1
|
Vermeersch G, Gouwy M, Proost P, Struyf S, Devos T. Neutrophils in BCR::ABL1 negative MPN: Contributors or bystanders of fibrosis? Blood Rev 2025:101285. [PMID: 40133166 DOI: 10.1016/j.blre.2025.101285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/20/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
BCR::ABL1 negative myeloproliferative neoplasms (MPNs) are a heterogenous group of disorders characterized by clonal proliferation of hematopoietic stem and progenitor cells (HSPCs) within the bone marrow. Although the identification of somatic key driver mutations significantly increased both understanding and diagnostic accuracy of MPNs, many questions about the exact pathophysiology remain unanswered. Increased neutrophil count at diagnosis is a well-recognized predictor of worse disease evolution and survival, nonetheless the exact role of neutrophilic granulocytes within MPN pathophysiology is almost unexplored. As the majority of these cells are residing within the bone marrow, they represent a non-negligible entity within the bone marrow niche and its homeostasis. This review describes how neutrophils might contribute to the development of the inflammatory bone marrow niche, and hereby also fibrosis, associated with MPNs. The versatile functions and effects in different contexts emphasize the necessity for future research oriented to bone marrow in addition to peripheral blood.
Collapse
Affiliation(s)
- Gaël Vermeersch
- Department of Hematology, University Hospitals Leuven, 3000 Leuven, Belgium; Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium.
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Timothy Devos
- Department of Hematology, University Hospitals Leuven, 3000 Leuven, Belgium; Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| |
Collapse
|
2
|
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
|
3
|
Todor SB, Ichim C, Boicean A, Mihaila RG. Cardiovascular Risk in Philadelphia-Negative Myeloproliferative Neoplasms: Mechanisms and Implications-A Narrative Review. Curr Issues Mol Biol 2024; 46:8407-8423. [PMID: 39194713 DOI: 10.3390/cimb46080496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs), encompassing disorders like polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are characterized by clonal hematopoiesis without the Philadelphia chromosome. The JAK2 V617F mutation is prevalent in PV, ET, and PMF, while mutations in MPL and CALR also play significant roles. These conditions predispose patients to thrombotic events, with PMF exhibiting the lowest survival among MPNs. Chronic inflammation, driven by cytokine release from aberrant leukocytes and platelets, amplifies cardiovascular risk through various mechanisms, including atherosclerosis and vascular remodeling. Additionally, MPN-related complications like pulmonary hypertension and cardiac fibrosis contribute to cardiovascular morbidity and mortality. This review consolidates recent research on MPNs' cardiovascular implications, emphasizing thrombotic risk, chronic inflammation, and vascular stiffness. Understanding these associations is crucial for developing targeted therapies and improving outcomes in MPN patients.
Collapse
Affiliation(s)
- Samuel Bogdan Todor
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Adrian Boicean
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | | |
Collapse
|
4
|
Karagianni A, Karkempetzaki AI, Brooks D, Matsuura S, Dambal V, Trackman PC, Ravid K. Deletion of mouse lysyl oxidase in megakaryocytes affects bone properties in a sex-dependent manner. Blood 2024; 143:2666-2670. [PMID: 38635757 PMCID: PMC11196861 DOI: 10.1182/blood.2024024620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024] Open
Abstract
ABSTRACT Lysyl oxidase (LOX) is a facilitator of extracellular matrix cross-linking. Using newly developed megakaryocyte-specific LOX knockout mice, we show that LOX expressed in these scarce bone marrow cells affects bone volume and collagen architecture in a sex-dependent manner.
Collapse
Affiliation(s)
- Aikaterini Karagianni
- Department of Medicine, Whitaker Cardiovascular Research Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
- Department of Internal Medicine, University of Crete, School of Medicine, Heraklion, Greece
| | - Anastasia Iris Karkempetzaki
- Department of Medicine, Whitaker Cardiovascular Research Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
- Department of Internal Medicine, University of Crete, School of Medicine, Heraklion, Greece
| | - Daniel Brooks
- Center for Musculoskeletal Research, Endocrine Unit, Massachusetts General Hospital, Boston, MA
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Boston, MA
| | - Shinobu Matsuura
- Department of Medicine, Whitaker Cardiovascular Research Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Vrinda Dambal
- Department of Translational Dental Medicine, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA
| | - Philip C. Trackman
- Department of Translational Dental Medicine, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA
- Forsyth Institute, Boston, MA
| | - Katya Ravid
- Department of Medicine, Whitaker Cardiovascular Research Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| |
Collapse
|
5
|
Abbonante V, Karkempetzaki AI, Leon C, Krishnan A, Huang N, Di Buduo CA, Cattaneo D, Ward CMT, Matsuura S, Guinard I, Weber J, De Acutis A, Vozzi G, Iurlo A, Ravid K, Balduini A. Newly identified roles for PIEZO1 mechanosensor in controlling normal megakaryocyte development and in primary myelofibrosis. Am J Hematol 2024; 99:336-349. [PMID: 38165047 PMCID: PMC10922533 DOI: 10.1002/ajh.27184] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024]
Abstract
Mechanisms through which mature megakaryocytes (Mks) and their progenitors sense the bone marrow extracellular matrix to promote lineage differentiation in health and disease are still partially understood. We found PIEZO1, a mechanosensitive cation channel, to be expressed in mouse and human Mks. Human mutations in PIEZO1 have been described to be associated with blood cell disorders. Yet, a role for PIEZO1 in megakaryopoiesis and proplatelet formation has never been investigated. Here, we show that activation of PIEZO1 increases the number of immature Mks in mice, while the number of mature Mks and Mk ploidy level are reduced. Piezo1/2 knockout mice show an increase in Mk size and platelet count, both at basal state and upon marrow regeneration. Similarly, in human samples, PIEZO1 is expressed during megakaryopoiesis. Its activation reduces Mk size, ploidy, maturation, and proplatelet extension. Resulting effects of PIEZO1 activation on Mks resemble the profile in Primary Myelofibrosis (PMF). Intriguingly, Mks derived from Jak2V617F PMF mice show significantly elevated PIEZO1 expression, compared to wild-type controls. Accordingly, Mks isolated from bone marrow aspirates of JAK2V617F PMF patients show increased PIEZO1 expression compared to Essential Thrombocythemia. Most importantly, PIEZO1 expression in bone marrow Mks is inversely correlated with patient platelet count. The ploidy, maturation, and proplatelet formation of Mks from JAK2V617F PMF patients are rescued upon PIEZO1 inhibition. Together, our data suggest that PIEZO1 places a brake on Mk maturation and platelet formation in physiology, and its upregulation in PMF Mks might contribute to aggravating some hallmarks of the disease.
Collapse
Affiliation(s)
- Vittorio Abbonante
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Anastasia Iris Karkempetzaki
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- University of Crete, School of Medicine, Heraklion, Greece
| | - Catherine Leon
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Anandi Krishnan
- Institute of Immunology, Stanford University School of Medicine, Palo Alto, California, United States
| | - Nasi Huang
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | | | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Christina Marie Torres Ward
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Shinobu Matsuura
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ines Guinard
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Josiane Weber
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Aurora De Acutis
- Interdepartmental Research Center "E. Piaggio", University of Pisa, Pisa, Italy
| | - Giovanni Vozzi
- Interdepartmental Research Center "E. Piaggio", University of Pisa, Pisa, Italy
| | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| |
Collapse
|
6
|
Yan X, Zhang N, Wei L, Zhang W, Huang T, Li W, Chen W, Yang A, You H. Selective inhibition of hepatic stellate cell and fibroblast-derived LOXL1 attenuates BDL- and Mdr2-/--induced cholestatic liver fibrosis. Am J Physiol Gastrointest Liver Physiol 2023; 325:G608-G621. [PMID: 37873581 DOI: 10.1152/ajpgi.00004.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023]
Abstract
Lysyl oxidase-like 1 (LOXL1) proteins are amine oxidases that play a crucial role in extracellular matrix remodeling due to their collagen cross-linking and intracellular functions. The role of LOXL1 in cholestatic liver fibrosis remains unexplored. We measured LOXL1 expression in two murine models of cholestasis [Mdr2 knockout (Mdr2-/-) and bile duct ligation (BDL)]. We used adeno-associated virus (AAV) serotype 6-mediated hepatic delivery against LOXL1 (AAV2/6-shLoxl1) to investigate the therapeutic efficacy of targeting LOXL1 in cholestatic liver fibrosis. NIH-3T3 murine fibroblasts were used to investigate the function and regulatory mechanisms of LOXL1 in vitro. LOXL1 expression was significantly upregulated in Mdr2-/- and BDL mice compared with their corresponding controls, predominantly in collagen-rich fibrous septa and portal areas. AAV2/6-shLoxl1 significantly reduced LOXL1 levels in Mdr2-/- and BDL mice, mainly in desmin-positive hepatic stellate cells (HSCs) and fibroblasts. Concomitant with reduced LOXL1 expression, there was reduced ductular reaction, inflammation, and fibrosis in both Mdr2-/- and BDL mice. In addition, Loxl1 intervention decreased Ki-67-positive cells in the desmin-positive areas in both Mdr2-/- and BDL mice. Overexpression of LOXL1 significantly promoted fibroblast proliferation by activating the platelet-derived growth factor receptor and extracellular signal-regulated kinase signaling pathways in vitro. Our findings demonstrated that selective inhibition of LOXL1 derived from HSCs/fibroblasts attenuated cholestatic liver/biliary fibrosis, inflammation, ductal reaction, and HSC/fibroblast proliferation. Based on our findings, LOXL1 could be a potential therapeutic target for cholestatic fibrosis.NEW & NOTEWORTHY Selectively, inhibition of HSC/fibroblasts-derived LOXL1 by AAV2/6-shLoxl1 could reduce collagen deposition, HSC/fibroblasts proliferation, and cholestatic liver fibrosis progression. In addition, overexpression of LOXL1 significantly promoted HSC/fibroblast proliferation by activating the PDGFRß/PI3K and ERK signaling pathways in vitro.
