1
|
Arora S, Vachhani P, Bose P. Investigational drugs in early phase trials for myelofibrosis. Expert Opin Investig Drugs 2024; 33:1231-1244. [PMID: 39604120 PMCID: PMC11669310 DOI: 10.1080/13543784.2024.2434696] [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: 10/05/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
INTRODUCTION Myelofibrosis (MF) is a chronic myeloproliferative neoplasm characterized by bone marrow fibrosis, cytopenias, and organomegaly. Four JAK inhibitors are US-FDA approved for treatment of MF. While these drugs reduce symptom burden and spleen size to varying degrees, they do not affect the natural disease course or decrease the risk of leukemic transformation. Therefore, there is a strong need for newer therapies to further advance the field and improve the outcomes of MF. In this review, we cover novel therapies for MF currently in early stages of development. AREAS COVERED We present the latest data from early phase clinical trials in MF using drugs with diverse therapeutic mechanisms, including novel JAK-STAT pathway inhibitors, epigenetic therapies, antifibrotic agents, and immunotherapeutic strategies. Additionally, we cover drugs targeted toward anemia improvement in MF. EXPERT OPINION Numerous agents representing diverse drug classes are in clinical development for MF. While deeper and durable improvements in splenomegaly, symptoms, and anemia are the main clinical objectives, a number of putative biomarkers are being assessed as measures of potential 'disease modification.' Although JAK inhibitor monotherapy represents the current standard, it is hoped that JAK inhibitor-based rational combinations and driver mutation-specific therapies will soon usher in a new era.
Collapse
Affiliation(s)
- Sankalp Arora
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Pankit Vachhani
- Department of Medicine, Division of Hematology and Oncology, The University of Alabama at Birmingham, Birmingham, AL
| | - Prithviraj Bose
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
2
|
Ghosh K, Shome DK, Kulkarni B, Ghosh MK, Ghosh K. Fibrosis and bone marrow: understanding causation and pathobiology. J Transl Med 2023; 21:703. [PMID: 37814319 PMCID: PMC10561412 DOI: 10.1186/s12967-023-04393-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/28/2023] [Indexed: 10/11/2023] Open
Abstract
Bone marrow fibrosis represents an important structural change in the marrow that interferes with some of its normal functions. The aetiopathogenesis of fibrosis is not well established except in its primary form. The present review consolidates current understanding of marrow fibrosis. We searched PubMed without time restriction using key words: bone marrow and fibrosis as the main stem against the terms: growth factors, cytokines and chemokines, morphology, megakaryocytes and platelets, myeloproliferative disorders, myelodysplastic syndrome, collagen biosynthesis, mesenchymal stem cells, vitamins and minerals and hormones, and mechanism of tissue fibrosis. Tissue marrow fibrosis-related papers were short listed and analysed for the review. It emerged that bone marrow fibrosis is the outcome of complex interactions between growth factors, cytokines, chemokines and hormones together with their facilitators and inhibitors. Fibrogenesis is initiated by mobilisation of special immunophenotypic subsets of mesenchymal stem cells in the marrow that transform into fibroblasts. Fibrogenic stimuli may arise from neoplastic haemopoietic or non-hematopoietic cells, as well as immune cells involved in infections and inflammatory conditions. Autoimmunity is involved in a small subset of patients with marrow fibrosis. Megakaryocytes and platelets are either directly involved or are important intermediaries in stimulating mesenchymal stem cells. MMPs, TIMPs, TGF-β, PDGRF, and basic FGF and CRCXL4 chemokines are involved in these processes. Genetic and epigenetic changes underlie many of these conditions.
Collapse
Affiliation(s)
- Kanjaksha Ghosh
- National Institute of Immunohaematology, 13 Th Fl KEM Hospital, Parel, Mumbai, 400012, India.
| | - Durjoy K Shome
- Department of Pathophysiology, American University of Antigua College of Medicine, Coolidge, Antigua and Barbuda
| | - Bipin Kulkarni
- Department of Molecular Biology and Haemostasis, National Institute of Immunohaematology, 13Th Fl KEM Hospital, Parel, Mumbai, 400012, India
| | - Malay K Ghosh
- Department of Haematology, Nilratan Sarkar Medical College, Kolkata, 700014, West Bengal, India
| | - Kinjalka Ghosh
- Department of Clinical Biochemistry, Tata Medical Centre and Homi Bhaba National Institute, Parel, Mumbai, 400012, India
| |
Collapse
|
3
|
Wang JN, Li BJ, Yuan J, Li Y. Brucellosis complicated by myelofibrosis: report of five cases and review of literature. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2023; 16:164-171. [PMID: 37559685 PMCID: PMC10408432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/21/2023] [Indexed: 08/11/2023]
Abstract
Myelofibrosis is a myeloproliferative tumor, that can be secondary to malignant hematologic or inflammatory diseases, such as chronic myeloid leukemia, polycythemia vera, primary thrombocythemia, multiple myeloma, disseminated tuberculosis, or vasculitis. However, few cases of brucellosis-associated myelofibrosis have been reported. Moreover, due to the rarity of this phenomenon, it is often overlooked by clinicians, resulting in misdiagnosis and mismanagement. Thus, brucellosis should be considered as a possible cause of myelofibrosis. In the present study, we report five cases of brucellosis, of which three had myelofibrosis. In addition, to further determine the potential link between brucellosis and myelofibrosis, we retrospectively analyzed the levels of various cytokines by collecting the clinicopathologic data of patients and using immunohistochemical staining. We found that brucellosis patients with myelofibrosis had elevated levels of cytokines such as interferon (IFN)-γ, interleukin (IL)-1β, basic fibroblast growth factor (b-FGF), vascular endothelial growth factor (VEGF), suggesting that the regulation of cytokines may play a central role in the development of myelofibrosis in patients with brucellosis.
Collapse
Affiliation(s)
- Jun-Nuan Wang
- Hebei Medical UniversityShijiazhuang, Hebei, China
- Department of Hematology, Hebei General HospitalShijiazhuang, Hebei, China
| | - Bing-Jie Li
- Department of Pathology, Hebei General HospitalShijiazhuang, Hebei, China
| | - Jun Yuan
- Department of Hematology, Hebei General HospitalShijiazhuang, Hebei, China
| | - Yan Li
- Department of Hematology, Hebei General HospitalShijiazhuang, Hebei, China
| |
Collapse
|
4
|
Picoli CC, Martins PR, Wong XLC, Righi T, Guimarães PPG, Pinto MCX, Amorim JH, Azevedo VAC, Pereira SR, Kanashiro A, Cruz FC, Resende RR, Mintz A, Frenette PS, Birbrair A. Whole bone subcutaneous transplantation as a strategy to study precisely the bone marrow niche. Stem Cell Rev Rep 2022; 19:906-927. [PMID: 36585572 DOI: 10.1007/s12015-022-10496-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2022] [Indexed: 01/01/2023]
Abstract
Hematopoietic stem cells are maintained in a specialized microenvironment, known as the 'niche', within the bone marrow. Understanding the contribution of cellular and molecular components within the bone marrow niche for the maintenance of hematopoietic stem cells is crucial for the success of therapeutic applications. So far, the roles of crucial mechanisms within the bone marrow niche have been explored in transgenic animals in which genetic modifications are ubiquitously introduced in the whole body. The lack of precise tools to explore genetic alterations exclusively within the bone marrow prevents our determination of whether the observed outcomes result from confounding effects from other organs. Here, we developed a new method - 'whole bone subcutaneous transplantation'- to study the bone marrow niche in transgenic animals precisely. Using immunolabeling of CD45.1 (donor) vs. CD45.2 (recipient) hematopoeitic stem cells, we demonstrated that hematopoeitic stem cells from the host animals colonize the subcutaneously transplanted femurs after transplantation, while the hematopoietic stem cells from the donor disappear. Strikinlgy, the bone marrow niche of these subcutaneously transplanted femurs remain from the donor mice, enabling us to study specifically cells of the bone marrow niche using this model. We also showed that genetic ablation of peri-arteriolar cells specifically in donor femurs reduced the numbers of hematopoietic stem cells in these bones. This supports the use of this strategy as a model, in combination with genetic tools, to evaluate how bone marrow niche specific modifications may impact non-modified hematopoietic stem cells. Thus, this approach can be utilized for genetic manipulation in vivo of specific cell types only within the bone marrow. The combination of whole bone subcutaneous transplantation with rodent transgenic models will facilitate a more precise, complex and comprehensive understanding of existing problems in the study of the hematopoietic stem cell bone marrow niche.
Collapse
Affiliation(s)
- Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Xiao Lin Casey Wong
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA
| | - Thamires Righi
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA.,Department of Pharmacology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Pedro P G Guimarães
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro C X Pinto
- Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Jaime H Amorim
- Center of Biological Sciences and Health, Federal University of West Bahia, Barreiras, BA, Brazil
| | - Vasco A C Azevedo
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Alexandre Kanashiro
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA
| | - Fabio Cardoso Cruz
- Department of Pharmacology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Paul S Frenette
- Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, NY, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA. .,Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
5
|
Politis PK, Charonis AS. Calreticulin in renal fibrosis: A short review. J Cell Mol Med 2022; 26:5949-5954. [PMID: 36440574 PMCID: PMC9753439 DOI: 10.1111/jcmm.17627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/02/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022] Open
Abstract
Fibrosis is a common denominator of several pathological conditions. Over the last decade, Calreticulin has emerged as a critical player in the fibrotic processes in many tissues and organs. Here we review the recent advances in our understanding of the regulatory roles of Calreticulin in renal fibrosis. In particular, a proteomic screen that we performed more than 15 years ago, for the identification of novel components involved in the mechanisms of renal fibrosis, led to the observation that Calreticulin is associated with the initiation and progression of kidney fibrosis in a rodent model. We also showed that altered expression levels of Calreticulin in vitro and in vivo are significantly affecting the fibrotic phenotype in cellular systems and animal models, respectively. We also identified an upstream regulatory mechanism that mediates the transcriptional control of Calreticulin expression during the progression of renal fibrosis, by showing that the druggable orphan nuclear receptor NR5A2 and its SUMOylation is involved in this action. These data provide novel targets for future pharmacological interventions against fibrosis. In addition, further proteomic analysis uncovered a correlation between the up-regulation of Calreticulin and that of 14-3-3σ protein. Collectively, our previous observations suggest that Calreticulin is a central node in a regulatory axis that controls the initiation and progression of renal fibrosis.
