1
|
Bulté D, Barzaghi F, Mesa-Nuñez C, Rigamonti C, Basso-Ricci L, Visconti C, Crippa S, Pettinato E, Gilioli D, Milani R, Quaranta P, Caorsi R, Cafaro A, Cangemi G, Lupia M, Schena F, Grossi A, Di Colo G, Federici S, Insalaco A, De Benedetti F, Marktel S, Di Micco R, Bernardo ME, Scala S, Cicalese MP, Conti F, Miano M, Gattorno M, Dufour C, Aiuti A, Mortellaro A. Early bone marrow alterations in patients with adenosine deaminase 2 deficiency across disease phenotypes and severities. J Allergy Clin Immunol 2025; 155:616-627.e8. [PMID: 39284370 PMCID: PMC11804788 DOI: 10.1016/j.jaci.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Deficiency of adenosine deaminase 2 (DADA2) is a complex monogenic disease caused by recessive mutations in the ADA2 gene. DADA2 exhibits a broad clinical spectrum encompassing vasculitis, immunodeficiency, and hematologic abnormalities. Yet, the impact of DADA2 on the bone marrow (BM) microenvironment is largely unexplored. OBJECTIVE This study comprehensively examined the BM and peripheral blood of pediatric and adult patients with DADA2 presenting with rheumatologic/immunologic symptoms or severe hematologic manifestations. METHODS Immunophenotyping of hematopoietic stem cells (HSCs), progenitor cells, and mature cell populations was performed for 18 patients with DADA2. We also conducted a characterization of mesenchymal stromal cells. RESULTS Our study revealed a significant decrease in primitive HSCs and progenitor cells, alongside their reduced clonogenic capacity and multilineage differentiation potential. These BM defects were evident in patients with both severe and nonsevere hematologic manifestations, including pediatric patients, demonstrating that BM disruption can emerge silently and early on, even in patients who do not show obvious hematologic symptoms. Beyond stem cells, there was a reduction in mature cell populations in the BM and peripheral blood, affecting myeloid, erythroid, and lymphoid populations. Furthermore, BM mesenchymal stromal cells in patients with DADA2 exhibited reduced clonogenic and proliferation capabilities and were more prone to undergo cellular senescence marked by elevated DNA damage. CONCLUSIONS Our exploration into the BM landscape of patients with DADA2 sheds light on the critical hematologic dimension of the disease and emphasizes the importance of vigilant monitoring, even in the case of subclinical presentation.
Collapse
Affiliation(s)
- Dimitri Bulté
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Barzaghi
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Mesa-Nuñez
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Rigamonti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Luca Basso-Ricci
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Camilla Visconti
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefania Crippa
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuela Pettinato
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Diego Gilioli
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Milani
- Immunohematology and Transfusion Medicine Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Pamela Quaranta
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Caorsi
- UOC Rheumatology and Autoinflammatory Diseases, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Alessia Cafaro
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Giuliana Cangemi
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Michela Lupia
- Hematology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Francesca Schena
- UOC Rheumatology and Autoinflammatory Diseases, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Alice Grossi
- Laboratorio Genetica e Genomica delle Malattie Rare, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Giulia Di Colo
- Vita-Salute San Raffaele University, Milan, Italy; Immunology, Rheumatology, Allergy and Rare Disease Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Silvia Federici
- Division of Rheumatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Antonella Insalaco
- Division of Rheumatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Sarah Marktel
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Di Micco
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Ester Bernardo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy; Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Serena Scala
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Pia Cicalese
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy; Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Francesca Conti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Maurizio Miano
- Hematology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marco Gattorno
- UOC Rheumatology and Autoinflammatory Diseases, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Carlo Dufour
- Hematology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy; Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandra Mortellaro
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
2
|
Liu S, Vivona ES, Kurre P. Why hematopoietic stem cells fail in Fanconi anemia: Mechanisms and models. Bioessays 2025; 47:e2400191. [PMID: 39460396 DOI: 10.1002/bies.202400191] [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: 08/06/2024] [Revised: 09/27/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
Fanconi anemia (FA) is generally classified as a DNA repair disorder, conferring a genetic predisposition to cancer and prominent bone marrow failure (BMF) in early childhood. Corroborative human and murine studies point to a fetal origin of hematopoietic stem cell (HSC) attrition under replicative stress. Along with intriguing recent insights into non-canonical roles and domain-specific functions of FA proteins, these studies have raised the possibility of a DNA repair-independent BMF etiology. However, deeper mechanistic insight is critical as current curative options of allogeneic stem cell transplantation and emerging gene therapy have limited eligibility, carry significant side effects, and involve complex procedures restricted to resource-rich environments. To develop rational and broadly accessible therapies for FA patients, the field will need more faithful disease models that overcome the scarcity of patient samples, leverage technological advances, and adopt investigational clinical trial designs tailored for rare diseases.