Collapse
Affiliation(s)
- Xuzhen Yan
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- Beijing Clinical Medicine Institute, Beijing, People's Republic of China
- National Clinical Research Center of Digestive Diseases, Beijing, People's Republic of China
| | - Ning Zhang
- National Clinical Research Center of Digestive Diseases, Beijing, People's Republic of China
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, People's Republic of China
| | - Luyang Wei
- National Clinical Research Center of Digestive Diseases, Beijing, People's Republic of China
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wen Zhang
- National Clinical Research Center of Digestive Diseases, Beijing, People's Republic of China
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, People's Republic of China
| | - Tao Huang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- Beijing Clinical Medicine Institute, Beijing, People's Republic of China
- National Clinical Research Center of Digestive Diseases, Beijing, People's Republic of China
| | - Weiyu Li
- National Clinical Research Center of Digestive Diseases, Beijing, People's Republic of China
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, People's Republic of China
| | - Wei Chen
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- Beijing Clinical Medicine Institute, Beijing, People's Republic of China
- National Clinical Research Center of Digestive Diseases, Beijing, People's Republic of China
| | - Aiting Yang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- Beijing Clinical Medicine Institute, Beijing, People's Republic of China
- National Clinical Research Center of Digestive Diseases, Beijing, People's Republic of China
| | - Hong You
- Beijing Clinical Medicine Institute, Beijing, People's Republic of China
- National Clinical Research Center of Digestive Diseases, Beijing, People's Republic of China
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, People's Republic of China
| |
Collapse
|
7
|
Nakajima K, Suzuki M, Kawashima I, Koshiisi M, Kumagai T, Yamamoto T, Tanaka M, Kirito K. The chaperone protein GRP78 released from MPN cells increases the expression of lysyl oxidase in a human stromal cell line. Leuk Res 2023; 134:107389. [PMID: 37757654 DOI: 10.1016/j.leukres.2023.107389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 08/30/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Impaired function of the endoplasmic stress (ER) response causes numerous pathological conditions, including tissue fibrosis. In the present study, we aimed to determine the pathological role of ER stress response systems in myeloproliferative neoplasms (MPNs). We found increased expression of the chaperone protein glucose-regulated protein (GRP) 78, a central regulator of ER stress, in megakaryocytes from primary myelofibrosis or postessential thrombocythemia myelofibrosis patients. GRP78 was overexpressed in JAK2V617F-harboring cell lines; however, inhibitors of ER stress did not affect the expression levels of GRP78. In contrast, ruxolitinib, a well-known inhibitor of JAK2V617F, clearly blocked GRP78 expression in these cells through downregulation of transcription factor 4 (ATF4). Interestingly, GRP78 was secreted from HEL and SET-2 cells into culture media. Coculture of these cells with HS-5 cells, a human bone marrow stroma-derived cell line, induced enhanced expression of lysyl oxidase (LOX), which mediates cross-linking of collagen fibers and induces tissue fibrosis, in HS-5 cells. An anti-GRP78 neutralizing antibody abrogated LOX elevation; in contrast, recombinant GRP78 protein induced LOX protein expression in HS-5 cells. Our observations suggest that the oncogenic protein JAK2V617F induces overexpression and release of GRP78, which may induce a fibrotic phenotype in surrounding bone marrow stromal cells.
Collapse
Affiliation(s)
- Kei Nakajima
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Megumi Suzuki
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Ichiro Kawashima
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Megumi Koshiisi
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Takuma Kumagai
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Takeo Yamamoto
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Masaru Tanaka
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Keita Kirito
- Department of Hematology/Oncology, University of Yamanashi, Japan.
| |
Collapse
|
8
|
Vainchenker W, Yahmi N, Havelange V, Marty C, Plo I, Constantinescu SN. Recent advances in therapies for primary myelofibrosis. Fac Rev 2023; 12:23. [PMID: 37771602 PMCID: PMC10523375 DOI: 10.12703/r/12-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023] Open
Abstract
Primary myelofibrosis (PMF), polycythemia vera (PV) and essential thrombocythemia (ET) form the classical BCR-ABL1-negative myeloproliferative neoplasms (MPNs) that are driven by a constitutive activation of JAK2 signaling. PMF as well as secondary MF (post-ET and post-PV MF) are the most aggressive MPNs. Presently, there is no curative treatment, except allogenic hematopoietic stem cell transplantation. JAK inhibitors, essentially ruxolitinib, are the therapy of reference for intermediate and high-risk MF. However, presently the current JAK inhibitors behave mainly as anti-inflammatory drugs, improving general symptoms and spleen size without major impact on disease progression. A better understanding of the genetics of MF, the biology of its leukemic stem cells (LSCs), the mechanisms of fibrosis and of cytopenia and the role of inflammatory cytokines has led to new approaches with the development of numerous therapeutic agents that target epigenetic regulation, telomerase, apoptosis, cell cycle, cytokines and signaling. Furthermore, the use of a new less toxic form of interferon-α has been revived, as it is presently one of the only molecules that targets the mutated clone. These new approaches have different aims: (a) to provide alternative therapy to JAK inhibition; (b) to correct cytopenia; and (c) to inhibit fibrosis development. However, the main important goal is to find new disease modifier treatments, which will profoundly modify the progression of the disease without major toxicity. Presently the most promising approaches consist of the inhibition of telomerase and the combination of JAK2 inhibitors (ruxolitinib) with either a BCL2/BCL-xL or BET inhibitor. Yet, the most straightforward future approaches can be considered to be the development of and/or selective inhibition of JAK2V617F and the targeting MPL and calreticulin mutants by immunotherapy. It can be expected that the therapy of MF will be significantly improved in the coming years.
Collapse
Affiliation(s)
- William Vainchenker
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Nasrine Yahmi
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Violaine Havelange
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Cliniques universitaires Saint Luc, Department of Hematology, Université Catholique de Louvain, Brussels, Belgium
| | - Caroline Marty
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1287, Villejuif, France
| | - Stefan N Constantinescu
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, Brussels, Belgium
- WEL Research Institute, WELBIO Department, Wavre, Belgium
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| |
Collapse
|
9
|
Ding Z, Shi R, Hu W, Tian L, Sun R, Wu Y, Zhang X. Cancer-associated fibroblasts in hematologic malignancies: elucidating roles and spotlighting therapeutic targets. Front Oncol 2023; 13:1193978. [PMID: 37746306 PMCID: PMC10511871 DOI: 10.3389/fonc.2023.1193978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Hematologic malignancies comprise a diverse range of blood, bone marrow, and organ-related disorders that present significant challenges due to drug resistance, relapse, and treatment failure. Cancer-associated fibroblasts (CAFs) represent a critical component of the tumor microenvironment (TME) and have recently emerged as potential therapeutic targets. In this comprehensive review, we summarize the latest findings on the roles of CAFs in various hematologic malignancies, including acute leukemia, multiple myeloma, chronic lymphocytic leukemia, myeloproliferative neoplasms, and lymphoma. We also explore their involvement in tumor progression, drug resistance, and the various signaling pathways implicated in their activation and function. While the underlying mechanisms and the existence of multiple CAF subtypes pose challenges, targeting CAFs and their associated pathways offers a promising avenue for the development of innovative treatments to improve patient outcomes in hematologic malignancies.
Collapse
Affiliation(s)
- Ziyang Ding
- The Second Clinical School of Nanjing Medical University, Nanjing, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weikang Hu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Tian
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rong Sun
- Department of Radiation Oncology, Jinling Hospital, Nanjing, China
| | - Yang Wu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoyan Zhang
- Department of Hematology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Hasselbalch HC, Junker P, Skov V, Kjær L, Knudsen TA, Larsen MK, Holmström MO, Andersen MH, Jensen C, Karsdal MA, Willumsen N. Revisiting Circulating Extracellular Matrix Fragments as Disease Markers in Myelofibrosis and Related Neoplasms. Cancers (Basel) 2023; 15:4323. [PMID: 37686599 PMCID: PMC10486581 DOI: 10.3390/cancers15174323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 09/10/2023] Open
Abstract
Philadelphia chromosome-negative chronic myeloproliferative neoplasms (MPNs) arise due to acquired somatic driver mutations in stem cells and develop over 10-30 years from the earliest cancer stages (essential thrombocythemia, polycythemia vera) towards the advanced myelofibrosis stage with bone marrow failure. The JAK2V617F mutation is the most prevalent driver mutation. Chronic inflammation is considered to be a major pathogenetic player, both as a trigger of MPN development and as a driver of disease progression. Chronic inflammation in MPNs is characterized by persistent connective tissue remodeling, which leads to organ dysfunction and ultimately, organ failure, due to excessive accumulation of extracellular matrix (ECM). Considering that MPNs are acquired clonal stem cell diseases developing in an inflammatory microenvironment in which the hematopoietic cell populations are progressively replaced by stromal proliferation-"a wound that never heals"-we herein aim to provide a comprehensive review of previous promising research in the field of circulating ECM fragments in the diagnosis, treatment and monitoring of MPNs. We address the rationales and highlight new perspectives for the use of circulating ECM protein fragments as biologically plausible, noninvasive disease markers in the management of MPNs.