Collapse
Affiliation(s)
- Panagiotis K. Politis
- Center for Basic ResearchBiomedical Research Foundation of the Academy of AthensAthensGreece
| | - Aristidis S. Charonis
- Center for Clinical, Experimental Surgery and Translational ResearchBiomedical Research Foundation of the Academy of AthensAthensGreece,University Research Institute of Maternal and Child Health and Precision MedicineAthensGreece
| |
Collapse
|
6
|
Bassan VL, Barretto GD, de Almeida FC, Palma PVB, Binelli LS, da Silva JPL, Fontanari C, Castro RC, de Figueiredo Pontes LL, Frantz FG, de Castro FA. Philadelphia-negative myeloproliferative neoplasms display alterations in monocyte subpopulations frequency and immunophenotype. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:223. [PMID: 36175590 PMCID: PMC9522456 DOI: 10.1007/s12032-022-01825-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/14/2022] [Indexed: 11/06/2022]
Abstract
Philadelphia-negative myeloproliferative neoplasms (MPN) are clonal hematological diseases associated with driver mutations in JAK2, CALR, and MPL genes. Moreover, several evidence suggests that chronic inflammation and alterations in stromal and immune cells may contribute to MPN’s pathophysiology. We evaluated the frequency and the immunophenotype of peripheral blood monocyte subpopulations in patients with polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (MF). Peripheral blood monocytes from PV (n = 16), ET (n = 16), and MF (n = 15) patients and healthy donors (n = 10) were isolated and submitted to immunophenotyping to determine the frequency of monocyte subpopulations and surface markers expression density. Plasma samples were used to measure the levels of soluble CD163, a biomarker of monocyte activity. PV, ET, and MF patients presented increased frequency of intermediate and non-classical monocytes and reduced frequency of classical monocytes compared to controls. Positivity for JAK2 mutation was significantly associated with the percentage of intermediate monocytes. PV, ET, and MF patients presented high-activated monocytes, evidenced by higher HLA-DR expression and increased soluble CD163 levels. The three MPN categories presented increased frequency of CD56+ aberrant monocytes, and PV and ET patients presented reduced frequency of CD80/86+ monocytes. Therefore, alterations in monocyte subpopulations frequency and surface markers expression pattern may contribute to oncoinflammation and may be associated with the pathophysiology of MPN.
Collapse
Affiliation(s)
- Vitor Leonardo Bassan
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/nº - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14040-903, Brazil.
| | - Gabriel Dessotti Barretto
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/nº - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14040-903, Brazil
| | - Felipe Campos de Almeida
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/nº - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14040-903, Brazil
| | - Patrícia Vianna Bonini Palma
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501 - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14051-260, Brazil
| | - Larissa Sarri Binelli
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501 - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14051-260, Brazil.,Department of Medical Images, Hematology and Clinical Oncology, University Hospital of the Ribeirão Preto Medical School, University of São Paulo, Rua Tenente Catão Roxo, 3900 - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14015-010, Brazil
| | - João Paulo Lettieri da Silva
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501 - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14051-260, Brazil.,Department of Medical Images, Hematology and Clinical Oncology, University Hospital of the Ribeirão Preto Medical School, University of São Paulo, Rua Tenente Catão Roxo, 3900 - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14015-010, Brazil
| | - Caroline Fontanari
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/nº - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14040-903, Brazil
| | - Ricardo Cardoso Castro
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/nº - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14040-903, Brazil
| | - Lorena Lôbo de Figueiredo Pontes
- Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501 - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14051-260, Brazil.,Department of Medical Images, Hematology and Clinical Oncology, University Hospital of the Ribeirão Preto Medical School, University of São Paulo, Rua Tenente Catão Roxo, 3900 - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14015-010, Brazil
| | - Fabiani Gai Frantz
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/nº - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14040-903, Brazil
| | - Fabíola Attié de Castro
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/nº - Vila Monte Alegre, Ribeirão Preto, São Paulo, 14040-903, Brazil
| |
Collapse
|
7
|
Inhibition of proinflammatory signaling impairs fibrosis of bone marrow mesenchymal stromal cells in myeloproliferative neoplasms. Exp Mol Med 2022; 54:273-284. [PMID: 35288649 PMCID: PMC8980093 DOI: 10.1038/s12276-022-00742-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 11/04/2021] [Accepted: 12/21/2021] [Indexed: 12/03/2022] Open
Abstract
Although bone marrow-derived mesenchymal stromal cells (BM-MSCs) have been identified as a major cellular source of fibrosis, the exact molecular mechanism and signaling pathways involved have not been identified thus far. Here, we show that BM-MSCs contribute to fibrosis in myeloproliferative neoplasms (MPNs) by differentiating into αSMA-positive myofibroblasts. These cells display a dysregulated extracellular matrix with increased FN1 production and secretion of profibrotic MMP9 compared to healthy donor cells. Fibrogenic TGFβ and inflammatory JAK2/STAT3 and NFκB signaling pathway activity is increased in BM-MSCs of MPN patients. Moreover, coculture with mononuclear cells from MPN patients was sufficient to induce fibrosis in healthy BM-MSCs. Inhibition of JAK1/2, SMAD3 or NFκB significantly reduced the fibrotic phenotype of MPN BM-MSCs and was able to prevent the development of fibrosis induced by coculture of healthy BM-MSCs and MPN mononuclear cells with overly active JAK/STAT signaling, underlining their involvement in fibrosis. Combined treatment with JAK1/2 and SMAD3 inhibitors showed synergistic and the most favorable effects on αSMA and FN1 expression in BM-MSCs. These results support the combined inhibition of TGFβ and inflammatory signaling to extenuate fibrosis in MPN. The treatment of fibrosis in patients with rare bone marrow disorders could be improved with a combined therapy that targets inflammatory pathways. Myeloproliferative neoplasms (MPN) are a group of bone marrow disorders characterized by the over-production of blood cells, which can lead to fibrosis in the bone marrow. Vladan Čokić at the University of Belgrade, Serbia, and co-workers examined how stem cells known as mesenchymal stromal cells from the bone marrow contribute to MPN fibrosis. They found an increase in three pro-inflammatory signaling pathways in MPN patients, resulting in the stromal cells differentiating into cells with dysregulated extracellular matrices. The differentiated cells did not behave correctly nor degrade properly, triggering fibrosis. The team combined two drugs that target the inflammatory signaling pathways, and successfully inhibited the development of fibrosis in MPN cell cultures.
Collapse
|
8
|
Zingariello M, Verachi P, Gobbo F, Martelli F, Falchi M, Mazzarini M, Valeri M, Sarli G, Marinaccio C, Melo-Cardenas J, Crispino JD, Migliaccio AR. Resident Self-Tissue of Proinflammatory Cytokines Rather than Their Systemic Levels Correlates with Development of Myelofibrosis in Gata1low Mice. Biomolecules 2022; 12:biom12020234. [PMID: 35204735 PMCID: PMC8961549 DOI: 10.3390/biom12020234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Abstract
Serum levels of inflammatory cytokines are currently investigated as prognosis markers in myelofibrosis, the most severe Philadelphia-negative myeloproliferative neoplasm. We tested this hypothesis in the Gata1low model of myelofibrosis. Gata1low mice, and age-matched wild-type littermates, were analyzed before and after disease onset. We assessed cytokine serum levels by Luminex-bead-assay and ELISA, frequency and cytokine content of stromal cells by flow cytometry, and immunohistochemistry and bone marrow (BM) localization of GFP-tagged hematopoietic stem cells (HSC) by confocal microscopy. Differences in serum levels of 32 inflammatory-cytokines between prefibrotic and fibrotic Gata1low mice and their wild-type littermates were modest. However, BM from fibrotic Gata1low mice contained higher levels of lipocalin-2, CXCL1, and TGF-β1 than wild-type BM. Although frequencies of endothelial cells, mesenchymal cells, osteoblasts, and megakaryocytes were higher than normal in Gata1low BM, the cells which expressed these cytokines the most were malignant megakaryocytes. This increased bioavailability of proinflammatory cytokines was associated with altered HSC localization: Gata1low HSC were localized in the femur diaphysis in areas surrounded by microvessels, neo-bones, and megakaryocytes, while wild-type HSC were localized in the femur epiphysis around adipocytes. In conclusion, bioavailability of inflammatory cytokines in BM, rather than blood levels, possibly by reshaping the HSC niche, correlates with myelofibrosis in Gata1low mice.
Collapse
Affiliation(s)
| | - Paola Verachi
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
| | - Francesca Gobbo
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Mario Falchi
- National Center HIV/AIDS Research, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Mazzarini
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
| | - Mauro Valeri
- Center for Animal Experimentation and Well-Being, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy;
| | | | - Johanna Melo-Cardenas
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.M.-C.); (J.D.C.)
| | - John D. Crispino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.M.-C.); (J.D.C.)
| | - Anna Rita Migliaccio
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
- Center for Integrated Biomedical Research, Campus Bio-Medico, 00128 Rome, Italy
- Correspondence:
| |
Collapse
|
9
|
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: 4.5] [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
|
10
|
Kale V. Granulocytes Negatively Regulate Secretion of Transforming Growth Factor β1 by Bone Marrow Mononuclear Cells via Secretion of Erythropoietin Receptors in the Milieu. Stem Cell Rev Rep 2021; 18:1408-1416. [PMID: 34775556 DOI: 10.1007/s12015-021-10292-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2021] [Indexed: 11/27/2022]
Abstract
In my previous study, I demonstrated that bone marrow-derived mononuclear cells (BM MNCs) secrete copious amounts of Transforming Growth Factor β1 (TGFβ1) in response to erythropoietin (EPO). In this study, I investigated the principal cell type involved in the process. I found that a large percentage of various marrow cells, but not their mature counterparts present in the peripheral blood, express EPO-receptors (EPO-R). Cell depletion experiments showed that depletion of Glycophorin positive erythroblasts and CD41+ megakaryocytes - the prime suspects - did not affect the EPO-mediated TGFβ1 secretion by the BM MNCs. However, individual depletion of CD2+ T lymphocytes, CD14+ monocyte/macrophages, and CD19+ B cells affected the TGFβ1 secretion by EPO-primed MNCs: depletion of CD2+ cells had the most striking effect. Unexpectedly, and most interestingly, depletion of CD15+ granulocytes led to a significant increase in the TGFβ1 secretion by both naïve and EPO-primed BM MNCs, suggesting that these cells negatively regulate the process. Mechanistically, I show that the CD15+ cells exert this regulatory effect via secretion of both full-length and soluble EPO-R in the milieu. Overall my results, for the first time, unravel an in-built regulatory mechanism prevailing in the BM microenvironment that regulates the secretion of TGFβ1 by controlling EPO-EPO-R interaction.My data could be relevant in understanding the pathophysiology of several conditions associated with deregulated production of TGFβ1 in the marrow compartment.
