mediche Chirurgiche e Tecnologie Avanzate "G.F. Ingrassia", University of Catania, Catania 95123, Italy
| | - Arcangelo Liso
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71100, Italy
| |
Collapse
|
25
|
Baralle M, Baralle FE. Alternative splicing and liver disease. Ann Hepatol 2021; 26:100534. [PMID: 34547477 DOI: 10.1016/j.aohep.2021.100534] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 02/04/2023]
Abstract
Alternative splicing produces complex and dynamic changes in the protein isoforms that are necessary for the proper biological functioning of the metabolic pathways involved in liver development and hepatocyte homeostasis. Changes in the physiological state of alternatively spliced forms are increasingly linked to liver pathologies. This may occur when the expression or function of the set of proteins controlling the alternative splicing processes are altered by external effectors such as oxidative stress and other environmental variations. Studies addressing these modifications reveal a complex interplay between the expression levels of different proteins that regulate the alternative splicing process as well as the changes in alternative splicing. This interplay results in a cascade of different protein isoforms that correlate with the progression of non-alcoholic fatty liver disease, hepatocellular carcinoma, and alcoholic liver disease. However, research on the detailed molecular mechanism underlying the production of these isoforms is needed. It is imperative to identify the physiological processes affected by the differentially spliced isoforms and confirm their role on the onset and maintenance of the pathology. This is required to design potential therapeutic approaches targeting the key splicing changes to revert the pathological condition as well as identify prognostic markers. In this review, we describe the complexity of the splicing process through an example to encourage researchers to go down this path. Subsequently, rather than a catalog of splicing events we have hand-picked and discuss a few selected studies of specific liver pathologies and suggested ways to focus research on these areas.
Collapse
Affiliation(s)
- Marco Baralle
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, Italy
| | - Francisco E Baralle
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy.
| |
Collapse
|
26
|
Bone marrow microenvironment of MPN cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021. [PMID: 34756245 DOI: 10.1016/bs.ircmb.2021.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
In this chapter, we will discuss the current knowledge concerning the alterations of the cellular components in the bone marrow niche in Myeloproliferative Neoplasms (MPNs), highlighting the central role of the megakaryocytes in MPN progression, and the extracellular matrix components characterizing the fibrotic bone marrow.
Collapse
|
27
|
Leimkühler NB, Costa IG, Schneider RK. From cell to cell: Identification of actionable targets in bone marrow fibrosis using single-cell technologies. Exp Hematol 2021; 104:48-54. [PMID: 34601067 DOI: 10.1016/j.exphem.2021.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 11/25/2022]
Abstract
Single-cell technologies have rapidly developed in recent years and have already had a significant impact on the research of myeloproliferative neoplasms. The increasing number of publicly available data sets allows characterization of the bone marrow niche in patients and mouse models at unprecedented resolution. Single-cell RNA sequencing has successfully been used to identify and characterize disease-driving cell populations and to identify the alarmin S100A8/A9 as an important mediator of myelofibrosis and potent therapeutic target. It is now possible to execute a streamlined set of experiments to specifically identify and validate actionable target genes functionally with the advance of reliable in vivo models and the possibility of conducting single-cell analyses with a minimal amount of patient material. The advent of large-scale analyses of both hematopoietic and non-hematopoietic bone marrow cells will allow comprehensive network analyses guiding an increasingly detailed mapping of the MPN interactome.
Collapse
Affiliation(s)
- Nils B Leimkühler
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Rebekka K Schneider
- Department of Cell Biology, Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Oncode Institute, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
28
|
Abstract
Megakaryocytes give rise to platelets, which have a wide variety of functions in coagulation, immune response, inflammation, and tissue repair. Dysregulation of megakaryocytes is a key feature of in the myeloproliferative neoplasms, especially myelofibrosis. Megakaryocytes are among the main drivers of myelofibrosis by promoting myeloproliferation and bone marrow fibrosis. In vivo targeting of megakaryocytes by genetic and pharmacologic approaches ameliorates the disease, underscoring the important role of megakaryocytes in myeloproliferative neoplasms. Here we review the current knowledge of the function of megakaryocytes in the JAK2, CALR, and MPL-mutant myeloproliferative neoplasms.