Collapse
Affiliation(s)
- Hans Carl Hasselbalch
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Peter Junker
- Department of Rheumatology, Odense University Hospital, 5000 Odense, Denmark;
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Trine A. Knudsen
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Morten Kranker Larsen
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Morten Orebo Holmström
- National Center for Cancer Immune Therapy, Herlev Hospital, 2730 Herlev, Denmark; (M.O.H.); (M.H.A.)
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Herlev Hospital, 2730 Herlev, Denmark; (M.O.H.); (M.H.A.)
| | - Christina Jensen
- Nordic Bioscience A/S, 2730 Herlev, Denmark; (C.J.); (M.A.K.); (N.W.)
| | - Morten A. Karsdal
- Nordic Bioscience A/S, 2730 Herlev, Denmark; (C.J.); (M.A.K.); (N.W.)
| | | |
Collapse
|
11
|
Willekens C, Laplane L, Dagher T, Benlabiod C, Papadopoulos N, Lacout C, Rameau P, Catelain C, Alfaro A, Edmond V, Signolle N, Marchand V, Droin N, Hoogenboezem R, Schneider RK, Penson A, Abdel-Wahab O, Giraudier S, Pasquier F, Marty C, Plo I, Villeval JL, Constantinescu SN, Porteu F, Vainchenker W, Solary E. SRSF2-P95H decreases JAK/STAT signaling in hematopoietic cells and delays myelofibrosis development in mice. Leukemia 2023:10.1038/s41375-023-01878-0. [PMID: 37100881 DOI: 10.1038/s41375-023-01878-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 04/28/2023]
Abstract
Heterozygous mutation targeting proline 95 in Serine/Arginine-rich Splicing Factor 2 (SRSF2) is associated with V617F mutation in Janus Activated Kinase 2 (JAK2) in some myeloproliferative neoplasms (MPNs), most commonly primary myelofibrosis. To explore the interaction of Srsf2P95H with Jak2V617F, we generated Cre-inducible knock-in mice expressing these mutants under control of the stem cell leukemia (Scl) gene promoter. In transplantation experiments, Srsf2P95H unexpectedly delayed myelofibrosis induced by Jak2V617F and decreased TGFβ1 serum level. Srsf2P95H reduced the competitiveness of transplanted Jak2V617F hematopoietic stem cells while preventing their exhaustion. RNA sequencing of sorted megakaryocytes identified an increased number of splicing events when the two mutations were combined. Focusing on JAK/STAT pathway, Jak2 exon 14 skipping was promoted by Srsf2P95H, an event detected in patients with JAK2V617F and SRSF2P95 co-mutation. The skipping event generates a truncated inactive JAK2 protein. Accordingly, Srsf2P95H delays myelofibrosis induced by the thrombopoietin receptor agonist Romiplostim in Jak2 wild-type animals. These results unveil JAK2 exon 14 skipping promotion as a strategy to reduce JAK/STAT signaling in pathological conditions.
Collapse
Affiliation(s)
- Christophe Willekens
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Département d'hématologie, Gustave Roussy Cancer Campus, Villejuif, France
| | - Lucie Laplane
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Institut d'Histoire et Philosophie des Sciences et des Techniques, Université Paris I Panthéon-Sorbonne, Paris, France
| | - Tracy Dagher
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Camelia Benlabiod
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Institut d'Histoire et Philosophie des Sciences et des Techniques, Université Paris I Panthéon-Sorbonne, Paris, France
| | - Nicolas Papadopoulos
- Ludwig Institute for Cancer Research Brussels, Brussels, Belgium
- Université catholique de Louvain and de Duve Institute, Brussels, Belgium
| | | | | | | | | | - Valérie Edmond
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Valentine Marchand
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Nathalie Droin
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Remco Hoogenboezem
- Department of Hematology, Erasmus University, Rotterdam, The Netherlands
| | - Rebekka K Schneider
- Department of Hematology, Erasmus University, Rotterdam, The Netherlands
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, RWTH Aachen University, Aachen, Germany
| | - Alex Penson
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Florence Pasquier
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Département d'hématologie, Gustave Roussy Cancer Campus, Villejuif, France
| | - Caroline Marty
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Isabelle Plo
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Jean-Luc Villeval
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research Brussels, Brussels, Belgium
- Université catholique de Louvain and de Duve Institute, Brussels, Belgium
- WELBIO department, WEL Research Institute, Wavre, Belgium
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Françoise Porteu
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - William Vainchenker
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Eric Solary
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France.
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.
- Département d'hématologie, Gustave Roussy Cancer Campus, Villejuif, France.
| |
Collapse
|
12
|
Xu Q, Streuer A, Jann JC, Altrock E, Schmitt N, Flach J, Sens-Albert C, Rapp F, Wolf J, Nowak V, Weimer N, Obländer J, Palme I, Kuzina M, Jawhar A, Darwich A, Weis CA, Marx A, Wuchter P, Costina V, Jäger E, Sperk E, Neumaier M, Fabarius A, Metzgeroth G, Nolte F, Steiner L, Levkin PA, Jawhar M, Hofmann WK, Riabov V, Nowak D. Inhibition of lysyl oxidases synergizes with 5-azacytidine to restore erythropoiesis in myelodysplastic and myeloid malignancies. Nat Commun 2023; 14:1497. [PMID: 36932114 PMCID: PMC10023686 DOI: 10.1038/s41467-023-37175-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 03/01/2023] [Indexed: 03/19/2023] Open
Abstract
Limited response rates and frequent relapses during standard of care with hypomethylating agents in myelodysplastic neoplasms (MN) require urgent improvement of this treatment indication. Here, by combining 5-azacytidine (5-AZA) with the pan-lysyl oxidase inhibitor PXS-5505, we demonstrate superior restoration of erythroid differentiation in hematopoietic stem and progenitor cells (HSPCs) of MN patients in 20/31 cases (65%) versus 9/31 cases (29%) treated with 5-AZA alone. This effect requires direct contact of HSPCs with bone marrow stroma components and is dependent on integrin signaling. We further confirm these results in vivo using a bone marrow niche-dependent MN xenograft model in female NSG mice, in which we additionally demonstrate an enforced reduction of dominant clones as well as significant attenuation of disease expansion and normalization of spleen sizes. Overall, these results lay out a strong pre-clinical rationale for efficacy of combination treatment of 5-AZA with PXS-5505 especially for anemic MN.
Collapse
Affiliation(s)
- Qingyu Xu
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Alexander Streuer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Johann-Christoph Jann
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Eva Altrock
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Nanni Schmitt
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Johanna Flach
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Carla Sens-Albert
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Felicitas Rapp
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Julia Wolf
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Verena Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Nadine Weimer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Julia Obländer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Iris Palme
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Mariia Kuzina
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, 76344, Germany
| | - Ahmed Jawhar
- Department of Orthopedic Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Ali Darwich
- Department of Orthopedic Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Cleo-Aron Weis
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Alexander Marx
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg-Hessen, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Victor Costina
- Institute of Clinical Chemistry, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Evelyn Jäger
- Institute of Clinical Chemistry, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Elena Sperk
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Michael Neumaier
- Institute of Clinical Chemistry, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Alice Fabarius
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Georgia Metzgeroth
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Florian Nolte
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Laurenz Steiner
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Pavel A Levkin
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, 76344, Germany
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Karlsruhe, 76131, Germany
| | - Mohamad Jawhar
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Vladimir Riabov
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.
| |
Collapse
|
13
|
Saleh M, Essawy E, Shaalan M, Osman S, Ahmed F, El-Matbouli M. Therapeutic Intervention with Dietary Chitosan Nanoparticles Alleviates Fish Pathological and Molecular Systemic Inflammatory Responses against Infections. Mar Drugs 2022; 20:md20070425. [PMID: 35877718 PMCID: PMC9315998 DOI: 10.3390/md20070425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Marine bio-sourced chitosan nanoparticles (CSNP) are antimicrobial and immunomodulatory agents beneficial for fish medicine. Herein, dietary CSNP was investigated for the amelioration of the systemic inflammatory responses of an induced fish model. One hundred and forty-four rainbow trout were assigned to one pathogen-free and non-supplemented group (negative control), and three challenged groups: non-supplemented (positive control), CSNP-preventive, and CSNP-therapeutic. After a feeding experiment extended for 21 days, the organosomatic indices (OSI) and molecular aspects were assessed. After a challenge experiment extended for further 28 days, CSNP-therapeutic intervention was assessed on fish survival and systemic inflammatory responses on pathology, histo-morphology, and molecular aspects. With CSNP administration, OSI nonsignificantly decreased and the relative expression of targeted inflammatory-mediator genes was significantly increased. The CSNP-therapeutic fish showed an RPS of 80% as compared to the positive control group, and CSNP-therapeutic administration retained the highest gene expression augmentation up to 28 days after the challenge. Notably, the splenic reticulin fibers framework of the CSNP-therapeutic group retained the highest integrity among the groups during the infection. After recovery, reticulin fibers density in the CSNP-therapeutic samples was significantly higher than in the negative control group, which indicates high innate immunity. Thus, CSNP showed promising biotherapeutic features enhancing fish resistance against infections.
Collapse
Affiliation(s)
- Mona Saleh
- Clinical Division of Fish Medicine, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria; (E.E.); (M.S.); (M.E.-M.)