Collapse
Affiliation(s)
- Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis School of Biological Sciences, Symbiosis International University Symbiosis Knowledge park, Lavale, Pune, 412115, India.
- National Centre for Cell Science, Ganeshkhind, Pune, 411007, India.
| |
Collapse
|
11
|
Lucchesi A, Napolitano R, Bochicchio MT, Giordano G, Napolitano M. Platelets Contribution to Thrombin Generation in Philadelphia-Negative Myeloproliferative Neoplasms: The "Circulating Wound" Model. Int J Mol Sci 2021; 22:ijms222111343. [PMID: 34768772 PMCID: PMC8583863 DOI: 10.3390/ijms222111343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Current cytoreductive and antithrombotic strategies in MPNs are mostly based on cell counts and on patient's demographic and clinical history. Despite the numerous studies conducted on platelet function and on the role of plasma factors, an accurate and reliable method to dynamically quantify the hypercoagulability states of these conditions is not yet part of clinical practice. Starting from our experience, and after having sifted through the literature, we propose an in-depth narrative report on the contribution of the clonal platelets of MPNs-rich in tissue factor (TF)-in promoting a perpetual procoagulant mechanism. The whole process results in an unbalanced generation of thrombin and is self-maintained by Protease Activated Receptors (PARs). We chose to define this model as a "circulating wound", as it indisputably links the coagulation, inflammation, and fibrotic progression of the disease, in analogy with what happens in some solid tumours. The platelet contribution to thrombin generation results in triggering a vicious circle supported by the PARs/TGF-beta axis. PAR antagonists could therefore be a good option for target therapy, both to contain the risk of vascular events and to slow the progression of the disease towards end-stage forms. Both the new and old strategies, however, will require tools capable of measuring procoagulant or prohaemorrhagic states in a more extensive and dynamic way to favour a less empirical management of MPNs and their potential clinical complications.
Collapse
MESH Headings
- Animals
- Biological Assay
- Blood Platelets/metabolism
- Humans
- Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/drug therapy
- Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/metabolism
- Models, Biological
- Receptors, Fibrinogen/metabolism
- Thrombin/antagonists & inhibitors
- Thrombin/biosynthesis
- Thrombophilia/physiopathology
Collapse
Affiliation(s)
- Alessandro Lucchesi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Roberta Napolitano
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
- Correspondence:
| | - Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Giulio Giordano
- Internal Medicine Division, Hematology Service, Regional Hospital “A. Cardarelli”, 86100 Campobasso, Italy;
| | - Mariasanta Napolitano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties and Infectious Disease Unit, University Hospital “P. Giaccone”, 90127 Palermo, Italy;
| |
Collapse
|
12
|
Lambert J, Saliba J, Calderon C, Sii-Felice K, Salma M, Edmond V, Alvarez JC, Delord M, Marty C, Plo I, Kiladjian JJ, Soler E, Vainchenker W, Villeval JL, Rousselot P, Prost S. PPARγ agonists promote the resolution of myelofibrosis in preclinical models. J Clin Invest 2021; 131:136713. [PMID: 33914703 DOI: 10.1172/jci136713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
Myelofibrosis (MF) is a non-BCR-ABL myeloproliferative neoplasm associated with poor outcomes. Current treatment has little effect on the natural history of the disease. MF results from complex interactions between (a) the malignant clone, (b) an inflammatory context, and (c) remodeling of the bone marrow (BM) microenvironment. Each of these points is a potential target of PPARγ activation. Here, we demonstrated the therapeutic potential of PPARγ agonists in resolving MF in 3 mouse models. We showed that PPARγ agonists reduce myeloproliferation, modulate inflammation, and protect the BM stroma in vitro and ex vivo. Activation of PPARγ constitutes a relevant therapeutic target in MF, and our data support the possibility of using PPARγ agonists in clinical practice.
Collapse
Affiliation(s)
- Juliette Lambert
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Department of Hematology and Oncology, Centre Hospitalier de Versailles, Le Chesnay, France.,Opale Carnot Institute, Paris, France
| | - Joseph Saliba
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Carolina Calderon
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Opale Carnot Institute, Paris, France
| | - Karine Sii-Felice
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Mohammad Salma
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | - Valérie Edmond
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Jean-Claude Alvarez
- Département de Pharmacologie-Toxicologie, Hôpitaux Universitaires Paris Ile-de-France Ouest, AP-HP, Hôpital Raymond-Poincaré, FHU Sepsis, Garches, France.,MasSpecLab, Plateforme de spectrométrie de masse, INSERM U-1173, Université Paris-Saclay (Versailles Saint-Quentin-en-Yvelines), UFR des sciences de la santé, Montigny-le-Bretonneux, France
| | - Marc Delord
- Recherche Clinique, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Caroline Marty
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Jean-Jacques Kiladjian
- Opale Carnot Institute, Paris, France.,Université de Paris, AP-HP, Hôpital Saint-Louis, Centre d'Investigations Cliniques CIC 1427, INSERM, Paris, France
| | - Eric Soler
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | | | - Jean-Luc Villeval
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Philippe Rousselot
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Department of Hematology and Oncology, Centre Hospitalier de Versailles, Le Chesnay, France.,Opale Carnot Institute, Paris, France.,Université Paris-Saclay (Versailles Saint-Quentin-en-Yvelines), UFR des sciences de la santé, Montigny-le-Bretonneux, France
| | - Stéphane Prost
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Opale Carnot Institute, Paris, France
| |
Collapse
|
13
|
Renal Diseases Associated with Hematologic Malignancies and Thymoma in the Absence of Renal Monoclonal Immunoglobulin Deposits. Diagnostics (Basel) 2021; 11:diagnostics11040710. [PMID: 33921123 PMCID: PMC8071536 DOI: 10.3390/diagnostics11040710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/02/2021] [Accepted: 04/13/2021] [Indexed: 12/05/2022] Open
Abstract
In addition to kidney diseases characterized by the precipitation and deposition of overproduced monoclonal immunoglobulin and kidney damage due to chemotherapy agents, a broad spectrum of renal lesions may be found in patients with hematologic malignancies. Glomerular diseases, in the form of paraneoplastic glomerulopathies and acute kidney injury with various degrees of proteinuria due to specific lymphomatous interstitial and/or glomerular infiltration, are two major renal complications observed in the lymphoid disorder setting. However, other hematologic neoplasms, including chronic lymphocytic leukemia, thymoma, myeloproliferative disorders, Castleman disease and hemophagocytic syndrome, have also been associated with the development of kidney lesions. These renal disorders require prompt recognition by the clinician, due to the need to implement specific treatment, depending on the chemotherapy regimen, to decrease the risk of subsequent chronic kidney disease. In the context of renal disease related to hematologic malignancies, renal biopsy remains crucial for accurate pathological diagnosis, with the aim of optimizing medical care for these patients. In this review, we provide an update on the epidemiology, clinical presentation, pathophysiological processes and diagnostic strategy for kidney diseases associated with hematologic malignancies outside the spectrum of monoclonal gammopathy of renal significance.
Collapse
|
14
|
Leguit RJ, Raymakers RAP, Hebeda KM, Goldschmeding R. CCN2 (Cellular Communication Network factor 2) in the bone marrow microenvironment, normal and malignant hematopoiesis. J Cell Commun Signal 2021; 15:25-56. [PMID: 33428075 PMCID: PMC7798015 DOI: 10.1007/s12079-020-00602-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023] Open
Abstract
CCN2, formerly termed Connective Tissue Growth Factor, is a protein belonging to the Cellular Communication Network (CCN)-family of secreted extracellular matrix-associated proteins. As a matricellular protein it is mainly considered to be active as a modifier of signaling activity of several different signaling pathways and as an orchestrator of their cross-talk. Furthermore, CCN2 and its fragments have been implicated in the regulation of a multitude of biological processes, including cell proliferation, differentiation, adhesion, migration, cell survival, apoptosis and the production of extracellular matrix products, as well as in more complex processes such as embryonic development, angiogenesis, chondrogenesis, osteogenesis, fibrosis, mechanotransduction and inflammation. Its function is complex and context dependent, depending on cell type, state of differentiation and microenvironmental context. CCN2 plays a role in many diseases, especially those associated with fibrosis, but has also been implicated in many different forms of cancer. In the bone marrow (BM), CCN2 is highly expressed in mesenchymal stem/stromal cells (MSCs). CCN2 is important for MSC function, supporting its proliferation, migration and differentiation. In addition, stromal CCN2 supports the maintenance and longtime survival of hematopoietic stem cells, and in the presence of interleukin 7, stimulates the differentiation of pro-B lymphocytes into pre-B lymphocytes. Overexpression of CCN2 is seen in the majority of B-acute lymphoblastic leukemias, especially in certain cytogenetic subgroups associated with poor outcome. In acute myeloid leukemia, CCN2 expression is increased in MSCs, which has been associated with leukemic engraftment in vivo. In this review, the complex function of CCN2 in the BM microenvironment and in normal as well as malignant hematopoiesis is discussed. In addition, an overview is given of data on the remaining CCN family members regarding normal and malignant hematopoiesis, having many similarities and some differences in their function.