Collapse
|
29
|
Qi HM, Cao Q, Liu Q. TLR4 regulates vascular smooth muscle cell proliferation in hypertension via modulation of the NLRP3 inflammasome. Am J Transl Res 2021; 13:314-325. [PMID: 33527026 PMCID: PMC7847527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/11/2020] [Indexed: 06/12/2023]
Abstract
UNLABELLED Backgroud: Toll-like receptor 4 (TLR4), a key mediator of inflammatory responses, which is associated with vascular remodeling. The association between TLR4 and NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome in the regulation of vascular smooth muscle cell (VSMC) proliferation remains unclear. This study was to explore the role and underlying mechanisms of TLR4 in the proliferation of VSMC in hypertension. METHODS VSMC proliferation after TLR4 overexpression or downregulation was determined by CCK-8, EdU Incorporation and colony formation assays. Western blots were carried out to investigate the expression of TLR4 and NLRP3 inflammasome components in VSMCs. Next, blood pressure measurements and Hematoxylin and Eosin (HE) staining assays were performed in spontaneously hypertensive rats (SHR). Media thickness (M) and diameter lumen (L) were measured as indicators of vascular remodeling. The expression of TLR4, PCNA and NLRP3 inflammasome complex was analyzed by Western blots in the aorta of SHR. RESULTS We showed that TLR4 overexpression with cDNA enhanced, while knockdown of TLR4 with shRNA inhibited Ang II-induced VSMC proliferation. Besides, TLR4 overexpression upregulated the proteion expression of the NLRP3 inflammasome components including NLRP3, ASC and caspase-1, whereas their corresponding levels of expression were observed to decrease in TLR4 shRNA-transfected VSMCs. Knockdown of TLR4 attenuated vascular remodeling, blood pressure (BP) and the levels of NLRP3, ASC, caspase-1, IL-1β and IL-18 in SHR aortas. CONCLUSION This study revealed that TLR4 regulated Ang II-induced VSMC proliferation through modulating the NLRP3 inflammasome. Knockdown of TLR4 attenuated the BP and vascular remodeling by inhibiting the expression of the NLRP3 inflammasome component in SHR. Our results support that TLR4 regulates VSMC proliferation in hypertension via triggering the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Hui-Meng Qi
- Department of General Practice, The First Hospital of China Medical UniversityShenyang, China
| | - Qin Cao
- Department of Gastroenterolog, The First Hospital of China Medical UniversityShenyang, China
| | - Qiang Liu
- Department of Nephrology, The First Hospital of China Medical UniversityShenyang, China
| |
Collapse
|
30
|
Wirth F, Lubosch A, Hamelmann S, Nakchbandi IA. Fibronectin and Its Receptors in Hematopoiesis. Cells 2020; 9:cells9122717. [PMID: 33353083 PMCID: PMC7765895 DOI: 10.3390/cells9122717] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/08/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
Fibronectin is a ubiquitous extracellular matrix protein that is produced by many cell types in the bone marrow and distributed throughout it. Cells of the stem cell niche produce the various isoforms of this protein. Fibronectin not only provides the cells a scaffold to bind to, but it also modulates their behavior by binding to receptors on the adjacent hematopoietic stem cells and stromal cells. These receptors, which include integrins such as α4β1, α9β1, α4β7, α5β1, αvβ3, Toll-like receptor-4 (TLR-4), and CD44, are found on the hematopoietic stem cell. Because the knockout of fibronectin is lethal during embryonal development and because fibronectin is produced by almost all cell types in mammals, the study of its role in hematopoiesis is difficult. Nevertheless, strong and direct evidence exists for its stimulation of myelopoiesis and thrombopoiesis using in vivo models. Other reviewed effects can be deduced from the study of fibronectin receptors, which showed their activation modifies the behavior of hematopoietic stem cells. Erythropoiesis was only stimulated under hemolytic stress, and mostly late stages of lymphocytic differentiation were modulated. Because fibronectin is ubiquitously expressed, these interactions in health and disease need to be taken into account whenever any molecule is evaluated in hematopoiesis.