- Correspondence: (M.S.); (F.A.)
| | - Ehab Essawy
- Clinical Division of Fish Medicine, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria; (E.E.); (M.S.); (M.E.-M.)
- Department of Chemistry, Biochemistry Division, Faculty of Science, Helwan University, Cairo 11790, Egypt
- Bioinformatics Center, Faculty of Science, Helwan University, Cairo 11790, Egypt
| | - Mohamed Shaalan
- Clinical Division of Fish Medicine, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria; (E.E.); (M.S.); (M.E.-M.)
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo 12613, Egypt
| | - Shaaban Osman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt;
| | - Fatma Ahmed
- Department of Zoology, Faculty of Science, Sohag University, Sohag 82524, Egypt
- Correspondence: (M.S.); (F.A.)
| | - Mansour El-Matbouli
- Clinical Division of Fish Medicine, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria; (E.E.); (M.S.); (M.E.-M.)
- Division of Aquatic Animal Health, School of Veterinary Medicine, Badr Universiy, Cairo 11829, Egypt
| |
Collapse
|
14
|
Leiva O, Hobbs G, Ravid K, Libby P. Cardiovascular Disease in Myeloproliferative Neoplasms: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2022; 4:166-182. [PMID: 35818539 PMCID: PMC9270630 DOI: 10.1016/j.jaccao.2022.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 11/24/2022] Open
Abstract
Myeloproliferative neoplasms are associated with increased risk for thrombotic complications. These conditions most commonly involve somatic mutations in genes that lead to constitutive activation of the Janus-associated kinase signaling pathway (eg, Janus kinase 2, calreticulin, myeloproliferative leukemia protein). Acquired gain-of-function mutations in these genes, particularly Janus kinase 2, can cause a spectrum of disorders, ranging from clonal hematopoiesis of indeterminate potential, a recently recognized age-related promoter of cardiovascular disease, to frank hematologic malignancy. Beyond thrombosis, patients with myeloproliferative neoplasms can develop other cardiovascular conditions, including heart failure and pulmonary hypertension. The authors review the pathophysiologic mechanisms of cardiovascular complications of myeloproliferative neoplasms, which involve inflammation, prothrombotic and profibrotic factors (including transforming growth factor-beta and lysyl oxidase), and abnormal function of circulating clones of mutated leukocytes and platelets from affected individuals. Anti-inflammatory therapies may provide cardiovascular benefit in patients with myeloproliferative neoplasms, a hypothesis that requires rigorous evaluation in clinical trials.
Collapse
Key Words
- ASXL1, additional sex Combs-like 1
- CHIP, clonal hematopoiesis of indeterminate potential
- DNMT3a, DNA methyltransferase 3 alpha
- IL, interleukin
- JAK, Janus-associated kinase
- JAK2, Janus kinase 2
- LOX, lysyl oxidase
- MPL, myeloproliferative leukemia protein
- MPN, myeloproliferative neoplasm
- STAT, signal transducer and activator of transcription
- TET2, tet methylcytosine dioxygenase 2
- TGF, transforming growth factor
- atherosclerosis
- cardiovascular complications
- clonal hematopoiesis
- myeloproliferative neoplasms
- thrombosis
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gabriela Hobbs
- Division of Hematology Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Huang DY, Wang GM, Ke ZR, Zhou Y, Yang HH, Ma TL, Guan CX. Megakaryocytes in pulmonary diseases. Life Sci 2022; 301:120602. [DOI: 10.1016/j.lfs.2022.120602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/06/2023]
|
16
|
Deletion of Grin1 in mouse megakaryocytes reveals NMDA receptor role in platelet function and proplatelet formation. Blood 2022; 139:2673-2690. [PMID: 35245376 DOI: 10.1182/blood.2021014000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/18/2022] [Indexed: 11/20/2022] Open
Abstract
The process of proplatelet formation (PPF) requires coordinated interaction between megakaryocytes (MKs) and the extracellular matrix (ECM), followed by a dynamic reorganization of the actin and microtubule cytoskeleton. Localized fluxes of intracellular calcium ions (Ca2+) facilitate MK-ECM interaction and PPF. Glutamate-gated N-methyl-D--aspartate receptor (NMDAR) is highly permeable to Ca2+. NMDAR antagonists inhibit MK maturation ex vivo, however there is no in vivo data. Using the Cre-loxP system, we generated a platelet lineage-specific knockout mouse model of reduced NMDAR function in MKs and platelets (Pf4-Grin1-/- mice). Effects of NMDAR deletion were examined using well-established assays of platelet function and production in vivo and ex vivo. We found that Pf4-Grin1-/- mice had defects in megakaryopoiesis, thrombopoiesis and platelet function, which manifested as reduced platelet counts, lower rates of platelet production in the immune model of thrombocytopenia, and a prolonged tail bleeding time. Platelet activation was impaired to a range of agonists associated with reduced Ca2+ responses, including metabotropic-like, and defective platelet spreading. MKs showed reduced colony and proplatelet formation. Impaired reorganization of intracellular F-actin and α-tubulin was identified as the main cause of reduced platelet function and production. Pf4-Grin1-/- MKs also had lower levels of transcripts encoding crucial ECM elements and enzymes, suggesting NMDAR signaling is involved in ECM remodeling. In summary, we provide the first genetic evidence that NMDAR plays an active role in platelet function and production. NMDARs regulate PPF through the mechanism that involves MK-ECM interaction and cytoskeletal reorganization. Our results suggest that NMDAR helps guide PPF in vivo.
Collapse
|
17
|
Gaye MM, Ward CM, Piasecki AJ, Stahl VL, Karagianni A, Costello CE, Ravid K. Characterization of Glycoproteoforms of Integrins α2 and β1 in Megakaryocytes in the Occurrence of JAK2V617F Mutation-Induced Primary Myelofibrosis. Mol Cell Proteomics 2022; 21:100213. [PMID: 35182768 PMCID: PMC8968581 DOI: 10.1016/j.mcpro.2022.100213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/14/2022] [Accepted: 02/14/2022] [Indexed: 12/22/2022] Open
Abstract
Primary myelofibrosis (PMF) is a neoplasm prone to leukemic transformation, for which limited treatment is available. Among individuals diagnosed with PMF, the most prevalent mutation is the JAK2V617F somatic point mutation that activates the Janus kinase 2 (JAK2) enzyme. Our earlier reports on hyperactivity of β1 integrin and enhanced adhesion activity of the α2β1 complex in JAK2V617F megakaryocytes (MKs) led us to examine the new hypothesis that this mutation leads to posttranslational modification via changes in glycosylation. Samples were derived from immunoprecipitation of MKs obtained from Vav1-hJAK2V617F and WT mice. Immunoprecipitated fractions were separated by SDS-PAGE and analyzed using LC-MS/MS techniques in a bottom-up glycoproteomics workflow. In the immunoprecipitate, glycopeptiforms corresponding to 11 out of the 12 potential N-glycosylation sites of integrin β1 and to all nine potential glycosylation sites of integrin α2 were observed. Glycopeptiforms were compared across WT and JAK2V617F phenotypes for both integrins. The overall trend observed is that JAK2V617F mutation in PMF MKs leads to changes in β1 glycosylation; in most cases, it results in an increase in the integrated area of glycopeptiforms. We also observed that in mutated MKs, changes in integrin α2 glycosylation were more substantial than those observed for integrin β1 glycosylation, a finding that suggests that altered integrin α2 glycosylation may also affect activation. Additionally, the identification of proteins associated to the cytoskeleton that were co-immunoprecipitated with integrins α2 and β1 demonstrated the potential of the methodology employed in this study to provide some insight, at the peptide level, into the consequences of integrin activation in MKs. The extensive and detailed glycosylation patterns we uncovered provide a basis for future functional studies of each site in control cells as compared to JAK2V617F-mutated cells. Data are available via ProteomeXchange with identifier PXD030550.
Collapse
Affiliation(s)
- Maissa M. Gaye
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, Massachusetts, USA,Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Christina M. Ward
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Andrew J. Piasecki
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Vanessa L. Stahl
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Aikaterini Karagianni
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA,Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Catherine E. Costello
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, Massachusetts, USA,For correspondence: Catherine E. Costello; Katya Ravid
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
18
|
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
|
19
|
Migliaccio AR. A Novel Megakaryocyte Subpopulation Poised to Exert the Function of HSC Niche as Possible Driver of Myelofibrosis. Cells 2021; 10:3302. [PMID: 34943811 PMCID: PMC8699046 DOI: 10.3390/cells10123302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
Careful morphological investigations, coupled with experimental hematology studies in animal models and in in vitro human cultures, have identified that platelets are released in the circulation by mature megakaryocytes generated by hematopoietic stem cells by giving rise to lineage-restricted progenitor cells and then to morphologically recognizable megakaryocyte precursors, which undergo a process of terminal maturation. Advances in single cell profilings are revolutionizing the process of megakaryocytopoiesis as we have known it up to now. They identify that, in addition to megakaryocytes responsible for producing platelets, hematopoietic stem cells may generate megakaryocytes, which exert either immune functions in the lung or niche functions in organs that undergo tissue repair. Furthermore, it has been discovered that, in addition to hematopoietic stem cells, during ontogeny, and possibly in adult life, megakaryocytes may be generated by a subclass of specialized endothelial precursors. These concepts shed new light on the etiology of myelofibrosis, the most severe of the Philadelphia negative myeloproliferative neoplasms, and possibly other disorders. This perspective will summarize these novel concepts in thrombopoiesis and discuss how they provide a framework to reconciliate some of the puzzling data published so far on the etiology of myelofibrosis and their implications for the therapy of this disease.