Collapse
Affiliation(s)
- Roos J. Leguit
- Department of Pathology, University Medical Center Utrecht, H04-312, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - Reinier A. P. Raymakers
- Department of Hematology, UMCU Cancer Center, Heidelberglaan 100 B02.226, 3584 CX Utrecht, The Netherlands
| | - Konnie M. Hebeda
- Department of Pathology, Radboud University Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Centre Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
15
|
Nasillo V, Riva G, Paolini A, Forghieri F, Roncati L, Lusenti B, Maccaferri M, Messerotti A, Pioli V, Gilioli A, Bettelli F, Giusti D, Barozzi P, Lagreca I, Maffei R, Marasca R, Potenza L, Comoli P, Manfredini R, Maiorana A, Tagliafico E, Luppi M, Trenti T. Inflammatory Microenvironment and Specific T Cells in Myeloproliferative Neoplasms: Immunopathogenesis and Novel Immunotherapies. Int J Mol Sci 2021; 22:ijms22041906. [PMID: 33672997 PMCID: PMC7918142 DOI: 10.3390/ijms22041906] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
The Philadelphia-negative myeloproliferative neoplasms (MPNs) are malignancies of the hematopoietic stem cell (HSC) arising as a consequence of clonal proliferation driven by somatically acquired driver mutations in discrete genes (JAK2, CALR, MPL). In recent years, along with the advances in molecular characterization, the role of immune dysregulation has been achieving increasing relevance in the pathogenesis and evolution of MPNs. In particular, a growing number of studies have shown that MPNs are often associated with detrimental cytokine milieu, expansion of the monocyte/macrophage compartment and myeloid-derived suppressor cells, as well as altered functions of T cells, dendritic cells and NK cells. Moreover, akin to solid tumors and other hematological malignancies, MPNs are able to evade T cell immune surveillance by engaging the PD-1/PD-L1 axis, whose pharmacological blockade with checkpoint inhibitors can successfully restore effective antitumor responses. A further interesting cue is provided by the recent discovery of the high immunogenic potential of JAK2V617F and CALR exon 9 mutations, that could be harnessed as intriguing targets for innovative adoptive immunotherapies. This review focuses on the recent insights in the immunological dysfunctions contributing to the pathogenesis of MPNs and outlines the potential impact of related immunotherapeutic approaches.
Collapse
Affiliation(s)
- Vincenzo Nasillo
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy; (G.R.); (B.L.); (E.T.); (T.T.)
- Correspondence: ; Tel.: +39-059-422-2173
| | - Giovanni Riva
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy; (G.R.); (B.L.); (E.T.); (T.T.)
| | - Ambra Paolini
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Fabio Forghieri
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Luca Roncati
- Institute of Pathology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (L.R.); (A.M.)
| | - Beatrice Lusenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy; (G.R.); (B.L.); (E.T.); (T.T.)
| | - Monica Maccaferri
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Andrea Messerotti
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Valeria Pioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Andrea Gilioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Francesca Bettelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Davide Giusti
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Patrizia Barozzi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Ivana Lagreca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Rossana Maffei
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Roberto Marasca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Leonardo Potenza
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Patrizia Comoli
- Pediatric Hematology/Oncology Unit and Cell Factory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, 27100 Pavia, Italy;
| | - Rossella Manfredini
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Antonino Maiorana
- Institute of Pathology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (L.R.); (A.M.)
| | - Enrico Tagliafico
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy; (G.R.); (B.L.); (E.T.); (T.T.)
| | - Mario Luppi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy; (G.R.); (B.L.); (E.T.); (T.T.)
| |
Collapse
|
16
|
Nasillo V, Riva G, Paolini A, Forghieri F, Roncati L, Lusenti B, Maccaferri M, Messerotti A, Pioli V, Gilioli A, Bettelli F, Giusti D, Barozzi P, Lagreca I, Maffei R, Marasca R, Potenza L, Comoli P, Manfredini R, Maiorana A, Tagliafico E, Luppi M, Trenti T. Inflammatory Microenvironment and Specific T Cells in Myeloproliferative Neoplasms: Immunopathogenesis and Novel Immunotherapies. Int J Mol Sci 2021. [PMID: 33672997 DOI: 10.3390/ijms22041906.pmid:33672997;pmcid:pmc7918142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
The Philadelphia-negative myeloproliferative neoplasms (MPNs) are malignancies of the hematopoietic stem cell (HSC) arising as a consequence of clonal proliferation driven by somatically acquired driver mutations in discrete genes (JAK2, CALR, MPL). In recent years, along with the advances in molecular characterization, the role of immune dysregulation has been achieving increasing relevance in the pathogenesis and evolution of MPNs. In particular, a growing number of studies have shown that MPNs are often associated with detrimental cytokine milieu, expansion of the monocyte/macrophage compartment and myeloid-derived suppressor cells, as well as altered functions of T cells, dendritic cells and NK cells. Moreover, akin to solid tumors and other hematological malignancies, MPNs are able to evade T cell immune surveillance by engaging the PD-1/PD-L1 axis, whose pharmacological blockade with checkpoint inhibitors can successfully restore effective antitumor responses. A further interesting cue is provided by the recent discovery of the high immunogenic potential of JAK2V617F and CALR exon 9 mutations, that could be harnessed as intriguing targets for innovative adoptive immunotherapies. This review focuses on the recent insights in the immunological dysfunctions contributing to the pathogenesis of MPNs and outlines the potential impact of related immunotherapeutic approaches.
Collapse
Affiliation(s)
- Vincenzo Nasillo
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy
| | - Giovanni Riva
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy
| | - Ambra Paolini
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Fabio Forghieri
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Luca Roncati
- Institute of Pathology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Beatrice Lusenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy
| | - Monica Maccaferri
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Andrea Messerotti
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Valeria Pioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Andrea Gilioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Francesca Bettelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Davide Giusti
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Patrizia Barozzi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Ivana Lagreca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Rossana Maffei
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Roberto Marasca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Leonardo Potenza
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Patrizia Comoli
- Pediatric Hematology/Oncology Unit and Cell Factory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, 27100 Pavia, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine "S. Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Antonino Maiorana
- Institute of Pathology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Enrico Tagliafico
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy
| | - Mario Luppi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy
| |
Collapse
|
17
|
Yung Y, Lee E, Chu HT, Yip PK, Gill H. Targeting Abnormal Hematopoietic Stem Cells in Chronic Myeloid Leukemia and Philadelphia Chromosome-Negative Classical Myeloproliferative Neoplasms. Int J Mol Sci 2021; 22:ijms22020659. [PMID: 33440869 PMCID: PMC7827471 DOI: 10.3390/ijms22020659] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 02/02/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are unique hematopoietic stem cell disorders sharing mutations that constitutively activate the signal-transduction pathways involved in haematopoiesis. They are characterized by stem cell-derived clonal myeloproliferation. The key MPNs comprise chronic myeloid leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). CML is defined by the presence of the Philadelphia (Ph) chromosome and BCR-ABL1 fusion gene. Despite effective cytoreductive agents and targeted therapy, complete CML/MPN stem cell eradication is rarely achieved. In this review article, we discuss the novel agents and combination therapy that can potentially abnormal hematopoietic stem cells in CML and MPNs and the CML/MPN stem cell-sustaining bone marrow microenvironment.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Autophagy
- Biomarkers, Tumor
- Cell Survival/drug effects
- Cell Transformation, Neoplastic/genetics
- Combined Modality Therapy
- Disease Susceptibility
- Genetic Predisposition to Disease
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Molecular Targeted Therapy
- Myeloproliferative Disorders/etiology
- Myeloproliferative Disorders/pathology
- Myeloproliferative Disorders/therapy
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Philadelphia Chromosome
- Signal Transduction/drug effects
- Stem Cell Niche
- Tumor Microenvironment
Collapse
Affiliation(s)
| | | | | | | | - Harinder Gill
- Correspondence: ; Tel.: +852-2255-4542; Fax: +852-2816-2863
| |
Collapse
|
18
|
Ambrosi TH, Chan CKF. Skeletal Stem Cells as the Developmental Origin of Cellular Niches for Hematopoietic Stem and Progenitor Cells. Curr Top Microbiol Immunol 2021; 434:1-31. [PMID: 34850280 PMCID: PMC8864730 DOI: 10.1007/978-3-030-86016-5_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The skeletal system is a highly complex network of mesenchymal, hematopoietic, and vasculogenic stem cell lineages that coordinate the development and maintenance of defined microenvironments, so-called niches. Technological advancements in recent years have allowed for the dissection of crucial cell types as well as their autocrine and paracrine signals that regulate these niches during development, homeostasis, regeneration, and disease. Ingress of blood vessels and bone marrow hematopoiesis are initiated by skeletal stem cells (SSCs) and their more committed downstream lineage cell types that direct shape and form of skeletal elements. In this chapter, we focus on the role of SSCs as the developmental origin of niches for hematopoietic stem and progenitor cells. We discuss latest updates in the definition of SSCs, cellular processes establishing and maintaining niches, as well as alterations of stem cell microenvironments promoting malignancies. We conclude with an outlook on future studies that could take advantage of SSC-niche engineering as a basis for the development of new therapeutic tools to not only treat bone-related diseases but also maladies stemming from derailed niche dynamics altering hematopoietic output.
Collapse
Affiliation(s)
- Thomas H Ambrosi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Charles K F Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
19
|
Bose P, Masarova L, Verstovsek S. Novel Concepts of Treatment for Patients with Myelofibrosis and Related Neoplasms. Cancers (Basel) 2020; 12:cancers12102891. [PMID: 33050168 PMCID: PMC7599937 DOI: 10.3390/cancers12102891] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Myelofibrosis (MF) is an advanced form of a group of rare, related bone marrow cancers termed myeloproliferative neoplasms (MPNs). Some patients develop myelofibrosis from the outset, while in others, it occurs as a complication of the more indolent MPNs, polycythemia vera (PV) or essential thrombocythemia (ET). Patients with PV or ET who require drug treatment are typically treated with the chemotherapy drug hydroxyurea, while in MF, the targeted therapies termed Janus kinase (JAK) inhibitors form the mainstay of treatment. However, these and other drugs (e.g., interferons) have important limitations. No drug has been shown to reliably prevent the progression of PV or ET to MF or transformation of MPNs to acute myeloid leukemia. In PV, it is not conclusively known if JAK inhibitors reduce the risk of blood clots, and in MF, these drugs do not improve low blood counts. New approaches to treating MF and related MPNs are, therefore, necessary. Abstract Janus kinase (JAK) inhibition forms the cornerstone of the treatment of myelofibrosis (MF), and the JAK inhibitor ruxolitinib is often used as a second-line agent in patients with polycythemia vera (PV) who fail hydroxyurea (HU). In addition, ruxolitinib continues to be studied in patients with essential thrombocythemia (ET). The benefits of JAK inhibition in terms of splenomegaly and symptoms in patients with MF are undeniable, and ruxolitinib prolongs the survival of persons with higher risk MF. Despite this, however, “disease-modifying” effects of JAK inhibitors in MF, i.e., bone marrow fibrosis and mutant allele burden reduction, are limited. Similarly, in HU-resistant/intolerant PV, while ruxolitinib provides excellent control of the hematocrit, symptoms and splenomegaly, reduction in the rate of thromboembolic events has not been convincingly demonstrated. Furthermore, JAK inhibitors do not prevent disease evolution to MF or acute myeloid leukemia (AML). Frontline cytoreductive therapy for PV generally comprises HU and interferons, which have their own limitations. Numerous novel agents, representing diverse mechanisms of action, are in development for the treatment of these three classic myeloproliferative neoplasms (MPNs). JAK inhibitor-based combinations, all of which are currently under study for MF, have been covered elsewhere in this issue. In this article, we focus on agents that have been studied as monotherapy in patients with MF, generally after JAK inhibitor resistance/intolerance, as well as several novel compounds in development for PV/ET.