Collapse
Affiliation(s)
- Franziska Wirth
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; (F.W.); (A.L.); (S.H.)
| | - Alexander Lubosch
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; (F.W.); (A.L.); (S.H.)
| | - Stefan Hamelmann
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; (F.W.); (A.L.); (S.H.)
| | - Inaam A. Nakchbandi
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; (F.W.); (A.L.); (S.H.)
- Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-622-156-8744
| |
Collapse
|
31
|
FN-EDA mediates angiogenesis of hepatic fibrosis via integrin-VEGFR2 in a CD63 synergetic manner. Cell Death Discov 2020; 6:140. [PMID: 33293521 PMCID: PMC7722740 DOI: 10.1038/s41420-020-00378-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 10/29/2020] [Accepted: 11/13/2020] [Indexed: 01/13/2023] Open
Abstract
Pathological angiogenesis is an important component of hepatic fibrosis along with fibrous deposition, but its role is not well understood. Here, we demonstrated that fibronectin containing extra domain A(FN-EDA), a fibronectin splice variant highly expressed in hepatic fibrosis, mediated angiogenesis in disease progression. FN-EDA was positively correlated with pathological angiogenesis in hepatic fibrosis, and a reduction in FN-EDA expression was associated with diminished intrahepatic angiogenesis and fibrosis. FN-EDA mostly colocalized with hepatic stellate cells (HSCs) and interference or blockage of FN-EDA attenuated migration and tube formation in co-cultured endothelial cells. Mechanistic studies indicated that FN-EDA was secreted to promote phosphorylation of VEGFR2 with the assistance of integrin and CD63. Targeting FN-EDA-integrin combination postponed the progression of hepatic angiogenesis and fibrosis in vivo. These results indicated that FN-EDA plays an emerging role in angiogenesis in hepatic fibrosis and could be a potential therapeutic intervention for the disease.
Collapse
|
32
|
Schino M, Fiorentino V, Rossi E, Betti S, Di Cecca M, Ranucci V, Chiusolo P, Martini M, De Stefano V, Larocca LM. Bone marrow megakaryocytic activation predicts fibrotic evolution of Philadelphia-negative myeloproliferative neoplasms. Haematologica 2020; 106:3162-3169. [PMID: 33543865 PMCID: PMC8634198 DOI: 10.3324/haematol.2020.264143] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Indexed: 11/13/2022] Open
Abstract
Philadelphia-negative chronic myeloproliferative neoplasms (MPN) have been traditionally considered as indistinctly slowly progressing conditions; recent evidence proves that a subset of cases have a rapid evolution, so that MPN prognosis needs to be personalized. We identified a new morphological parameter, defined as megakaryocytic activation (M-ACT) based on the coexistence of megakaryocytic emperipolesis, megakaryocytes (MK) cluster formation and evidence of arrangement of collagen fibers around the perimeter of MK. We retrospectively analyzed the bone marrow biopsy of two MPN cohorts of patients with polycythemia (PV) (n=64) and non-PV patients (including essential thrombocythemia, and early/prefibrotic primary myelofibrosis [PMF]) (n=222). M-ACT showed a significant correlation with splenomegaly, white blood cell count, and lactate dehydrogenase serum levels in both groups, with JAK2 V617F allele burden in PV patients, and with CALR mutations, and platelet count in non-PV patients. Progression-free survival, defined as PV-to-secondary MF progression and non-PV-to-overt PMF, was worse in both PV and early/prefibrotic PMF patients with M-ACT in comparison to those without M-ACT (P<0.0001). Interestingly, M-ACT was not found in the subgroup of essential thrombocythemia patients. In conclusion, M-ACT can be helpful in the differential diagnosis of MPN and can represent a new morphologic parameter with a predictive value for progression of MPN.