Collapse
Affiliation(s)
- Anna Rita Migliaccio
- Department of Medicine and Surgery, Campus Bio-Medico University, 00128 Rome, Italy; or amigliaccio.altius.org
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
| |
Collapse
|
20
|
Bone marrow microenvironment of MPN cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021. [PMID: 34756245 DOI: 10.1016/bs.ircmb.2021.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
In this chapter, we will discuss the current knowledge concerning the alterations of the cellular components in the bone marrow niche in Myeloproliferative Neoplasms (MPNs), highlighting the central role of the megakaryocytes in MPN progression, and the extracellular matrix components characterizing the fibrotic bone marrow.
Collapse
|
21
|
Liu Y, Gu Z, Cao H, Kaphle P, Lyu J, Zhang Y, Hu W, Chung SS, Dickerson KE, Xu J. Convergence of oncogenic cooperation at single-cell and single-gene levels drives leukemic transformation. Nat Commun 2021; 12:6323. [PMID: 34732703 PMCID: PMC8566485 DOI: 10.1038/s41467-021-26582-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/13/2021] [Indexed: 12/17/2022] Open
Abstract
Cancers develop from the accumulation of somatic mutations, yet it remains unclear how oncogenic lesions cooperate to drive cancer progression. Using a mouse model harboring NRasG12D and EZH2 mutations that recapitulates leukemic progression, we employ single-cell transcriptomic profiling to map cellular composition and gene expression alterations in healthy or diseased bone marrows during leukemogenesis. At cellular level, NRasG12D induces myeloid lineage-biased differentiation and EZH2-deficiency impairs myeloid cell maturation, whereas they cooperate to promote myeloid neoplasms with dysregulated transcriptional programs. At gene level, NRasG12D and EZH2-deficiency independently and synergistically deregulate gene expression. We integrate results from histopathology, leukemia repopulation, and leukemia-initiating cell assays to validate transcriptome-based cellular profiles. We use this resource to relate developmental hierarchies to leukemia phenotypes, evaluate oncogenic cooperation at single-cell and single-gene levels, and identify GEM as a regulator of leukemia-initiating cells. Our studies establish an integrative approach to deconvolute cancer evolution at single-cell resolution in vivo.
Collapse
Affiliation(s)
- Yuxuan Liu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Zhimin Gu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hui Cao
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Pranita Kaphle
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Junhua Lyu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yuannyu Zhang
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Wenhuo Hu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Stephen S Chung
- Division of Hematology Oncology, Department of Internal Medicine, and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kathryn E Dickerson
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jian Xu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
22
|
Abstract
Thrombocytopoiesis is a complex process beginning at the level of hematopoietic stem cells, which ultimately generate megakaryocytes, large marrow cells with a distinctive morphology, and then, through a process of terminal maturation, megakaryocytes shed thousands of platelets into the circulation. This process is controlled by intrinsic and extrinsic factors. Emerging data indicate that an important intrinsic control on the late stages of thrombopoiesis is exerted by integrins, a family of transmembrane receptors composed of one α and one β subunit. One β subunit expressed by megakaryocytes is the β1 integrin, the role of which in the regulation of platelet formation is beginning to be clarified. Here, we review recent data indicating that activation of β1 integrin by outside-in and inside-out signaling regulates the interaction of megakaryocytes with the endosteal niche, which triggers their maturation, while its inactivation by galactosylation determines the migration of these cells to the perivascular niche, where they complete their terminal maturation and release platelets in the bloodstream. Furthermore, β1 integrin mediates the activation of transforming growth factor β (TGF-β), a protein produced by megakaryocytes that may act in an autocrine fashion to halt their maturation and affect the composition of their surrounding extracellular matrix. These findings suggest that β1 integrin could be a therapeutic target for inherited and acquired disorders of platelet production.
Collapse
Affiliation(s)
- Maria Mazzarini
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Italy
| | - Paola Verachi
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Italy
| | - Fabrizio Martelli
- National Center for Preclinical and Clinical Research and Evaluation of Pharmaceutical Drugs, Rome, Italy
| | - Anna Rita Migliaccio
- University Campus Biomedico, Rome, Italy
- Myeloproliferative Neoplasm-Research Consortium, New York, NY, USA
| |
Collapse
|
23
|
Varricchio L, Iancu-Rubin C, Upadhyaya B, Zingariello M, Martelli F, Verachi P, Clementelli C, Denis JF, Rahman AH, Tremblay G, Mascarenhas J, Mesa RA, O'Connor-McCourt M, Migliaccio AR, Hoffman R. TGFβ1 protein trap AVID200 beneficially affects hematopoiesis and bone marrow fibrosis in myelofibrosis. JCI Insight 2021; 6:e145651. [PMID: 34383713 PMCID: PMC8492354 DOI: 10.1172/jci.insight.145651] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 08/11/2021] [Indexed: 11/17/2022] Open
Abstract
Myelofibrosis (MF) is a progressive chronic myeloproliferative neoplasm characterized by hyperactivation of JAK/STAT signaling and dysregulation of the transcription factor GATA1 in megakaryocytes (MKs). TGF-β plays a pivotal role in the pathobiology of MF by promoting BM fibrosis and collagen deposition and by enhancing the dormancy of normal hematopoietic stem cells (HSCs). In this study, we show that MF-MKs elaborated significantly greater levels of TGF-β1 than TGF-β2 and TGF-β3 to a varying degree, and we evaluated the ability of AVID200, a potent TGF-β1/TGF-β3 protein trap, to block the excessive TGF-β signaling. Treatment of human mesenchymal stromal cells with AVID200 significantly reduced their proliferation, decreased phosphorylation of SMAD2, and interfered with the ability of TGF-β1 to induce collagen expression. Moreover, treatment of MF mononuclear cells with AVID200 led to increased numbers of progenitor cells (PCs) with WT JAK2 rather than mutated JAK2V617F. This effect of AVID200 on MF PCs was attributed to its ability to block TGF-β1–induced p57Kip2 expression and SMAD2 activation, thereby allowing normal rather than MF PCs to preferentially proliferate and form hematopoietic colonies. To assess the in vivo effects of AVID200, Gata1lo mice, a murine model of MF, were treated with AVID200, resulting in the reduction in BM fibrosis and an increase in BM cellularity. AVID200 treatment also increased the frequency and numbers of murine progenitor cells as well as short-term and long-term HSCs. Collectively, these data provide the rationale for TGF-β1 blockade, with AVID200 as a therapeutic strategy for patients with MF.
Collapse
Affiliation(s)
- Lilian Varricchio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Camelia Iancu-Rubin
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Bhaskar Upadhyaya
- Human Immune Monitoring Core, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | | | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Verachi
- Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy
| | - Cara Clementelli
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | | | - Adeeb H Rahman
- Human Immune Monitoring Core, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | | | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Ruben A Mesa
- Hematology Oncology, Mays Cancer Center, San Antonio, United States of America
| | | | | | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
| |
Collapse
|
24
|
Morsia E, Gangat N. Myelofibrosis: challenges for preclinical models and emerging therapeutic targets. Expert Opin Ther Targets 2021; 25:211-222. [PMID: 33844952 DOI: 10.1080/14728222.2021.1915992] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Myelofibrosis (MF) is characterized by anemia, splenomegaly, constitutional symptoms and bone marrow fibrosis. MF has no curative treatment to date, except for a small subset of patients that are eligible for allogeneic hematopoietic stem cell transplant. The discovery in recent years of the MF mutational landscape and the role of bone marrow microenvironment in disease pathogenesis has led to further insights into disease biology and consequentially rationally derived therapies.Areas covered: We searched PubMed/Medline/American Society of Hematology (ASH) abstracts until November 2020 using the following terms: myelofibrosis, mouse models, pre-clinical studies and clinical trials. The development of targeted therapies is aimed to modify the history of the disease. Although JAK inhibitors showed encouraging results in terms of spleen and symptoms response, long term remissions and disease modifying ability is lacking. Beyond JAK inhibitors, a range of agents targeting proliferative, metabolic, apoptotic pathways, the microenvironment, epigenetic modification and immunomodulation are in various stages of investigations. We review pre-clinical data, preliminary clinical results of these agents, and finally offer insights on the management of MF patients.Expert opinion: MF patients refractory or with suboptimal response to JAK inhibitors, may be managed by addition of agents with differing mechanisms, such as bromodomain (BET), lysine demethylase 1 (LSD1), MDM2, or Bcl-Xl inhibitors which could prevent emergence of resistance. Immunotherapies as long-acting interferons, and calreticulin directed antibodies or peptide vaccination are eagerly awaited. Historically, therapeutic challenges in MF have arisen due to the fact that rationally derived therapies that are based on murine models have limited impact on fibrosis and underlying disease biology in human studies, the latter illustrates the complex multi-faceted disease pathogenesis of MF. Together, we not only suggest individualized therapy in MF that is guided by genomic signature but also its early implementation potentially in prefibrotic MF.