Collapse
|
20
|
Saikia P, Crabb JS, Dibbin LL, Juszczak MJ, Willard B, Jang GF, Shiju TM, Crabb JW, Wilson SE. Quantitative proteomic comparison of myofibroblasts derived from bone marrow and cornea. Sci Rep 2020; 10:16717. [PMID: 33028893 PMCID: PMC7541534 DOI: 10.1038/s41598-020-73686-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Myofibroblasts are fibroblastic cells that function in wound healing, tissue repair and fibrosis, and arise from bone marrow (BM)-derived fibrocytes and a variety of local progenitor cells. In the cornea, myofibroblasts are derived primarily from stromal keratocytes and from BM-derived fibrocytes after epithelial-stromal and endothelial-stromal injuries. Quantitative proteomic comparison of mature alpha-smooth muscle actin (α-SMA)+ myofibroblasts (verified by immunocytochemistry for vimentin, α-SMA, desmin, and vinculin) generated from rabbit corneal fibroblasts treated with transforming growth factor (TGF) beta-1 or generated directly from cultured BM treated with TGF beta-1 was pursued for insights into possible functional differences. Paired cornea-derived and BM-derived α-SMA+ myofibroblast primary cultures were generated from four New Zealand white rabbits and confirmed to be myofibroblasts by immunocytochemistry. Paired cornea- and BM-derived myofibroblast specimens from each rabbit were analyzed by LC MS/MS iTRAQ technology using an Orbitrap Fusion Lumos Tribrid mass spectrometer, the Mascot search engine, the weighted average quantification method and the UniProt rabbit and human databases. From 2329 proteins quantified with ≥ 2 unique peptides from ≥ 3 rabbits, a total of 673 differentially expressed (DE) proteins were identified. Bioinformatic analysis of DE proteins with Ingenuity Pathway Analysis implicate progenitor-dependent functional differences in myofibroblasts that could impact tissue development. Our results suggest BM-derived myofibroblasts may be more prone to the formation of excessive cellular and extracellular material that are characteristic of fibrosis.
Collapse
Affiliation(s)
- Paramananda Saikia
- Cole Eye Institute, I-32, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| | - Jack S Crabb
- Cole Eye Institute, I-32, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44195, USA
- Lerner Research Institute, Cleveland, OH, 44195, USA
| | - Luciana L Dibbin
- Cole Eye Institute, I-32, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| | - Madison J Juszczak
- Cole Eye Institute, I-32, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| | | | - Geeng-Fu Jang
- Cole Eye Institute, I-32, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44195, USA
- Lerner Research Institute, Cleveland, OH, 44195, USA
| | - Thomas Michael Shiju
- Cole Eye Institute, I-32, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| | - John W Crabb
- Cole Eye Institute, I-32, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44195, USA.
- Lerner Research Institute, Cleveland, OH, 44195, USA.
- Cleveland Clinic, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA.
| | - Steven E Wilson
- Cole Eye Institute, I-32, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44195, USA.
- Cleveland Clinic, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA.
| |
Collapse
|
21
|
Teodorescu P, Pasca S, Jurj A, Gafencu G, Joelsson JP, Selicean S, Moldovan C, Munteanu R, Onaciu A, Tigu AB, Buse M, Zimta AA, Stiufiuc R, Petrushev B, Desmirean M, Dima D, Vlad C, Bergthorsson JT, Berce C, Ciurea S, Ghiaur G, Tomuleasa C. Transforming growth factor β-mediated micromechanics modulates disease progression in primary myelofibrosis. J Cell Mol Med 2020; 24:11100-11110. [PMID: 32889753 PMCID: PMC7576271 DOI: 10.1111/jcmm.15526] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Primary myelofibrosis (PMF) is a Ph‐negative myeloproliferative neoplasm (MPN), characterized by advanced bone marrow fibrosis and extramedullary haematopoiesis. The bone marrow fibrosis results from excessive proliferation of fibroblasts that are influenced by several cytokines in the microenvironment, of which transforming growth factor‐β (TGF‐β) is the most important. Micromechanics related to the niche has not yet been elucidated. In this study, we hypothesized that mechanical stress modulates TGF‐β signalling leading to further activation and subsequent proliferation and invasion of bone marrow fibroblasts, thus showing the important role of micromechanics in the development and progression of PMF, both in the bone marrow and in extramedullary sites. Using three PMF‐derived fibroblast cell lines and transforming growth factor‐β receptor (TGFBR) 1 and 2 knock‐down PMF‐derived fibroblasts, we showed that mechanical stress does stimulate the collagen synthesis by the fibroblasts in patients with myelofibrosis, through the TGFBR1, which however seems to be activated through alternative pathways, other than TGFBR2.
Collapse
Affiliation(s)
- Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania
| | - Sergiu Pasca
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Grigore Gafencu
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Molecular Haematology Unit - Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Jon-Petur Joelsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavík, Iceland.,Department of Laboratory Hematology, Landspitali University Hospital, Reykjavík, Iceland
| | - Sonia Selicean
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Graduate School for Cellular and Biomedical Sciences, Universität Bern, Bern, Switzerland
| | - Cristian Moldovan
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Anca Onaciu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Adrian-Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Mihail Buse
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Alina-Andreea Zimta
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Rares Stiufiuc
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Bobe Petrushev
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Minodora Desmirean
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Pathology, Constantin Papilian Military Hospital, Cluj Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Research Center, Cluj Napoca, Romania
| | - Cristina Vlad
- Department of Cardiology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Cardiology, Rehabilitation Hospital, Cluj Napoca, Romania
| | - Jon Thor Bergthorsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavík, Iceland.,Department of Laboratory Hematology, Landspitali, University Hospital, Reykjavík, Iceland
| | - Cristian Berce
- Animal Facility, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Stefan Ciurea
- Department of Cellular Therapies and Stem Cell Transplantation, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel Ghiaur
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.,Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| |
Collapse
|
22
|
Kuykendall AT, Horvat NP, Pandey G, Komrokji R, Reuther GW. Finding a Jill for JAK: Assessing Past, Present, and Future JAK Inhibitor Combination Approaches in Myelofibrosis. Cancers (Basel) 2020; 12:E2278. [PMID: 32823910 PMCID: PMC7464183 DOI: 10.3390/cancers12082278] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/11/2022] Open
Abstract
Myelofibrosis (MF) is a myeloproliferative neoplasm hallmarked by the upregulation of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway with associated extramedullary hematopoiesis and a high burden of disease-related symptoms. While JAK inhibitor therapy is central to the management of MF, it is not without limitations. In an effort to improve treatment for MF patients, there have been significant efforts to identify combination strategies that build upon the substantial benefits of JAK inhibition. Early efforts to combine agents with additive therapeutic profiles have given way to rationally designed combinations hoping to demonstrate clinical synergism and modify the underlying disease. In this article, we review the preclinical basis and existing clinical data for JAK inhibitor combination strategies while highlighting emerging strategies of particular interest.
Collapse
Affiliation(s)
- Andrew T. Kuykendall
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Nathan P. Horvat
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612 USA;
| | - Garima Pandey
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (G.P.); (G.W.R.)
| | - Rami Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Gary W. Reuther
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (G.P.); (G.W.R.)
| |
Collapse
|
23
|
Longhitano L, Li Volti G, Giallongo C, Spampinato M, Barbagallo I, Di Rosa M, Romano A, Avola R, Tibullo D, Palumbo GA. The Role of Inflammation and Inflammasome in Myeloproliferative Disease. J Clin Med 2020; 9:E2334. [PMID: 32707883 PMCID: PMC7464195 DOI: 10.3390/jcm9082334] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/01/2020] [Accepted: 07/17/2020] [Indexed: 12/22/2022] Open
Abstract
Polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF) are rare hematological conditions known as myeloproliferative neoplasms (MPNs). They are characterized for being BCR-ABL negative malignancies and affected patients often present with symptoms which can significantly impact their quality of life. MPNs are characterized by a clonal proliferation of an abnormal hematopoietic stem/progenitor cell. In MPNs; cells of all myeloid lineages; including those involved in the immune and inflammatory response; may belong to the malignant clone thus leading to an altered immune response and an overexpression of cytokines and inflammatory receptors; further worsening chronic inflammation. Many of these cytokines; in particular, IL-1β and IL-18; are released in active form by activating the inflammasome complexes which in turn mediate the inflammatory process. Despite this; little is known about the functional effects of stem cell-driven inflammasome signaling in MPN pathogenesis. In this review we focused on the role of inflammatory pathway and inflammasome in MPN diseases. A better understanding of the inflammatory-state-driving MPNs and of the role of the inflammasome may provide new insights on possible therapeutic strategies.