Collapse
Affiliation(s)
- Mattia Schino
- Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome.
| | - Vincenzo Fiorentino
- Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome.
| | - Elena Rossi
- Department of Radiological and Hematological Sciences, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| | - Silvia Betti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| | - Monica Di Cecca
- Department of Radiological and Hematological Sciences, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome.
| | - Valentina Ranucci
- Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome.
| | - Patrizia Chiusolo
- Department of Radiological and Hematological Sciences, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| | - Maurizio Martini
- Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| | - Valerio De Stefano
- Department of Radiological and Hematological Sciences, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| | - Luigi Maria Larocca
- Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome.
| |
Collapse
|
33
|
Abstract
Sterile inflammation within primary tumor tissues can spread to distant organs that are devoid of tumor cells. This happens in a manner dependent on tumor-led secretome, before the actual metastasis occurs. The premetastatic microenvironment is established in this way and is at least partly regulated by hijacking the host innate immune system. The biological manifestation of premetastasis include increased vascular permeability, cell mobilization via the blood stream, degradation of the extracellular matrix, immunosuppression, and host antineoplastic activities.
Collapse
Affiliation(s)
- Yoshiro Maru
- Department of Pharmacology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
34
|
Yuan X, Yang S, Li W, Li J, Lin J, Wu Y, Chen Y. Construction of fibronectin conditional gene knock-out mice and the effect of fibronectin gene knockout on hematopoietic, biochemical and immune parameters in mice. PeerJ 2020; 8:e10224. [PMID: 33194415 PMCID: PMC7605225 DOI: 10.7717/peerj.10224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/29/2020] [Indexed: 12/03/2022] Open
Abstract
Fibronectin (FN) is a multi-functional glycoprotein that primarily acts as a cell adhesion molecule and tethers cells to the extra cellular matrix. In order to clarify the effect of FN deficiency on hematopoiesis, biochemical and immune parameters in mice. We constructed a tamoxifen-induced conditional (cre-loxp system) fibronectin knock-out (FnKO) mouse model on a C57BL/6 background, and monitored their behavior, fertility, histological, hematopoietic, biochemical and immunological indices. We found that the Fn KO mice had reduced fertility, high platelet counts, smaller bone marrow megakaryocytes and looser attachment between the hepatocyte and vascular endothelial junctions compared to the wild type (WT) mice. In contrast, the behavior, hematological counts, serum biochemical indices and vital organ histology were similar in both Fn KO and WT mice. This model will greatly help in elucidating the role of FN in immune-related diseases in future.
Collapse
Affiliation(s)
- Xiaohong Yuan
- Insitute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Shu Yang
- Insitute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Wen Li
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jinggang Li
- Insitute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jia Lin
- Insitute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yong Wu
- Insitute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yuanzhong Chen
- Insitute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
35
|
Abbonante V, Di Buduo CA, Malara A, Laurent PA, Balduini A. Mechanisms of platelet release: in vivo studies and in vitro modeling. Platelets 2020; 31:717-723. [PMID: 32522064 DOI: 10.1080/09537104.2020.1774532] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mechanisms related to platelet release in the context of the bone marrow niche are not completely known. In this review we discuss what has been discovered about four critical aspects of this process: 1) the bone marrow niche organization, 2) the role of the extracellular matrix components, 3) the mechanisms by which megakaryocytes release platelets and 4) the novel approaches to mimic the bone marrow environment and produce platelets ex vivo.