Collapse
Affiliation(s)
- Erika Morsia
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
25
|
Song H, Liu J, Tian X, Liu D, Li J, Zhao X, Mei Z, Yan C, Han Y. Thrombopoietic effects of CCAAT/enhancer-binding protein β on the early-stage differentiation of megakaryocytes. Arch Biochem Biophys 2021; 703:108846. [PMID: 33744198 DOI: 10.1016/j.abb.2021.108846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/25/2021] [Accepted: 03/11/2021] [Indexed: 11/30/2022]
Abstract
CCAAT/enhancer-binding protein β (C/EBPβ) is a transcription factor that is involved in adipocytic and monocytic differentiation. However, the physiological role of C/EBPβ in megakaryocytes (MKs) is not clear. In this study, we investigated the effects of C/EBPβ on the early-stage differentiation of MKs, and explored the potential mechanisms of action. We established a cytosine arabinoside-induced thrombocytopenia mouse model using C57BL/6 mice. In the thrombocytopenia mice, the platelet count was found to be decreased, and the mRNA and protein expression levels of C/EBPβ in MKs were also reduced. Furthermore, the maturation of Dami (MKs cell line) cells was induced by phorbol 12-myristate 13-acetate. When C/EBPβ was silenced in Dami cells by transfection using C/EBPβ-small interfering RNA, the expression of MKs-specific markers CD41 and CD62P, was dramatically decreased, resulting in morphological changes and differentiation retardation in low ploidy, which were evaluated using flow cytometry, real-time polymerase chain reaction, western blot, and confocal microscopy. The mitogen activated protein kinase-extracellular signal-regulated kinase signaling pathway was found to be required for the differentiation of MKs; knockdown of C/EBPβ in MEK/ERK1/2 pathway attenuated MKs differentiation. Overexpression of C/EBPβ in MEK/ERK1/2 pathway inhibited by U0126 did not promote MKs differentiation. To the best of our knowledge, C/EBPβ plays an important role in MKs differentiation and polyploidy cell cycle control. Taken together, C/EBPβ may have thrombopoietic effects in the differentiation of MKs, and may assist in the development of treatments for various disorders.
Collapse
Affiliation(s)
- HaiXu Song
- Air Force Medical University, Xi'an, China
| | - Jiahao Liu
- Xiamen Special Service Health Center of the Army, Xiamen, China
| | - Xiaoxiang Tian
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Dan Liu
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Jiayin Li
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaojie Zhao
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Zhu Mei
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China
| | - Yaling Han
- Air Force Medical University, Xi'an, China; Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, Shenyang, China.
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Pemmaraju N, Chen NC, Verstovsek S. Immunotherapy and Immunomodulation in Myeloproliferative Neoplasms. Hematol Oncol Clin North Am 2021; 35:409-429. [PMID: 33641877 DOI: 10.1016/j.hoc.2020.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Myeloproliferative neoplasms are characterized by chronic inflammation. The discovery of constitutively active JAK-STAT signaling associated with driver mutations has led to clinical and translational breakthroughs. Insights into the other pathways and novel factors of potential importance are being actively investigated. Various classes of agents with immunomodulating or immunosuppressive properties have been used with varying degrees of success in treating myeloproliferative neoplasms. Early clinical trials are investigating the feasibility, effectiveness, and safety of immune checkpoint inhibitors, cell-based immunotherapies, and SMAC mimetics. The dynamic landscape of immunotherapy and immunomodulation in myeloproliferative neoplasms is the topic of the present review.
Collapse
Affiliation(s)
- Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard #3000, Houston, TX 77030, USA.
| | - Natalie C Chen
- Department of Internal Medicine, The University of Texas School of Health Sciences at Houston, 6431 Fannin, MSB 1.150, Houston, TX 77030, USA
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard #428, Houston, TX 77030, USA
| |
Collapse
|
29
|
The extracellular matrix: A key player in the pathogenesis of hematologic malignancies. Blood Rev 2020; 48:100787. [PMID: 33317863 DOI: 10.1016/j.blre.2020.100787] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/10/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022]
Abstract
Hematopoietic stem and progenitor cells located in the bone marrow lay the foundation for multiple lineages of mature hematologic cells. Bone marrow niches are architecturally complex with specific cellular, physiochemical, and biomechanical factors. Increasing evidence suggests that the bone marrow microenvironment contributes to the pathogenesis of hematological neoplasms. Numerous studies have deciphered the role of genetic mutations and chromosomal translocations in the development hematologic malignancies. Significant progress has also been made in understanding how the cellular components and cytokine interactions within the bone marrow microenvironment promote the evolution of hematologic cancers. Although the extracellular matrix is known to be a key player in the pathogenesis of various diseases, it's role in the progression of hematologic malignancies is less understood. In this review, we discuss the interactions between the extracellular matrix and malignant cells, and provide an overview of the role of extracellular matrix remodeling in sustaining hematologic malignancies.
Collapse
|
30
|
Piasecki A, Leiva O, Ravid K. Lysyl oxidase inhibition in primary myelofibrosis: A renewed strategy. ARCHIVES OF STEM CELL AND THERAPY 2020; 1:23-27. [PMID: 33738462 PMCID: PMC7968867 DOI: 10.46439/stemcell.1.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Primary myelofibrosis (PMF) is a type of myeloproliferative neoplasm (MPN) that portends a poor prognosis and has limited options for treatment. PMF is often driven by clonal mutations in one of three genes that regulate the JAK-STAT signaling pathway, leading to hyperactivation of this signaling pathway and over-proliferation of megakaryocytes (MKs) and their precursors. PMF presents with debilitating symptoms such as splenomegaly and weight loss. The few available treatments for PMF include a JAK2 inhibitor, ruxolitinib, which causes side effects and is not always effective. The extracellular matrix (ECM) and bone marrow (BM) microenvironment may play an important role in the pathogenesis of PMF. Lysyl oxidase (LOX), an enzyme that plays a key role in the ECM by facilitating the cross-linking of collagen and elastin fibers, has been shown to be upregulated in MKs of PMF mice and in PMF patients, suggesting its role in the progression of BM fibrosis. Recently, LOX has been identified as a potential novel therapeutic target for PMF and the development of new small molecule LOX inhibitors, PXS-LOX_1 and PXS-LOX_2, has shown some promise in slowing the progression of PMF in pre-clinical studies. Given that these inhibitors displayed an ability to target the dysregulation of the ECM via LOX inhibition, they show promise as therapeutic agents for an underappreciated aspect of PMF.
Collapse
Affiliation(s)
- Andrew Piasecki
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston MA 02118, USA
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Orly Leiva
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston MA 02118, USA
- Author for correspondence:
| |
Collapse
|
31
|
Jutzi JS, Mullally A. Remodeling the Bone Marrow Microenvironment - A Proposal for Targeting Pro-inflammatory Contributors in MPN. Front Immunol 2020; 11:2093. [PMID: 32983162 PMCID: PMC7489333 DOI: 10.3389/fimmu.2020.02093] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/31/2020] [Indexed: 12/31/2022] Open
Abstract
Philadelphia-negative myeloproliferative neoplasms (MPN) are malignant bone marrow (BM) disorders, typically arising from a single somatically mutated hematopoietic stem cell. The most commonly mutated genes, JAK2, CALR, and MPL lead to constitutively active JAK-STAT signaling. Common clinical features include myeloproliferation, splenomegaly and constitutional symptoms. This review covers the contributions of cellular components of MPN pathology (e.g., monocytes, megakaryocytes, and mesenchymal stromal cells) as well as cytokines and soluble mediators to the development of myelofibrosis (MF) and highlights recent therapeutic advances. These findings outline the importance of malignant and non-malignant BM constituents to the pathogenesis and treatment of MF.
Collapse
Affiliation(s)
- Jonas Samuel Jutzi
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States.,Cancer Program, Broad Institute, Cambridge, MA, United States
| |
Collapse
|
32
|
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
|
33
|
Laczko R, Csiszar K. Lysyl Oxidase (LOX): Functional Contributions to Signaling Pathways. Biomolecules 2020; 10:biom10081093. [PMID: 32708046 PMCID: PMC7465975 DOI: 10.3390/biom10081093] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Cu-dependent lysyl oxidase (LOX) plays a catalytic activity-related, primary role in the assembly of the extracellular matrix (ECM), a dynamic structural and regulatory framework which is essential for cell fate, differentiation and communication during development, tissue maintenance and repair. LOX, additionally, plays both activity-dependent and independent extracellular, intracellular and nuclear roles that fulfill significant functions in normal tissues, and contribute to vascular, cardiac, pulmonary, dermal, placenta, diaphragm, kidney and pelvic floor disorders. LOX activities have also been recognized in glioblastoma, diabetic neovascularization, osteogenic differentiation, bone matrix formation, ligament remodeling, polycystic ovary syndrome, fetal membrane rupture and tumor progression and metastasis. In an inflammatory context, LOX plays a role in diminishing pluripotent mesenchymal cell pools which are relevant to the pathology of diabetes, osteoporosis and rheumatoid arthritis. Most of these conditions involve mechanisms with complex cell and tissue type-specific interactions of LOX with signaling pathways, not only as a regulatory target, but also as an active player, including LOX-mediated alterations of cell surface receptor functions and mutual regulatory activities within signaling loops. In this review, we aim to provide insight into the diverse ways in which LOX participates in signaling events, and explore the mechanistic details and functional significance of the regulatory and cross-regulatory interactions of LOX with the EGFR, PDGF, VEGF, TGF-β, mechano-transduction, inflammatory and steroid signaling pathways.