Collapse
Affiliation(s)
- Suying Liu
- Comprehensive Bone Marrow Failure Center, Children's Hospital of Philadelphia, Philadelphia, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - E S Vivona
- Comprehensive Bone Marrow Failure Center, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Peter Kurre
- Comprehensive Bone Marrow Failure Center, Children's Hospital of Philadelphia, Philadelphia, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
3
|
Jayabal P, Ma X, Shiio Y. Roles of USP1 in Ewing sarcoma. Genes Cancer 2024; 15:15-27. [PMID: 38323120 PMCID: PMC10843185 DOI: 10.18632/genesandcancer.235] [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] [Received: 08/28/2023] [Accepted: 12/19/2023] [Indexed: 02/08/2024] Open
Abstract
Ewing sarcoma is a cancer of bone and soft tissue in children and young adults that is driven by the EWS-ETS fusion transcription factor, most commonly EWS-FLI1. We previously reported that Ewing sarcoma harbors two populations of cells, the CD133high population displaying higher growth rate and the CD133low population displaying chemotherapy resistance. We now find that the ubiquitin-specific protease 1 (USP1) is a transcriptional target of the EWS-FLI1 fusion oncoprotein, expressed at high and low levels in the CD133high and the CD133low populations, respectively, and determines chemo-sensitivity. We also find that USP1 inhibits cdc42, increases EWS-FLI1 transcriptional output, and simulates Ewing sarcoma growth. We show that chemo-sensitization by USP1 is independent of cdc42. A pharmacological inhibitor of USP1 was able to activate cdc42 and inhibit Ewing sarcoma growth. These results uncover critical roles for USP1 in Ewing sarcoma, which regulates growth and chemo-sensitivity via distinct mechanisms.
Collapse
Affiliation(s)
- Panneerselvam Jayabal
- Greehey Children’s Cancer Research Institute, The University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Xiuye Ma
- Greehey Children’s Cancer Research Institute, The University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Yuzuru Shiio
- Greehey Children’s Cancer Research Institute, The University of Texas Health Science Center, San Antonio, TX 78229, USA
- Mays Cancer Center, The University of Texas Health Science Center, San Antonio, TX 78229, USA
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center, San Antonio, TX 78229, USA
| |
Collapse
|
4
|
Özdemir C, Muratoğlu B, Özel BN, Alpdündar-Bulut E, Tonyalı G, Ünal Ş, Uçkan-Çetinkaya D. Multiparametric analysis of etoposide exposed mesenchymal stem cells and Fanconi anemia cells: implications in development of secondary myeloid malignancy. Clin Exp Med 2023; 23:4511-4524. [PMID: 37179284 DOI: 10.1007/s10238-023-01087-0] [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: 01/01/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023]
Abstract
Secondary acute myeloid leukemia (sAML) may develop following a prior therapy or may evolve from an antecedent hematological disorder such as Fanconi Anemia (FA). Pathophysiology of leukemic evolution is not clear. Etoposide (Eto) is a chemotherapeutic agent implicated in development of sAML. FA is an inherited bone marrow (BM) failure disease characterized by genomic instability and xenobiotic susceptibility. Here, we hypothesized that alterations in the BM niche may play a critical/driver role in development of sAML in both conditions. Expression of selected genes involved in xenobiotic metabolism, DNA double-strand break response, endoplasmic reticulum (ER) stress, heat shock response and cell cycle regulation were determined in BM mesenchymal stem cells (MSCs) of healthy controls and FA patients at steady state and upon exposure to Eto at different concentrations and in recurrent doses. Expression of CYPA1, p53, CCNB1, Dicer1, CXCL12, FLT3L and TGF-Beta genes were significantly downregulated in FA-MSCs compared with healthy controls. Eto exposure induced significant alterations in healthy BM-MSCs with increased expression of CYP1A1, GAD34, ATF4, NUPR1, CXCL12, KLF4, CCNB1 and nuclear localization of Dicer1. Interestingly, FA-MSCs did not show significant alterations in these genes upon Eto exposure. As opposed to healthy MSCs DICER1 gene expression and intracellular localization was not altered on FA BM-MSCs after Eto treatment. These results showed that Eto is a highly potent molecule and has pleiotropic effects on BM-MSCs, FA cells show altered expression profile compared to healthy controls and Eto exposure on FA cells shows differential profile than healthy controls.