Collapse
Affiliation(s)
| | | | - Alessandro Malara
- Department of Molecular Medicine, University of Pavia , Pavia, Italy
| | | | | |
Collapse
|
36
|
Efthymiou G, Saint A, Ruff M, Rekad Z, Ciais D, Van Obberghen-Schilling E. Shaping Up the Tumor Microenvironment With Cellular Fibronectin. Front Oncol 2020; 10:641. [PMID: 32426283 PMCID: PMC7203475 DOI: 10.3389/fonc.2020.00641] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/06/2020] [Indexed: 12/25/2022] Open
Abstract
Normal tissue homeostasis and architecture restrain tumor growth. Thus, for a tumor to develop and spread, malignant cells must overcome growth-repressive inputs from surrounding tissue and escape immune surveillance mechanisms that curb cancer progression. This is achieved by promoting the conversion of a physiological microenvironment to a pro-tumoral state and it requires a constant dialog between malignant cells and ostensibly normal cells of adjacent tissue. Pro-tumoral reprogramming of the stroma is accompanied by an upregulation of certain extracellular matrix (ECM) proteins and their cognate receptors. Fibronectin (FN) is one such component of the tumor matrisome. This large multidomain glycoprotein dimer expressed over a wide range of human cancers is assembled by cell-driven forces into a fibrillar array that provides an obligate scaffold for the deposition of other matrix proteins and binding sites for functionalization by soluble factors in the tumor microenvironment. Encoded by a single gene, FN regulates the proliferation, motile behavior and fate of multiple cell types, largely through mechanisms that involve integrin-mediated signaling. These processes are coordinated by distinct isoforms of FN, collectively known as cellular FN (as opposed to circulating plasma FN) that arise through alternative splicing of the FN1 gene. Cellular FN isoforms differ in their solubility, receptor binding ability and spatiotemporal expression, and functions that have yet to be fully defined. FN induction at tumor sites constitutes an important step in the acquisition of biological capabilities required for several cancer hallmarks such as sustaining proliferative signaling, promoting angiogenesis, facilitating invasion and metastasis, modulating growth suppressor activity and regulating anti-tumoral immunity. In this review, we will first provide an overview of ECM reprogramming through tumor-stroma crosstalk, then focus on the role of cellular FN in tumor progression with respect to these hallmarks. Last, we will discuss the impact of dysregulated ECM on clinical efficacy of classical (radio-/chemo-) therapies and emerging treatments that target immune checkpoints and explore how our expanding knowledge of the tumor ECM and the central role of FN can be leveraged for therapeutic benefit.
Collapse
Affiliation(s)
| | - Angélique Saint
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France.,Centre Antoine Lacassagne, Nice, France
| | - Michaël Ruff
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France
| | - Zeinab Rekad
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France
| | | | | |
Collapse
|
37
|
Marín Oyarzún CP, Glembotsky AC, Goette NP, Lev PR, De Luca G, Baroni Pietto MC, Moiraghi B, Castro Ríos MA, Vicente A, Marta RF, Schattner M, Heller PG. Platelet Toll-Like Receptors Mediate Thromboinflammatory Responses in Patients With Essential Thrombocythemia. Front Immunol 2020; 11:705. [PMID: 32425934 PMCID: PMC7203216 DOI: 10.3389/fimmu.2020.00705] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/30/2020] [Indexed: 01/03/2023] Open
Abstract
Essential thrombocythemia (ET) is comprised among chronic myeloproliferative neoplasms (MPN) and is caused by driver mutations in JAK2, CALR, and MPL, which lead to megakaryocyte proliferation and prominent thrombocytosis. Thrombosis remains the main cause of morbidity in ET and is driven by the interplay between blood cells, the endothelium, the clotting cascade, and host-derived inflammatory mediators. Platelet activation plays a key role in the thrombotic predisposition, although the underlying mechanisms remain poorly defined. In addition to their role in hemostasis, platelets participate in innate immunity and inflammation owing to the expression of toll-like receptors (TLR), which recognize inflammatory signals, triggering platelet functional responses. Considering the impact of inflammation on ET procoagulant state, we assessed the contribution of TLR2 and TLR4 to platelet hemostatic and inflammatory properties in ET patients, by using Pam3CSK4 and lipopolysaccharide (LPS) as specific TLR2 and TLR4 ligands, respectively. TLR2 ligation induced increased surface translocation of α-granule-derived P-selectin and CD40L, which mediate platelet interaction with leukocytes and endothelial cells, respectively, and higher levels of dense granule-derived CD63 in patients, whereas PAC-1 binding was not increased and LPS had no effect on these platelet responses. Platelet-neutrophil aggregate formation was elevated in ET at baseline and after stimulation of both TLR2 and TLR4. In addition, ET patients displayed higher TLR2- and TLR4-triggered platelet secretion of the chemokine RANTES (CCL5), whereas von Willebrand factor release was not enhanced, revealing a differential releasate pattern for α-granule-stored inflammatory molecules. TLR-mediated hyperresponsiveness contrasted with impaired or preserved responses to classic platelet hemostatic agonists, such as TRAP-6 and thrombin. TLR2 and TLR4 expression on the platelet surface was normal, whereas phosphorylation of downstream effector ERK1/2 was higher in patients at baseline and after incubation with Pam3CSK4, which may partly explain the enhanced TLR2 response. In conclusion, exacerbated response to TLR stimulation may promote platelet activation in ET, boosting platelet/leukocyte/endothelial interactions and secretion of inflammatory mediators, overall reinforcing the thromboinflammatory state. These findings highlight the role of platelets as inflammatory sentinels in MPN prothrombotic scenario and provide additional evidence for the close intertwining between thrombosis and inflammation in this setting.