Collapse
|
34
|
Matsuura S, Thompson CR, Ng SK, Ward CM, Karagianni A, Mazzeo C, Malara A, Balduini A, Ravid K. Adhesion to fibronectin via α5β1 integrin supports expansion of the megakaryocyte lineage in primary myelofibrosis. Blood 2020; 135:2286-2291. [PMID: 32294178 PMCID: PMC7316217 DOI: 10.1182/blood.2019004230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/10/2020] [Indexed: 12/11/2022] Open
Abstract
Excessive accumulation of extracellular matrix (ECM) is a hallmark of bone marrow (BM) milieu in primary myelofibrosis (PMF). Because cells have the ability to adhere to the surrounding ECM through integrin receptors, we examined the hypothesis that an abnormal ECM-integrin receptor axis contributes to BM megakaryocytosis in JAK2V617F+ PMF. Secretion of ECM protein fibronectin (FN) by BM stromal cells from PMF patients correlates with fibrosis and disease severity. Here, we show that Vav1-hJAK2V617F transgenic mice (JAK2V617F+) have high BM FN content associated with megakaryocytosis and fibrosis. Further, megakaryocytes from JAK2V617F+ mice have increased cell surface expression of the α5 subunit of the α5β1 integrin, the major FN receptor in megakaryocytes, and augmented adhesion to FN compared with wild-type controls. Reducing adhesion to FN by an inhibitory antibody to the α5 subunit effectively reduces the percentage of CD41+ JAK2V617F+ megakaryocytes in vitro and in vivo. Corroborating our findings in mice, JAK2V617F+ megakaryocytes from patients showed elevated expression of α5 subunit, and a neutralizing antibody to α5 subunit reduced adhesion to FN and megakaryocyte number derived from CD34+ cells. Our findings reveal a previously unappreciated contribution of FN-α5β1 integrin to megakaryocytosis in JAK2V617F+ PMF.
Collapse
Affiliation(s)
- Shinobu Matsuura
- Department of Medicine-Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
| | - Cristal Reyna Thompson
- Department of Medicine-Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
| | - Seng Kah Ng
- Department of Medicine-Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
| | - Christina Marie Ward
- Department of Medicine-Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
| | - Aikaterini Karagianni
- Department of Medicine-Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
- Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Carla Mazzeo
- Department of Medicine-Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
| | | | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, MA; and
| | - Katya Ravid
- Department of Medicine-Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
- Departments of Biochemistry and Biology, Boston University School of Medicine, Boston, MA
| |
Collapse
|
35
|
Psaila B, Wang G, Rodriguez-Meira A, Li R, Heuston EF, Murphy L, Yee D, Hitchcock IS, Sousos N, O'Sullivan J, Anderson S, Senis YA, Weinberg OK, Calicchio ML, Iskander D, Royston D, Milojkovic D, Roberts I, Bodine DM, Thongjuea S, Mead AJ. Single-Cell Analyses Reveal Megakaryocyte-Biased Hematopoiesis in Myelofibrosis and Identify Mutant Clone-Specific Targets. Mol Cell 2020; 78:477-492.e8. [PMID: 32386542 PMCID: PMC7217381 DOI: 10.1016/j.molcel.2020.04.008] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 02/04/2020] [Accepted: 04/06/2020] [Indexed: 12/20/2022]
Abstract
Myelofibrosis is a severe myeloproliferative neoplasm characterized by increased numbers of abnormal bone marrow megakaryocytes that induce fibrosis, destroying the hematopoietic microenvironment. To determine the cellular and molecular basis for aberrant megakaryopoiesis in myelofibrosis, we performed single-cell transcriptome profiling of 135,929 CD34+ lineage- hematopoietic stem and progenitor cells (HSPCs), single-cell proteomics, genomics, and functional assays. We identified a bias toward megakaryocyte differentiation apparent from early multipotent stem cells in myelofibrosis and associated aberrant molecular signatures. A sub-fraction of myelofibrosis megakaryocyte progenitors (MkPs) are transcriptionally similar to healthy-donor MkPs, but the majority are disease specific, with distinct populations expressing fibrosis- and proliferation-associated genes. Mutant-clone HSPCs have increased expression of megakaryocyte-associated genes compared to wild-type HSPCs, and we provide early validation of G6B as a potential immunotherapy target. Our study paves the way for selective targeting of the myelofibrosis clone and illustrates the power of single-cell multi-omics to discover tumor-specific therapeutic targets and mediators of tissue fibrosis.
Collapse
Affiliation(s)
- Bethan Psaila
- Haematopoietic Stem Cell Biology Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford OX3 9DS, UK; MRC Molecular Haematology Unit, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK; Hematopoiesis Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-4442, USA.
| | - Guanlin Wang
- Haematopoietic Stem Cell Biology Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford OX3 9DS, UK; MRC Molecular Haematology Unit, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK; MRC WIMM Centre for Computational Biology, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK
| | - Alba Rodriguez-Meira
- Haematopoietic Stem Cell Biology Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford OX3 9DS, UK; MRC Molecular Haematology Unit, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK; MRC WIMM Centre for Computational Biology, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK
| | - Rong Li
- Haematopoietic Stem Cell Biology Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford OX3 9DS, UK; MRC Molecular Haematology Unit, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK
| | - Elisabeth F Heuston
- Hematopoiesis Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-4442, USA
| | - Lauren Murphy
- Haematopoietic Stem Cell Biology Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford OX3 9DS, UK; MRC Molecular Haematology Unit, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK
| | - Daniel Yee
- York Biomedical Research Institute and Department of Biology, University of York, Heslington, York YO10 5DD, UK
| | - Ian S Hitchcock
- York Biomedical Research Institute and Department of Biology, University of York, Heslington, York YO10 5DD, UK
| | - Nikolaos Sousos
- Haematopoietic Stem Cell Biology Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford OX3 9DS, UK; MRC Molecular Haematology Unit, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK
| | - Jennifer O'Sullivan
- Haematopoietic Stem Cell Biology Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford OX3 9DS, UK; MRC Molecular Haematology Unit, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK
| | - Stacie Anderson
- NHGRI Flow Cytometry Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-4442, USA
| | - Yotis A Senis
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche-S 1255, Etablissement Français du Sang Grand Est, Strasbourg 67065, France
| | - Olga K Weinberg
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Monica L Calicchio
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Deena Iskander
- Centre for Haematology, Hammersmith Hospital, Imperial College of Medicine, London W12 OHS, UK
| | - Daniel Royston
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Dragana Milojkovic
- Centre for Haematology, Hammersmith Hospital, Imperial College of Medicine, London W12 OHS, UK
| | - Irene Roberts
- MRC Molecular Haematology Unit, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK; Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK
| | - David M Bodine
- Hematopoiesis Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-4442, USA
| | - Supat Thongjuea
- NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK; MRC WIMM Centre for Computational Biology, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK.
| | - Adam J Mead
- Haematopoietic Stem Cell Biology Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford OX3 9DS, UK; MRC Molecular Haematology Unit, MRC WIMM, University of Oxford, Oxford OX3 9DS, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK.
| |
Collapse
|
36
|
Matrix Mechanosensation in the Erythroid and Megakaryocytic Lineages. Cells 2020; 9:cells9040894. [PMID: 32268541 PMCID: PMC7226728 DOI: 10.3390/cells9040894] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 12/21/2022] Open
Abstract
The biomechanical properties of the bone marrow microenvironment emerge from a combination of interactions between various extracellular matrix (ECM) structural proteins and soluble factors. Matrix stiffness directs stem cell fate, and both bone marrow stromal and hematopoietic cells respond to biophysical cues. Within the bone marrow, the megakaryoblasts and erythroblasts are thought to originate from a common progenitor, giving rise to fully mature magakaryocytes (the platelet precursors) and erythrocytes. Erythroid and megakaryocytic progenitors sense and respond to the ECM through cell surface adhesion receptors such as integrins and mechanosensitive ion channels. While hematopoietic stem progenitor cells remain quiescent on stiffer ECM substrates, the maturation of the erythroid and megakaryocytic lineages occurs on softer ECM substrates. This review surveys the major matrix structural proteins that contribute to the overall biomechanical tone of the bone marrow, as well as key integrins and mechanosensitive ion channels identified as ECM sensors in context of megakaryocytosis or erythropoiesis.
Collapse
|
37
|
Zingariello M, Martelli F, Verachi P, Bardelli C, Gobbo F, Mazzarini M, Migliaccio AR. Novel targets to cure primary myelofibrosis from studies on Gata1 low mice. IUBMB Life 2019; 72:131-141. [PMID: 31749302 DOI: 10.1002/iub.2198] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/24/2019] [Indexed: 01/06/2023]
Abstract
In 2002, we discovered that mice carrying the hypomorphic Gata1low mutation that reduces expression of the transcription factor GATA1 in megakaryocytes (Gata1low mice) develop myelofibrosis, a phenotype that recapitulates the features of primary myelofibrosis (PMF), the most severe of the Philadelphia-negative myeloproliferative neoplasms (MPNs). At that time, this discovery had a great impact on the field because mutations driving the development of PMF had yet to be discovered. Later studies identified that PMF, as the others MPNs, is associated with mutations activating the thrombopoietin/JAK2 axis raising great hope that JAK inhibitors may be effective to treat the disease. Unfortunately, ruxolitinib, the JAK1/2 inhibitor approved by FDA and EMEA for PMF, ameliorates symptoms but does not improve the natural course of the disease, and the cure of PMF is still an unmet clinical need. Although GATA1 is not mutated in PMF, reduced GATA1 content in megakaryocytes as a consequence of ribosomal deficiency is a hallmark of myelofibrosis (both in humans and mouse models) and, in fact, a driving event in the disease. Conversely, mice carrying the hypomorphic Gata1low mutation express an activated TPO/JAK2 pathway and partially respond to JAK inhibitors in a fashion similar to PMF patients (reduction of spleen size but limited improvement of the natural history of the disease). These observations cross-validated Gata1low mice as a bona fide animal model for PMF and prompted the use of this model to identify abnormalities that might be targeted to cure the disease. We will summarize here data generated in Gata1low mice indicating that the TGF-β/P-selectin axis is abnormal in PMF and represents a novel target for its treatment.