Collapse
Affiliation(s)
- Lucia Longhitano
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (L.L.); (G.L.V.); (M.S.); (R.A.)
| | - Giovanni Li Volti
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (L.L.); (G.L.V.); (M.S.); (R.A.)
| | - Cesarina Giallongo
- Department of Scienze Mediche Chirurgiche e Tecnologie Avanzate “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | - Mariarita Spampinato
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (L.L.); (G.L.V.); (M.S.); (R.A.)
| | - Ignazio Barbagallo
- Section of Biochemistry, Department of Drug Sciences, University of Catania, 95123 Catania, Italy;
| | - Michelino Di Rosa
- Section of Human Anatomy, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Alessandra Romano
- Division of Hematology, Department of General Surgery and Medical-Surgical Specialties, A.O.U. “Policlinico-Vittorio Emanuele”, University of Catania, 95123 Catania, Italy;
| | - Roberto Avola
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (L.L.); (G.L.V.); (M.S.); (R.A.)
| | - Daniele Tibullo
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (L.L.); (G.L.V.); (M.S.); (R.A.)
| | - Giuseppe Alberto Palumbo
- Department of Scienze Mediche Chirurgiche e Tecnologie Avanzate “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| |
Collapse
|
24
|
Smith JNP, Witkin MD, Jogasuria AP, Christo KF, Raffay TM, Markowitz SD, Desai AB. Therapeutic targeting of 15-PGDH in murine pulmonary fibrosis. Sci Rep 2020; 10:11657. [PMID: 32669620 PMCID: PMC7363833 DOI: 10.1038/s41598-020-68336-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 06/23/2020] [Indexed: 12/31/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterized by interstitial remodeling and pulmonary dysfunction. The etiology of IPF is not completely understood but involves pathologic inflammation and subsequent failure to resolve fibrosis in response to epithelial injury. Treatments for IPF are limited to anti-inflammatory and immunomodulatory agents, which are only partially effective. Prostaglandin E2 (PGE2) disrupts TGFβ signaling and suppresses myofibroblast differentiation, however practical strategies to raise tissue PGE2 during IPF have been limited. We previously described the discovery of a small molecule, (+)SW033291, that binds with high affinity to the PGE2-degrading enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH) and increases PGE2 levels. Here we evaluated pulmonary 15-PGDH expression and activity and tested whether pharmacologic 15-PGDH inhibition (PGDHi) is protective in a mouse model of bleomycin-induced pulmonary fibrosis (PF). Long-term PGDHi was well-tolerated, reduced the severity of pulmonary fibrotic lesions and extracellular matrix remodeling, and improved pulmonary function in bleomycin-treated mice. Moreover, PGDHi attenuated both acute inflammation and weight loss, and decreased mortality. Endothelial cells and macrophages are likely targets as these cell types highly expressed 15-PGDH. In conclusion, PGDHi ameliorates inflammatory pathology and fibrosis in murine PF, and may have clinical utility to treat human disease.
Collapse
Affiliation(s)
- Julianne N P Smith
- Department of Medicine, and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Matthew D Witkin
- Department of Medicine, and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Alvin P Jogasuria
- Department of Medicine, and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Kelsey F Christo
- Department of Medicine, and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Thomas M Raffay
- Department of Medicine, and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sanford D Markowitz
- Department of Medicine, and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA. .,University Hospitals Seidman Cancer Center, Cleveland, OH, 44106, USA.
| | - Amar B Desai
- Department of Medicine, and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
25
|
Øbro NF, Grinfeld J, Belmonte M, Irvine M, Shepherd MS, Rao TN, Karow A, Riedel LM, Harris OB, Baxter EJ, Nangalia J, Godfrey A, Harrison CN, Li J, Skoda RC, Campbell PJ, Green AR, Kent DG. Longitudinal Cytokine Profiling Identifies GRO-α and EGF as Potential Biomarkers of Disease Progression in Essential Thrombocythemia. Hemasphere 2020; 4:e371. [PMID: 32647796 PMCID: PMC7306314 DOI: 10.1097/hs9.0000000000000371] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are characterized by deregulation of mature blood cell production and increased risk of myelofibrosis (MF) and leukemic transformation. Numerous driver mutations have been identified but substantial disease heterogeneity remains unexplained, implying the involvement of additional as yet unidentified factors. The inflammatory microenvironment has recently attracted attention as a crucial factor in MPN biology, in particular whether inflammatory cytokines and chemokines contribute to disease establishment or progression. Here we present a large-scale study of serum cytokine profiles in more than 400 MPN patients and identify an essential thrombocythemia (ET)-specific inflammatory cytokine signature consisting of Eotaxin, GRO-α, and EGF. Levels of 2 of these markers (GRO-α and EGF) in ET patients were associated with disease transformation in initial sample collection (GRO-α) or longitudinal sampling (EGF). In ET patients with extensive genomic profiling data (n = 183) cytokine levels added significant prognostic value for predicting transformation from ET to MF. Furthermore, CD56+CD14+ pro-inflammatory monocytes were identified as a novel source of increased GRO-α levels. These data implicate the immune cell microenvironment as a significant player in ET disease evolution and illustrate the utility of cytokines as potential biomarkers for reaching beyond genomic classification for disease stratification and monitoring.
Collapse
Affiliation(s)
- Nina F. Øbro
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - Jacob Grinfeld
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Miriam Belmonte
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5NG, United Kingdom
| | - Melissa Irvine
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - Mairi S. Shepherd
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - Tata Nageswara Rao
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Axel Karow
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lisa M. Riedel
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - Oliva B. Harris
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - E. Joanna Baxter
- Department of Hematology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Jyoti Nangalia
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Anna Godfrey
- Department of Hematology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Claire N. Harrison
- Department of Hematology, Guy's and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Juan Li
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - Radek C. Skoda
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Peter J. Campbell
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Anthony R. Green
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - David G. Kent
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5NG, United Kingdom
| |
Collapse
|
26
|
Neoplastic fibrocytes play an essential role in bone marrow fibrosis in Jak2V617F-induced primary myelofibrosis mice. Leukemia 2020; 35:454-467. [PMID: 32472085 PMCID: PMC7862060 DOI: 10.1038/s41375-020-0880-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/07/2020] [Accepted: 05/18/2020] [Indexed: 11/23/2022]
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) characterized by clonal myeloproliferation, progressive bone marrow (BM) fibrosis, splenomegaly, and anemia. BM fibrosis was previously thought to be a reactive phenomenon induced by mesenchymal stromal cells that are stimulated by the overproduction of cytokines such as transforming growth factor (TGF)-β1. However, the involvement of neoplastic fibrocytes in BM fibrosis was recently reported. In this study, we showed that the vast majority of collagen- and fibronectin-producing cells in the BM and spleens of Jak2V617F-induced myelofibrosis (MF) mice were fibrocytes derived from neoplastic hematopoietic cells. Neoplastic monocyte depletion eliminated collagen- and fibronectin-producing fibrocytes in BM and spleen, and ameliorated most characteristic MF features in Jak2V617F transgenic mice, including BM fibrosis, anemia, and splenomegaly, while had little effect on the elevated numbers of megakaryocytes and stem cells in BM, and leukothrombocytosis in peripheral blood. TGF-β1, which was produced by hematopoietic cells including fibrocytes, promoted the differentiation of neoplastic monocytes to fibrocytes, and elevated plasma TGF-β1 levels were normalized by monocyte depletion. Collectively, our data suggest that neoplastic fibrocytes are the major contributor to BM fibrosis in PMF, and TGF-β1 is required for their differentiation.
Collapse
|
27
|
Bharadwaj U, Kasembeli MM, Robinson P, Tweardy DJ. Targeting Janus Kinases and Signal Transducer and Activator of Transcription 3 to Treat Inflammation, Fibrosis, and Cancer: Rationale, Progress, and Caution. Pharmacol Rev 2020; 72:486-526. [PMID: 32198236 PMCID: PMC7300325 DOI: 10.1124/pr.119.018440] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Before it was molecularly cloned in 1994, acute-phase response factor or signal transducer and activator of transcription (STAT)3 was the focus of intense research into understanding the mammalian response to injury, particularly the acute-phase response. Although known to be essential for liver production of acute-phase reactant proteins, many of which augment innate immune responses, molecular cloning of acute-phase response factor or STAT3 and the research this enabled helped establish the central function of Janus kinase (JAK) family members in cytokine signaling and identified a multitude of cytokines and peptide hormones, beyond interleukin-6 and its family members, that activate JAKs and STAT3, as well as numerous new programs that their activation drives. Many, like the acute-phase response, are adaptive, whereas several are maladaptive and lead to chronic inflammation and adverse consequences, such as cachexia, fibrosis, organ dysfunction, and cancer. Molecular cloning of STAT3 also enabled the identification of other noncanonical roles for STAT3 in normal physiology, including its contribution to the function of the electron transport chain and oxidative phosphorylation, its basal and stress-related adaptive functions in mitochondria, its function as a scaffold in inflammation-enhanced platelet activation, and its contributions to endothelial permeability and calcium efflux from endoplasmic reticulum. In this review, we will summarize the molecular and cellular biology of JAK/STAT3 signaling and its functions under basal and stress conditions, which are adaptive, and then review maladaptive JAK/STAT3 signaling in animals and humans that lead to disease, as well as recent attempts to modulate them to treat these diseases. In addition, we will discuss how consideration of the noncanonical and stress-related functions of STAT3 cannot be ignored in efforts to target the canonical functions of STAT3, if the goal is to develop drugs that are not only effective but safe. SIGNIFICANCE STATEMENT: Key biological functions of Janus kinase (JAK)/signal transducer and activator of transcription (STAT)3 signaling can be delineated into two broad categories: those essential for normal cell and organ development and those activated in response to stress that are adaptive. Persistent or dysregulated JAK/STAT3 signaling, however, is maladaptive and contributes to many diseases, including diseases characterized by chronic inflammation and fibrosis, and cancer. A comprehensive understanding of JAK/STAT3 signaling in normal development, and in adaptive and maladaptive responses to stress, is essential for the continued development of safe and effective therapies that target this signaling pathway.
Collapse
Affiliation(s)
- Uddalak Bharadwaj
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Moses M Kasembeli
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Prema Robinson
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - David J Tweardy
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine (U.B., M.M.K., P.R., D.J.T.), and Department of Molecular and Cellular Oncology (D.J.T.), University of Texas, MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
28
|
Gene expression profiling distinguishes prefibrotic from overtly fibrotic myeloproliferative neoplasms and identifies disease subsets with distinct inflammatory signatures. PLoS One 2019; 14:e0216810. [PMID: 31071164 PMCID: PMC6534080 DOI: 10.1371/journal.pone.0216810] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
The Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) share similar molecular characteristics in that they frequently harbor hotspot mutations in JAK2, CALR or MPL, leading to activated JAK/STAT signaling. However, these MPN have distinct symptoms, morphology, and natural histories, including different tendencies to progress to fibrosis. Although the role of inflammation in tissue fibrosis is well recognized, inflammatory gene expression in bone marrows involved by MPN has been understudied. We analyzed the expression of inflammatory genes by directly measuring RNA transcript abundance in bone marrow biopsies of 108 MPN patients. Unsupervised analyses identified gene expression patterns that distinguish prefibrotic (grade 1–2) MPN from overtly fibrotic (grade 2–3) MPN with high sensitivity and specificity in two independent cohorts. Furthermore, prefibrotic and overtly fibrotic MPN are separable into subsets with different activities in biological pathways linked to inflammation, including cytokines, chemokines, interferon response, and toll-like receptor signaling. In conclusion, this study demonstrates that MPN with overt fibrosis is associated with significant inflammatory gene upregulation in the bone marrow, revealing potential roles for multiple pro-inflammatory signaling networks in the development of myelofibrosis and suggesting potential therapeutic mechanisms to alleviate this process in the bone marrow microenvironment.