Collapse
Affiliation(s)
- Cecilia P Marín Oyarzún
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Ana C Glembotsky
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Nora P Goette
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Paola R Lev
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Geraldine De Luca
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - María C Baroni Pietto
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Beatriz Moiraghi
- Department of Hematology, Hospital General de Agudos José María Ramos Mejía, Buenos Aires, Argentina
| | | | - Angeles Vicente
- Department of Hematology, Hospital Alemán, Buenos Aires, Argentina
| | - Rosana F Marta
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (IMEX)- CONICET, National Academy of Medicine, Buenos Aires, Argentina
| | - Paula G Heller
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
38
|
D'Atri LP, Rodríguez CS, Miguel CP, Pozner RG, Ortiz Wilczyñski JM, Negrotto S, Carrera-Silva EA, Heller PG, Schattner M. Activation of toll-like receptors 2 and 4 on CD34 + cells increases human megakaryo/thrombopoiesis induced by thrombopoietin. J Thromb Haemost 2019; 17:2196-2210. [PMID: 31397069 DOI: 10.1111/jth.14605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/07/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Platelet Toll-like receptor (TLR)2/4 are key players in amplifying the host immune response; however, their role in human megakaryo/thrombopoiesis has not yet been defined. OBJECTIVES We evaluated whether Pam3CSK4 or lipopolysaccharide (LPS), TLR2/4 ligands respectively, modulate human megakaryocyte development and platelet production. METHODS CD34+ cells from human umbilical cord were stimulated with LPS or Pam3CSK4 with or without thrombopoietin (TPO). RESULTS CD34+ cells and megakaryocytes express TLR2 and TLR4 at both RNA and protein level; however, direct stimulation of CD34+ cells with LPS or Pam3CSK4 had no effect on cell growth. Interestingly, both TLR ligands markedly increased TPO-induced CD34+ cell proliferation, megakaryocyte number and maturity, proplatelet and platelet production when added at day 0. In contrast, this synergism was not observed when TLR agonists were added 7 days after TPO addition. Interleukin-6 (IL-6) release was observed upon CD34+ or megakaryocyte stimulation with LPS or Pam3CSK4 but not with TPO and this effect was potentiated in combination with TPO. The increased proliferation and IL-6 production induced by TPO + LPS or Pam3CSK4 were suppressed by TLR2/4 or IL-6 neutralizing antibodies, as well as by PI3K/AKT and nuclear factor-κB inhibitors. Additionally, increased proplatelet and platelet production were associated with enhanced nuclear translocation of nuclear factor-E2. Finally, the supernatants of CD34+ cells stimulated with TPO+LPS-induced CFU-M colonies. CONCLUSIONS Our data suggest that the activation of TLR2 and TLR4 in CD34+ cells and megakaryocytes in the presence of TPO may contribute to warrant platelet provision during infection episodes by an autocrine IL-6 loop triggered by PI3K/NF-κB axes.