Collapse
Affiliation(s)
- Cansu Özdemir
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey.
| | - Bihter Muratoğlu
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey
| | - Buse Nurten Özel
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
| | - Esin Alpdündar-Bulut
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey
- Division of Hematology-Oncology, Faculty of Medicine, Department of Pediatrics, Hacettepe University, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey
| | - Gülsena Tonyalı
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey
| | - Şule Ünal
- Division of Hematology-Oncology, Faculty of Medicine, Department of Pediatrics, Hacettepe University, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey
- Research Center for Fanconi Anemia and Other IBMFSs, Hacettepe University, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey
| | - Duygu Uçkan-Çetinkaya
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey.
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey.
- Division of Hematology-Oncology, Faculty of Medicine, Department of Pediatrics, Hacettepe University, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey.
- Research Center for Fanconi Anemia and Other IBMFSs, Hacettepe University, 06100 Gevher Nesibe Street, Sihhiye, Altındağ, Ankara, Turkey.
| |
Collapse
|
5
|
Oppezzo A, Monney L, Kilian H, Slimani L, Maczkowiak-Chartois F, Rosselli F. Fanca deficiency is associated with alterations in osteoclastogenesis that are rescued by TNFα. Cell Biosci 2023; 13:115. [PMID: 37355617 DOI: 10.1186/s13578-023-01067-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/09/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Hematopoietic stem cells (HSCs) reside in the bone marrow (BM) niche, which includes bone-forming and bone-resorbing cells, i.e., osteoblasts (OBs) and osteoclasts (OCs). OBs originate from mesenchymal progenitors, while OCs are derived from HSCs. Self-renewal, proliferation and differentiation of HSCs are under the control of regulatory signals generated by OBs and OCs within the BM niche. Consequently, OBs and OCs control both bone physiology and hematopoiesis. Since the human developmental and bone marrow failure genetic syndrome fanconi anemia (FA) presents with skeletal abnormalities, osteoporosis and HSC impairment, we wanted to test the hypothesis that the main pathological abnormalities of FA could be related to a defect in OC physiology and/or in bone homeostasis. RESULTS We revealed here that the intrinsic differentiation of OCs from a Fanca-/- mouse is impaired in vitro due to overactivation of the p53-p21 axis and defects in NF-kB signaling. The OC differentiation abnormalities observed in vitro were rescued by treating Fanca-/- cells with the p53 inhibitor pifithrin-α, by treatment with the proinflammatory cytokine TNFα or by coculturing them with Fanca-proficient or Fanca-deficient osteoblastic cells. CONCLUSIONS Overall, our results highlight an unappreciated role of Fanca in OC differentiation that is potentially circumvented in vivo by the presence of OBs and TNFα in the BM niche.
Collapse
Affiliation(s)
- Alessia Oppezzo
- CNRS UMR9019, Équipe labellisée La Ligue contre le Cancer, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94805, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
- Université Paris Saclay, Orsay, France
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Lovely Monney
- CNRS UMR9019, Équipe labellisée La Ligue contre le Cancer, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94805, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
- Université Paris Saclay, Orsay, France
| | - Henri Kilian
- URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV), FHU-DDS-net, Dental School, Université de Paris, Montrouge, France
| | - Lofti Slimani
- URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV), FHU-DDS-net, Dental School, Université de Paris, Montrouge, France
| | - Frédérique Maczkowiak-Chartois
- CNRS UMR9019, Équipe labellisée La Ligue contre le Cancer, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94805, Villejuif, France
- Gustave Roussy Cancer Center, Villejuif, France
- Université Paris Saclay, Orsay, France
| | - Filippo Rosselli
- CNRS UMR9019, Équipe labellisée La Ligue contre le Cancer, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94805, Villejuif, France.
- Gustave Roussy Cancer Center, Villejuif, France.
- Université Paris Saclay, Orsay, France.