Collapse
|
29
|
Autoimmune-Associated Hemophagocytosis and Myelofibrosis in a Newly Diagnosed Lupus Patient: Case Report and Literature Review. Case Rep Hematol 2019; 2019:3879148. [PMID: 30729051 PMCID: PMC6343163 DOI: 10.1155/2019/3879148] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 12/16/2018] [Accepted: 12/19/2018] [Indexed: 02/07/2023] Open
Abstract
Bone marrow abnormalities in SLE are now becoming increasingly recognized, suggesting that the bone marrow may also be an important site of target organ damage. In this study, we present a rare case of concurrent autoimmune hemophagocytic syndrome and autoimmune myelofibrosis, potentially life-threatening conditions, in a newly diagnosed SLE patient. We report a case of a 30-year-old Filipino woman who presented with a one-year history of fever, constitutional symptoms, exertional dyspnea, joint pains, and alopecia and physical examination findings of fever, facial flushing, cervical lymphadenopathies, and knee joint effusions. Laboratory workup revealed pancytopenia with leukoerythroblastosis, elevated ESR, increased serum levels of transaminases, elevated CRP and LDH, hyperferritinemia, hypertriglyceridemia, proteinuria, hepatomegaly, and positive antinuclear antibody. Bone marrow aspiration and trephine biopsy revealed hemophagocytosis and moderate myelofibrosis. The patient was diagnosed with SLE with concomitant autoimmune-associated hemophagocytic syndrome and autoimmune myelofibrosis. Treatment with high-dose corticosteroids led to dramatic clinical improvement with normalization of laboratory data and complete resolution of bone marrow hemophagocytosis and myelofibrosis. Hemophagocytosis and myelofibrosis, although uncommon, are possible initial manifestations of SLE and should be included in the differential diagnosis of cytopenias in SLE. Thorough clinical assessment and microscopic bone marrow examination and timely initiation of corticosteroid therapy are essential in the diagnosis and management of these potentially life-threatening conditions. This case emphasizes that the bone marrow is an important site of target organ damage in SLE, and evaluation of cytopenias in SLE should take this into consideration.
Collapse
|
30
|
Li J, Ma X, Liu C, Li H, Zhuang J, Gao C, Zhou C, Liu L, Wang K, Sun C. Exploring the Mechanism of Danshen against Myelofibrosis by Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:8363295. [PMID: 30622613 PMCID: PMC6304517 DOI: 10.1155/2018/8363295] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/19/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023]
Abstract
Danshen (Salvia miltiorrhiza Bunge), a natural powerful drug for various conditions treatment, has traditionally been used in Asian countries for centuries as anticancer agent, anti-inflammatory agent, and antioxidant. More recently, it is explored in combination with other herbs for skeletal diseases therapy; bone-targeting compounds with pharmacological activities have been isolated from various sources of traditional Chinese medicine (TCM), including Danshen. In this case, some evidence supports that Danshen may treat myelofibrosis (MF) by exerting its antitumor effect. To study the specific mechanism of Danshen in the treatment of MF, we used bioinformatics databases to determine its active ingredients. Then, identification of target proteins related to MF was made using a network pharmacology analysis platform. In our results, 20 key active compounds and 457 key targets of Danshen were identified. In-depth network analysis of the top diseases, functions, and pathways suggested that a common underlying mechanism linked Danshen involvement with MF. Finally, 5 potential targets were confirmed by the analysis; these 5 targets, as well as 20 previously identified compounds, were subjected to molecular docking experiments. The results indicated that cryptotanshinone of Danshen may affect MF by acting on the key genes in the JAK-STAT signalling pathway and the TGF-β signalling pathway.
Collapse
Affiliation(s)
- Jie Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Xiaoran Ma
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Huayao Li
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261041, Shandong, China
| | - Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Chao Zhou
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261041, Shandong, China
| | - Lijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261041, Shandong, China
| | - Kejia Wang
- College of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, Shandong, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261041, Shandong, China
| |
Collapse
|
31
|
Rossi C, Zini R, Rontauroli S, Ruberti S, Prudente Z, Barbieri G, Bianchi E, Salati S, Genovese E, Bartalucci N, Guglielmelli P, Tagliafico E, Rosti V, Barosi G, Vannucchi AM, Manfredini R. Role of TGF-β1/miR-382-5p/SOD2 axis in the induction of oxidative stress in CD34+ cells from primary myelofibrosis. Mol Oncol 2018; 12:2102-2123. [PMID: 30259659 PMCID: PMC6275274 DOI: 10.1002/1878-0261.12387] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by an excessive production of pro-inflammatory cytokines resulting in chronic inflammation and genomic instability. Besides the driver mutations in JAK2, MPL, and CALR genes, the deregulation of miRNA expression may also contribute to the pathogenesis of PMF. To this end, we recently reported the upregulation of miR-382-5p in PMF CD34+ cells. In order to unveil the mechanistic details of the role of miR-382-5p in pathogenesis of PMF, we performed gene expression profiling of CD34+ cells overexpressing miR-382-5p. Among the downregulated genes, we identified superoxide dismutase 2 (SOD2), which is a predicted target of miR-382-5p. Subsequently, we confirmed miR-382-5p/SOD2 interaction by luciferase assay and we showed that miR-382-5p overexpression in CD34+ cells causes the decrease in SOD2 activity leading to reactive oxygen species (ROS) accumulation and oxidative DNA damage. In addition, our data indicate that inhibition of miR-382-5p in PMF CD34+ cells restores SOD2 function, induces ROS disposal, and reduces DNA oxidation. Since the pro-inflammatory cytokine transforming growth factor-β1 (TGF-β1) is a key player in PMF pathogenesis, we further investigated the effect of TGF-β1 on ROS and miR-382-5p levels. Our data showed that TGF-β1 treatment enhances miR-382-5p expression and reduces SOD2 activity leading to ROS accumulation. Finally, inhibition of TGF-β1 signaling in PMF CD34+ cells by galunisertib significantly reduced miR-382-5p expression and ROS accumulation and restored SOD2 activity. As a whole, this study reports that TGF-β1/miR-382-5p/SOD2 axis deregulation in PMF cells is linked to ROS overproduction that may contribute to enhanced oxidative stress and inflammation. Our results suggest that galunisertib may represent an effective drug reducing abnormal oxidative stress induced by TGF-β1 in PMF patients. DATABASE LINKING: GEO: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE103464.
Collapse
Affiliation(s)
- Chiara Rossi
- Department of Life Sciences, Centre for Regenerative Medicine, University of Modena and Reggio Emilia, Italy
| | - Roberta Zini
- Department of Life Sciences, Centre for Regenerative Medicine, University of Modena and Reggio Emilia, Italy
| | - Sebastiano Rontauroli
- Department of Life Sciences, Centre for Regenerative Medicine, University of Modena and Reggio Emilia, Italy
| | - Samantha Ruberti
- Department of Life Sciences, Centre for Regenerative Medicine, University of Modena and Reggio Emilia, Italy
| | - Zelia Prudente
- Department of Life Sciences, Centre for Regenerative Medicine, University of Modena and Reggio Emilia, Italy
| | - Greta Barbieri
- Department of Life Sciences, Centre for Regenerative Medicine, University of Modena and Reggio Emilia, Italy
| | - Elisa Bianchi
- Department of Life Sciences, Centre for Regenerative Medicine, University of Modena and Reggio Emilia, Italy
| | - Simona Salati
- Department of Life Sciences, Centre for Regenerative Medicine, University of Modena and Reggio Emilia, Italy
| | - Elena Genovese
- Department of Life Sciences, Centre for Regenerative Medicine, University of Modena and Reggio Emilia, Italy
| | - Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, AOU Careggi, University of Florence, Italy
| | - Paola Guglielmelli
- Department of Experimental and Clinical Medicine, CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, AOU Careggi, University of Florence, Italy
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, Italy
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnostics, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnostics, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Alessandro M Vannucchi
- Department of Experimental and Clinical Medicine, CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, AOU Careggi, University of Florence, Italy
| | - Rossella Manfredini
- Department of Life Sciences, Centre for Regenerative Medicine, University of Modena and Reggio Emilia, Italy
| | | |
Collapse
|
32
|
Talpaz M, Erickson-Viitanen S, Hou K, Hamburg S, Baer MR. Evaluation of an alternative ruxolitinib dosing regimen in patients with myelofibrosis: an open-label phase 2 study. J Hematol Oncol 2018; 11:101. [PMID: 30086777 PMCID: PMC6081850 DOI: 10.1186/s13045-018-0642-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/18/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Ruxolitinib improves splenomegaly and symptoms in patients with intermediate-2 or high-risk myelofibrosis; however, nearly half develop grade 3/4 anemia and/or thrombocytopenia, necessitating dose reductions and/or transfusions. We report findings from an open-label phase 2 study exploring a dose-escalation strategy aimed at preserving clinical benefit while reducing hematological adverse events early in ruxolitinib treatment. METHODS Patients with myelofibrosis received ruxolitinib 10 mg twice daily (BID), with incremental increases of 5 mg BID at weeks 12 and 18 for lack of efficacy (maximum, 20 mg BID). Symptom severity was measured using the Myelofibrosis Symptom Assessment Form Total Symptom Score (MFSAF TSS). RESULTS Forty-five patients were enrolled, 68.9% of whom had a Dynamic International Prognostic Scoring System score of 1 to 2 (i.e., intermediate-1 disease risk). Median percentage change in spleen volume from baseline to week 24 was - 17.3% (≥ 10% reduction achieved by 26 patients [57.8%]), with a clear dose response. Median percentage change in MFSAF TSS from baseline at week 24 was - 45.6%, also with a dose response. The most frequent treatment-emergent adverse events were anemia (26.7%), fatigue (22.2%), and arthralgias (20.0%). Grade 3/4 anemia (20.0%) and dose decreases due to anemia (11.1%) or thrombocytopenia (6.7%) were infrequent. CONCLUSIONS A dose-escalation approach may mitigate worsening anemia during early ruxolitinib therapy in some patients with myelofibrosis. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT01445769 . Registered September 23, 2011.