Collapse
Affiliation(s)
- Lina Paola D'Atri
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Camila Sofía Rodríguez
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Carolina Paula Miguel
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Roberto Gabriel Pozner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Juan Manuel Ortiz Wilczyñski
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Soledad Negrotto
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Eugenio Antonio Carrera-Silva
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Paula Graciela Heller
- Institute of Medical Research Dr. Alfredo Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
- Department of Hematology Research, National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Institute of Medical Research (IDIM), Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| |
Collapse
|
39
|
Yuan Y, Yang L, Liu T, Zhang H, Lu Q. Osteoclastogenesis inhibition by mutated IGSF23 results in human osteopetrosis. Cell Prolif 2019; 52:e12693. [PMID: 31560140 PMCID: PMC6869366 DOI: 10.1111/cpr.12693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/27/2019] [Accepted: 08/06/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Osteopetrosis is a rare inherited skeletal disease characterized by increased bone mineral density due to the loss of osteoclast function or differentiation potential. MATERIALS AND METHODS The study involved a Chinese patient with osteopetrosis (the proband) and her immediate family members and 180 controls without osteopetrosis. Bone density of the femoral neck, lumbar spine and total body was measured using dual-energy x-ray absorptiometry. Osteoclast differentiation by the participants' peripheral blood mononuclear cells (PBMCs) was investigated using tartrate-resistant acid phosphatase (TRAP) staining. Osteoblast differentiation was examined with Alizarin Red S staining. Reverse transcription-quantitative PCR was used to amplify immunoglobulin superfamily member 23 (IGSF23), c-FOS and nuclear factor of activated T cells 1 (NFATC1). RESULTS We found a homozygous mutation (c.295C>T) in the IGSF23 gene in two osteopetrosis samples. The mutation led to the formation of a stop codon, causing loss of the immunoglobulin-like domain and the whole transmembrane domain. PBMCs from the proband (IGSF23-/- ) exhibited poor ability for differentiating into mature osteoclasts in vitro. Overexpression of IGSF23 rescued the ability of IGSF23-/- PBMCs to differentiate into osteoclasts. Moreover, knockdown of IGSF23 reversed the bone loss in OVX mice by injecting AAV-shIGSF23 into mice femoral bone marrow cavity. Furthermore, we also found that the IGSF23 mutation led to decreased c-Fos and NFATC1 expression levels by inhibiting the mitogen-activated protein kinase signalling pathways. CONCLUSIONS IGSF23-mediated osteoclast differentiation of PBMCs may serve as a potential target in osteoporosis therapy.
Collapse
Affiliation(s)
- Ying Yuan
- Institute of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Yang
- Department of Endocrinology, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Ting Liu
- Institute of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Zhang
- Institute of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
40
|
Zhou Y, Zhang B, Li C, Huang X, Cheng H, Bao X, Zhao F, Cheng Q, Yue S, Han J, Luo Z. Megakaryocytes participate in the occurrence of bleomycin-induced pulmonary fibrosis. Cell Death Dis 2019; 10:648. [PMID: 31501415 PMCID: PMC6733875 DOI: 10.1038/s41419-019-1903-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/02/2019] [Accepted: 08/11/2019] [Indexed: 12/12/2022]
Abstract
Pulmonary fibrosis is characterized by the remodeling of fibrotic tissue and collagen deposition, which mainly results from aberrant fibroblasts proliferation and trans-differentiation to myofibroblasts. Patients with chronic myelogenous leukemia, myeloproliferative disorder, and scleroderma with pulmonary fibrosis complications show megakaryocyte infiltration in the lung. In this study, we demonstrated that the number of CD41+ megakaryocytes increased in bleomycin (BLM)-induced lung fibrosis tissues through the Chemokine (CXCmotif) ligand 12/Chemokine receptor 4 (CXCL12/CXCR4) axis. Pharmacological inhibition of the CXCL12/CXCR4 axis with WZ811 prevented migration of CD41+ megakaryocytes induced by BLM-injured lung tissue ex vivo and in vivo. In addition, WZ811 significantly attenuated lung fibrosis after BLM challenge. Moreover, megakaryocytes directly promoted fibroblast proliferation and trans-differentiation to myofibroblasts. We conclude that thrombopoietin (TPO) activated megakaryocytes through transforming growth factor β (TGF-β) pathway to promote fibroblast proliferation and trans-differentiation to myofibroblasts, which is abolished by treatment with selective TGF-βR-1/ALK5 inhibitors. Therefore, CD41+ megakaryocytes migrate to injured lung tissue partially through the CXCL12/CXCR4 axis to promote the proliferation and trans-differentiation of fibroblasts through direct contact and the TGF-β1 pathway.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bo Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chen Li
- Department of Physiology, Changzhi medical college, Changzhi, Shanxi, China
| | - XiaoTing Huang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - HaiPeng Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - XingWen Bao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - FeiYan Zhao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - QingMei Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - ShaoJie Yue
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - JianZhong Han
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| | - ZiQiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|