| |
Collapse
|
6
|
Yan J, Wan D. Dysregulation of circulating CDC42 and its correlation with demographic characteristics, comorbidities, tumor features, chemotherapeutic regimen and survival profile in non-small-cell lung cancer patients. J Clin Lab Anal 2021; 36:e24140. [PMID: 34952984 DOI: 10.1002/jcla.24140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/09/2021] [Accepted: 11/13/2021] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE Cell division control protein 42 (CDC42) induces the immune escape, represses the CD8+ T-cell activation, and further leads to the tumor metastasis in various neoplasms, whereas the correlation of circulating CDC42 with clinical features and prognosis of non-small-cell lung cancer (NSCLC) remains elusive. Hence, the current study aimed to investigate this topic. METHODS Peripheral blood mononuclear cells from 263 NSCLC patients before treatment and 50 health controls (HC) were used for CDC42 determination by reverse transcription quantitative polymerase chain reaction (RT-qPCR) assay. RESULTS CDC42 expression was higher in NSCLC patients than HCs (p < 0.001). Besides, elevated CDC42 expression was correlated with the occurrence of lymph node (LYN) metastasis (p = 0.003) and advanced TNM stage (p = 0.007), but not related to other tumor features, demographic characteristics, comorbidities, nor neoadjuvant/adjuvant chemotherapy (all p > 0.05). Additionally, elevated CDC42 expression was correlated with unfavorable accumulating disease-free survival (DFS) (p < 0.001) and overall survival (OS) (p = 0.025). More importantly, multivariate Cox's proportional hazard regression analysis revealed that elevated CDC42 expression (hazard ratio (HR): 1.284, p < 0.001) and higher TNM stage (HR: 1.428, p = 0.003) were independently associated with shorter DFS, meanwhile elevated CDC42 expression (HR: 1.193, p = 0.035), higher pathological grade (HR: 1.558, p = 0.003), higher TNM stage (HR: 1.703, p = 0.001) and higher Eastern Cooperative Oncology Group performance status (ECOG PS) score (HR: 1.538, p = 0.038) were independently correlated with unsatisfying OS. CONCLUSION Circulating CDC42 is highly expressed with its overexpression linked with LYN metastasis, poor DFS, and OS in NSCLC patients.
Collapse
Affiliation(s)
- Jie Yan
- Department of Thoracic and Cardiovascular Surgery, Huangshi Central Hospital, (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, China
| | - Daihong Wan
- Department of Surgical Anesthesiology, Huangshi Central Hospital, (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, China
| |
Collapse
|
7
|
Mastelaro de Rezende M, Zenker Justo G, Julian Paredes-Gamero E, Gosens R. Wnt-5A/B Signaling in Hematopoiesis throughout Life. Cells 2020; 9:cells9081801. [PMID: 32751131 PMCID: PMC7465103 DOI: 10.3390/cells9081801] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/08/2023] Open
Abstract
Wnt signaling is well-known to play major roles in the hematopoietic system, from embryogenesis to aging and disease. In addition to the main β-catenin-dependent pathway, it is now clear that Wnt5a and the structurally related Wnt5b are essential for hematopoiesis, bone marrow colonization and the final steps of hematopoietic stem cell (HSC) maturation via β-catenin-independent signaling. Wnt5a and Wnt5b ligands prevent hematopoietic exhaustion (by maintaining quiescent, long-term HSCs), induce the proliferation of progenitors, and guide myeloid development, in addition to being involved in the development of aging-related alterations. The aim of this review is to summarize the current knowledge on these roles of Wnt5a and Wn5b signaling in the hematopoietic field.
Collapse
Affiliation(s)
- Marina Mastelaro de Rezende
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo 04044-020, Brazil; (M.M.d.R.); (G.Z.J.); (E.J.P.-G.)
- Department of Molecular Pharmacology, University of Groningen, Groningen 9713 AV, The Netherlands
| | - Giselle Zenker Justo
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo 04044-020, Brazil; (M.M.d.R.); (G.Z.J.); (E.J.P.-G.)
- Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), Diadema 09913-030, Brazil
| | - Edgar Julian Paredes-Gamero
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo 04044-020, Brazil; (M.M.d.R.); (G.Z.J.); (E.J.P.-G.)