Collapse
Affiliation(s)
| | | | - Kevin Hou
- Incyte Corporation, Wilmington, DE, USA
| | - Solomon Hamburg
- Tower Hematology Oncology Medical Group, Beverly Hills, CA, USA
| | - Maria R Baer
- University of Maryland, Greenebaum Comprehensive Cancer Center, 22 S. Greene St, Baltimore, MD, 21201, USA.
| |
Collapse
|
33
|
Al-Fayoumi S, Hashiguchi T, Shirakata Y, Mascarenhas J, Singer JW. Pilot study of the antifibrotic effects of the multikinase inhibitor pacritinib in a mouse model of liver fibrosis. J Exp Pharmacol 2018; 10:9-17. [PMID: 29785143 PMCID: PMC5953271 DOI: 10.2147/jep.s150729] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Fibrotic diseases result from an exuberant response to chronic inflammation. Myelofibrosis is the end result of inflammation in bone, caused by an inflammatory process triggered by production of abnormal myeloid cells driven by mutations affecting the JAK-STAT pathway. Inflammatory cytokine overproduction leads to increased mesenchymal cell proliferation, culminating in fibrosis. Although JAK2 inhibitors, such as the JAK1/2 inhibitor ruxolitinib and the JAK2/FLT3/CSF1R/IRAK1 inhibitor pacritinib suppress abnormal clone expansion in myelofibrosis, ruxolitinib does not appear to prevent or reverse bone-marrow fibrosis in most patients. In two Phase III clinical trials, pacritinib, however, demonstrated improvements in platelet counts and hemoglobin and reductions in transfusion burden in some patients with baseline cytopenias, suggesting it may improve bone-marrow function. Unlike ruxolitinib, pacritinib suppresses signaling through IRAK1, a key control point for inflammatory and fibrotic signaling. Purpose To investigate potential antifibrotic effects of pacritinib in an animal model of liver fibrosis relevant to the observed course of human disease. Methods Pacritinib, negative control (vehicle), and positive control (the angiotensin 2-receptor antagonist and PPARγ partial agonist telmisartan) were assessed in the murine Stelic animal model, which mimics the clinically observed progression from hepatic steatosis to nonalcoholic steatohepatitis, liver fibrosis, and hepatocellular carcinoma. Histopathological analysis used hematoxylin and eosin staining. Body and liver weight changes, nonalcoholic fatty-liver disease activity scores, and plasma cytokeratin 18 fragment levels (a biomarker of hepatic necrosis) were measured. Results Pacritinib-treated mice had significantly (P<0.01) reduced fibrotic areas in liver compared to vehicle control and significantly (P<0.05) lower levels of CK18. The antifibrotic effect of pacritinib was comparable to that of telmisartan, but without significant effects on fat accumulation. Conclusion These results, the first to demonstrate hepatic antifibrotic effects for pacritinib in an animal model of liver disease, provide preliminary support for potential clinical applications of pacritinib in fibrotic diseases other than myelofibrosis.
Collapse
Affiliation(s)
| | | | | | - John Mascarenhas
- Tish Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
34
|
Goulard M, Dosquet C, Bonnet D. Role of the microenvironment in myeloid malignancies. Cell Mol Life Sci 2018; 75:1377-1391. [PMID: 29222645 PMCID: PMC5852194 DOI: 10.1007/s00018-017-2725-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/01/2017] [Accepted: 12/05/2017] [Indexed: 12/28/2022]
Abstract
The bone marrow microenvironment (BMM) regulates the fate of hematopoietic stem cells (HSCs) in homeostatic and pathologic conditions. In myeloid malignancies, new insights into the role of the BMM and its cellular and molecular actors in the progression of the diseases have started to emerge. In this review, we will focus on describing the major players of the HSC niche and the role of the altered niche function in myeloid malignancies, more specifically focusing on the mesenchymal stroma cell compartment.
Collapse
Affiliation(s)
- Marie Goulard
- INSERM, UMRS1131-Paris Diderot University, Saint Louis Hospital, Paris, France
| | - Christine Dosquet
- INSERM, UMRS1131-Paris Diderot University, Saint Louis Hospital, Paris, France
- Cell Biology Department, APHP, Saint Louis Hospital, Paris, France
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1, Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
35
|
Mäkitie RE, Niinimäki R, Kakko S, Honkanen T, Kovanen PE, Mäkitie O. Defective WNT signaling associates with bone marrow fibrosis-a cross-sectional cohort study in a family with WNT1 osteoporosis. Osteoporos Int 2018; 29:479-487. [PMID: 29147753 DOI: 10.1007/s00198-017-4309-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/07/2017] [Indexed: 01/28/2023]
Abstract
UNLABELLED This study explores bone marrow function in patients with defective WNT1 signaling. Bone marrow samples showed increased reticulin and altered granulopoiesis while overall hematopoiesis was normal. Findings did not associate with severity of osteoporosis. These observations provide new insight into the role of WNT signaling in bone marrow homeostasis. INTRODUCTION WNT signaling regulates bone homeostasis and survival and self-renewal of hematopoietic stem cells. Aberrant activation may lead to osteoporosis and bone marrow pathology. We aimed to explore bone marrow findings in a large family with early-onset osteoporosis due to a heterozygous WNT1 mutation. METHODS We analyzed peripheral blood samples, and bone marrow aspirates and biopsies from 10 subjects with WNT1 mutation p.C218G. One subject was previously diagnosed with idiopathic myelofibrosis and others had no previously diagnosed hematologic disorders. The findings were correlated with the skeletal phenotype, as evaluated by number of peripheral and spinal fractures and bone mineral density. RESULTS Peripheral blood samples showed no abnormalities in cell counts, morphology or distributions but mild increase in platelet count. Bone marrow aspirates (from 8/10 subjects) showed mild decrease in bone marrow iron storages in 6 and variation in cell distributions in 5 subjects. Bone marrow biopsies (from 6/10 subjects) showed increased bone marrow reticulin (grade MF-2 in the myelofibrosis subject and grade MF-1 in 4 others), and an increase in overall, and a shift towards early-phase, granulopoiesis. The bone marrow findings did not associate with the severity of skeletal phenotype. CONCLUSIONS Defective WNT signaling associates with a mild increase in bone marrow reticulin and may predispose to myelofibrosis, while overall hematopoiesis and peripheral blood values are unaltered in individuals with a WNT1 mutation. In this family with WNT1 osteoporosis, bone marrow findings were not related to the severity of osteoporosis.
Collapse
Affiliation(s)
- R E Mäkitie
- Folkhälsan Institute of Genetics, University of Helsinki, P.O. Box 63, FIN-00014, Helsinki, Finland.
| | - R Niinimäki
- Department of Children and Adolescents, Oulu University Hospital and Oulu University, Oulu, Finland
| | - S Kakko
- Internal Medicine and Clinical Research Center, University of Oulu, Oulu, Finland
| | - T Honkanen
- Department of Hematology, Päijät-Häme Central Hospital, Lahti, Finland
| | - P E Kovanen
- HUSLAB, Helsinki University Hospital and Department of Pathology, University of Helsinki, Helsinki, Finland
| | - O Mäkitie
- Folkhälsan Institute of Genetics, University of Helsinki, P.O. Box 63, FIN-00014, Helsinki, Finland
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Center for Molecular Medicine, Karolinska Institutet and Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
36
|
Yue L, Bartenstein M, Zhao W, Ho WT, Han Y, Murdun C, Mailloux AW, Zhang L, Wang X, Budhathoki A, Pradhan K, Rapaport F, Wang H, Shao Z, Ren X, Steidl U, Levine RL, Zhao ZJ, Verma A, Epling-Burnette PK. Efficacy of ALK5 inhibition in myelofibrosis. JCI Insight 2017; 2:e90932. [PMID: 28405618 DOI: 10.1172/jci.insight.90932] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Myelofibrosis (MF) is a bone marrow disorder characterized by clonal myeloproliferation, aberrant cytokine production, extramedullary hematopoiesis, and bone marrow fibrosis. Although somatic mutations in JAK2, MPL, and CALR have been identified in the pathogenesis of these diseases, inhibitors of the Jak2 pathway have not demonstrated efficacy in ameliorating MF in patients. TGF-β family members are profibrotic cytokines and we observed significant TGF-β1 isoform overexpression in a large cohort of primary MF patient samples. Significant overexpression of TGF-β1 was also observed in murine clonal MPLW515L megakaryocytic cells. TGF-β1 stimulated the deposition of excessive collagen by mesenchymal stromal cells (MSCs) by activating the TGF-β receptor I kinase (ALK5)/Smad3 pathway. MSCs derived from MPLW515L mice demonstrated sustained overproduction of both collagen I and collagen III, effects that were abrogated by ALK5 inhibition in vitro and in vivo. Importantly, use of galunisertib, a clinically active ALK5 inhibitor, significantly improved MF in both MPLW515L and JAK2V617F mouse models. These data demonstrate the role of malignant hematopoietic stem cell (HSC)/TGF-β/MSC axis in the pathogenesis of MF, and provide a preclinical rationale for ALK5 blockade as a therapeutic strategy in MF.
Collapse
Affiliation(s)
- Lanzhu Yue
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Matthias Bartenstein
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Wanke Zhao
- Department of Pathology, Peggy and Stephenson Cancer Center, Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Wanting Tina Ho
- Department of Pathology, Peggy and Stephenson Cancer Center, Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Ying Han
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Cem Murdun
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Adam W Mailloux
- Translational Science, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Ling Zhang
- Department of Hematopathology and Laboratory Medicine
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Anjali Budhathoki
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Kith Pradhan
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Franck Rapaport
- Leukemia Center, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Huaquan Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiubao Ren
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Ulrich Steidl
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Ross L Levine
- Leukemia Center, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Zhizhuang Joe Zhao
- Department of Pathology, Peggy and Stephenson Cancer Center, Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Amit Verma
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | | |
Collapse
|