- Faculdade de Ciências Farmacêuticas, Universidade Federal de Mato Grosso do Sul, Campo Grande 79070-900, Brazil
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen 9713 AV, The Netherlands
- Correspondence: ; Tel.: +31-50363-8177
| |
Collapse
|
8
|
Cagnan I, Cosgun E, Konu O, Uckan D, Gunel-Ozcan A. PKNOX2 expression and regulation in the bone marrow mesenchymal stem cells of Fanconi anemia patients and healthy donors. Mol Biol Rep 2019; 46:669-678. [PMID: 30515693 DOI: 10.1007/s11033-018-4522-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/22/2018] [Indexed: 12/30/2022]
|
9
|
Sivananthan A, Shields D, Fisher R, Hou W, Zhang X, Franicola D, Epperly MW, Wipf P, Greenberger JS. Continuous One Year Oral Administration of the Radiation Mitigator, MMS350, after Total-Body Irradiation, Restores Bone Marrow Stromal Cell Proliferative Capacity and Reduces Senescence in Fanconi Anemia (Fanca -/-) Mice. Radiat Res 2018; 191:139-153. [PMID: 30499383 DOI: 10.1667/rr15199.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We quantitated age-related accumulation of senescent cells in irradiated Fanconi anemia (FA) (Fanca-/- mouse cell lines in vitro, and monitored the effect of continuous administration (via drinking water) of the water-soluble radiation mitigator, MMS350, on tissues in vivo over one year after 7.5 Gy total-body irradiation (TBI). Irradiated Fanca-/- mouse bone marrow stromal cell lines showed increased numbers of beta-galactosidase- and p21-positive senescent cells compared to Fanca+/+ cell lines, which was reduced by MMS350. One week after 7.5 Gy TBI, Fanca-/- mice showed increased numbers of senescent cells in spleen compared to Fanca+/+ controls, decreased bone marrow cellularity, failure of explanted bone marrow to proliferate in vitro to form a hematopoietic microenvironment and no detectable single stromal cell cloning capacity. There was no detectable amelioration by MMS350 administration at one week. In contrast, one year post-TBI, Fanca-/- mice demonstrated fewer senescent cells in brain and spleen compared to Fanca+/+ controls. While Fanca-/- mouse bone marrow stromal cells explanted one year post-TBI still failed to proliferate in vitro, continuous oral administration of 400 µ M, MMS350 in drinking water restored explanted stromal cell proliferation. The data indicate that continuous administration of MMS350 modulated several properties of TBI-accelerated aging in Fanca-/- mice as well as control mice, and support further study of MMS350 as a modulator of radiation late effects.
Collapse
Affiliation(s)
- Aranee Sivananthan
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Donna Shields
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Renee Fisher
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Wen Hou
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Xichen Zhang
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Darcy Franicola
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Michael W Epperly
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Peter Wipf
- b Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Joel S Greenberger
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
10
|
Wu L, Amarachintha S, Xu J, Oley F, Du W. Mesenchymal COX2-PG secretome engages NR4A-WNT signalling axis in haematopoietic progenitors to suppress anti-leukaemia immunity. Br J Haematol 2018; 183:445-456. [PMID: 30106181 PMCID: PMC6391996 DOI: 10.1111/bjh.15548] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/05/2018] [Indexed: 02/02/2023]
Abstract
The bone marrow (BM) microenvironment (niche) plays important roles in supporting normal/abnormal haematopoiesis. We investigated the interaction between leukaemic mesenchymal niche and haematopoietic stem and progenitor cells (HSPCs) using the model of Fanconi anaemia (FA), a genetic disorder characterized by BM failure and leukaemia. Healthy donor HSPCs co-cultured on mesenchymal stromal cells (MSCs) derived from FA patients with acute myeloid leukaemia (AML) exhibited higher human engraftment and myeloid expansion in Non-obese diabetic severe combined immunodeficiency IL-2γ-/- /SGM3 recipients. Untargeted metabolomics analysis revealed the progressively elevated prostaglandins (PGs) in the MSCs of FA patients with myelodysplastic syndromes (MDS) and AML. Reduced secretion of PGs subsequent to inflammatory cyclooxygenase 2 (COX2) inhibition ameliorated HSPC/myeloid expansion. Transcriptome analysis demonstrated dysregulation of genes involved in the NR4A family of transcription factors (TFs) and WNT/β-catenin signalling pathway in FA-AML-MSC-co-cultured-CD34+ cells. COX2 inhibition led to significantly decreased NR4A TFs and WNT signalling genes expression. Mechanistically, NR4A1 and NR4A2 synergistically activate the CTNNB1 gene promoter . Knocking down CTNNB1 or NR4A1 in AML-MSC-co-cultured-CD34+ cells increased leukaemia-reactive T-effector cells production and rescued anti-leukaemia immunity. Together, these findings suggest that specific interactions between leukaemic mesenchymal niche and HSPCs orchestrate a novel COX2/PG-NR4A/WNT signalling axis, connecting inflammation, cellular metabolism and cancer immunity.
Collapse
MESH Headings
- Animals
- Cyclooxygenase 2/immunology
- Female
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Male
- Mesenchymal Stem Cells/immunology
- Mesenchymal Stem Cells/pathology
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Neoplasm Proteins/immunology
- Nuclear Receptor Subfamily 4, Group A, Member 1/immunology
- Nuclear Receptor Subfamily 4, Group A, Member 2/immunology
- Wnt Signaling Pathway/immunology
Collapse
Affiliation(s)
- Limei Wu
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV
| | - Surya Amarachintha
- The Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Jian Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Frank Oley
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV
| | - Wei Du
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV
- West Virginia University Cancer Institute, Morgantown, WV, USA
| |
Collapse
|