Collapse
Affiliation(s)
- Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | | | - Paola Verachi
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Claudio Bardelli
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Francesca Gobbo
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Maria Mazzarini
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy
| | - Anna Rita Migliaccio
- Department of Biological and Neurobiological Medicine, University of Bologna, Bologna, Italy.,Myeloproliferative Neoplasms Research Consortium, New York, New York
| |
Collapse
|
38
|
Leiva O, Ng SK, Matsuura S, Chitalia V, Lucero H, Findlay A, Turner C, Jarolimek W, Ravid K. Novel lysyl oxidase inhibitors attenuate hallmarks of primary myelofibrosis in mice. Int J Hematol 2019; 110:699-708. [PMID: 31637674 DOI: 10.1007/s12185-019-02751-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 12/22/2022]
Abstract
Primary myelofibrosis (PMF) is a chronic myeloproliferative neoplasm (MPN) that usually portends a poor prognosis with limited therapeutic options available. Currently, only allogeneic stem cell transplantation is curative in those who are candidates, while administration of the JAK1/2 inhibitor ruxolitinib carries a risk of worsening cytopenia. The limited therapeutic options available highlight the need for the development of novel treatments for PMF. Lysyl oxidase (LOX), an enzyme vital for collagen cross-linking and extracellular matrix stiffening, has been found to be upregulated in PMF. Herein, we evaluate two novel LOX inhibitors, PXS-LOX_1 and PXS-LOX_2, in two animal models of PMF (GATA1low and JAK2V617F-mutated mice). Specifically, PXS-LOX_1 or vehicle was given to 15- to 16-week-old GATA1low mice via intraperitoneal injection at a dose of 15 mg/kg four times a week for 9 weeks. PXS-LOX_1 was found to significantly decrease the bone marrow fibrotic burden and megakaryocyte number compared to vehicle in both male and female GATA1low mice. Given these results, PXS-LOX_1 was then tested in 15- to 17-week-old JAK2V617F-mutated mice at a dose of 30 mg/kg four times a week for 8 weeks. Again, we observed a significant decrease in bone marrow fibrotic burden. PXS-LOX_2, a LOX inhibitor with improved oral bioavailability, was next evaluated in 15- to 17-week-old JAK2V617F-mutated mice at a dose of 5 mg/kg p.o. four times a week for 8 weeks. This inhibitor also resulted in a significant decrease in bone marrow fibrosis, albeit with a more pronounced amelioration in female mice. Taking these results together, PXS-LOX_1 and PXS-LOX_2 appear to be promising new candidates for the treatment of fibrosis in PMF.
Collapse
Affiliation(s)
- Orly Leiva
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 700 Albany St., W-6, Boston, MA, 02118, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Seng Kah Ng
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 700 Albany St., W-6, Boston, MA, 02118, USA
| | - Shinobu Matsuura
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 700 Albany St., W-6, Boston, MA, 02118, USA
| | - Vipul Chitalia
- Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Hector Lucero
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 700 Albany St., W-6, Boston, MA, 02118, USA
| | - Alison Findlay
- Pharmaxis Ltd., 20 Rodborough Road, Frenchs Forest, NSW, Australia
| | - Craig Turner
- Pharmaxis Ltd., 20 Rodborough Road, Frenchs Forest, NSW, Australia
| | | | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 700 Albany St., W-6, Boston, MA, 02118, USA.
| |
Collapse
|
39
|
Dynamins 2 and 3 control the migration of human megakaryocytes by regulating CXCR4 surface expression and ITGB1 activity. Blood Adv 2019; 2:3540-3552. [PMID: 30538113 DOI: 10.1182/bloodadvances.2018021923] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/04/2018] [Indexed: 12/14/2022] Open
Abstract
Megakaryocyte (MK) migration from the bone marrow periosteal niche toward the vascular niche is a prerequisite for proplatelet extension and release into the circulation. The mechanism for this highly coordinated process is poorly understood. Here we show that dynasore (DNSR), a small-molecule inhibitor of dynamins (DNMs), or short hairpin RNA knockdown of DNM2 and DNM3 impairs directional migration in a human MK cell line or MKs derived from cultured CD34+ cells. Because cell migration requires actin cytoskeletal rearrangements, we measured actin polymerization and the activity of cytoskeleton regulator RhoA and found them to be decreased after inhibition of DNM2 and DNM3. Because SDF-1α is important for hematopoiesis, we studied the expression of its receptor CXCR4 in DNSR-treated cells. CXCR4 expression on the cell surface was increased, at least partially because of slower endocytosis and internalization after SDF-1α treatment. Combined inhibition of DNM2 and DNM3 or forced expression of dominant-negative Dnm2-K44A or GTPase-defective DNM3 diminished β1 integrin (ITGB1) activity. DNSR-treated MKs showed an abnormally clustered staining pattern of Rab11, a marker of recycling endosomes. This suggests decreased recruitment of the recycling pathway in DNSR-treated cells. Altogether, we show that the GTPase activity of DNMs, which governs endocytosis and regulates cell receptor trafficking, exerts control on MK migration toward SDF-1α gradients, such as those originating from the vascular niche. DNMs play a critical role in MKs by triggering membrane-cytoskeleton rearrangements downstream of CXCR4 and integrins.
Collapse
|
40
|
Eran Z, Zingariello M, Bochicchio MT, Bardelli C, Migliaccio AR. Novel strategies for the treatment of myelofibrosis driven by recent advances in understanding the role of the microenvironment in its etiology. F1000Res 2019; 8:F1000 Faculty Rev-1662. [PMID: 31583083 PMCID: PMC6758840 DOI: 10.12688/f1000research.18581.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/12/2019] [Indexed: 12/12/2022] Open
Abstract
Myelofibrosis is the advanced stage of the Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs), characterized by systemic inflammation, hematopoietic failure in the bone marrow, and development of extramedullary hematopoiesis, mainly in the spleen. The only potentially curative therapy for this disease is hematopoietic stem cell transplantation, an option that may be offered only to those patients with a compatible donor and with an age and functional status that may face its toxicity. By contrast, with the Philadelphia-positive MPNs that can be dramatically modified by inhibitors of the novel BCR-ABL fusion-protein generated by its genetic lesion, the identification of the molecular lesions that lead to the development of myelofibrosis has not yet translated into a treatment that can modify the natural history of the disease. Therefore, the cure of myelofibrosis remains an unmet clinical need. However, the excitement raised by the discovery of the genetic lesions has inspired additional studies aimed at elucidating the mechanisms driving these neoplasms towards their final stage. These studies have generated the feeling that the cure of myelofibrosis will require targeting both the malignant stem cell clone and its supportive microenvironment. We will summarize here some of the biochemical alterations recently identified in MPNs and the novel therapeutic approaches currently under investigation inspired by these discoveries.
Collapse
Affiliation(s)
- Zimran Eran
- Department of Hematology, Hadassah University Center, Jerusalem, Israel
| | - Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Maria Teresa Bochicchio
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.), IRCCS, Meldola (FC), Italy
| | - Claudio Bardelli
- Dipartimento di Scienze Biomediche e NeuroMotorie, Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| | - Anna Rita Migliaccio
- Dipartimento di Scienze Biomediche e NeuroMotorie, Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| |
Collapse
|
41
|
Lysyl oxidases: from enzyme activity to extracellular matrix cross-links. Essays Biochem 2019; 63:349-364. [DOI: 10.1042/ebc20180050] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/23/2019] [Accepted: 08/27/2019] [Indexed: 12/13/2022]
Abstract
AbstractThe lysyl oxidase family comprises five members in mammals, lysyl oxidase (LOX) and four lysyl oxidase like proteins (LOXL1-4). They are copper amine oxidases with a highly conserved catalytic domain, a lysine tyrosylquinone cofactor, and a conserved copper-binding site. They catalyze the first step of the covalent cross-linking of the extracellular matrix (ECM) proteins collagens and elastin, which contribute to ECM stiffness and mechanical properties. The role of LOX and LOXL2 in fibrosis, tumorigenesis, and metastasis, including changes in their expression level and their regulation of cell signaling pathways, have been extensively reviewed, and both enzymes have been identified as therapeutic targets. We review here the molecular features and three-dimensional structure/models of LOX and LOXLs, their role in ECM cross-linking, and the regulation of their cross-linking activity by ECM proteins, proteoglycans, and by inhibitors. We also make an overview of the major ECM cross-links, because they are the ultimate molecular readouts of LOX/LOXL activity in tissues. The recent 3D model of LOX, which recapitulates its known structural and biochemical features, will be useful to decipher the molecular mechanisms of LOX interaction with its various substrates, and to design substrate-specific inhibitors, which are potential antifibrotic and antitumor drugs.
Collapse
|
42
|
Leiva O, Bekendam RH, Garcia BD, Thompson C, Cantor A, Chitalia V, Ravid K. Emerging Factors Implicated in Fibrotic Organ-Associated Thrombosis: The Case of Two Organs. TH OPEN 2019; 3:e165-e170. [PMID: 31259299 PMCID: PMC6598088 DOI: 10.1055/s-0039-1692204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022] Open
Abstract
Thrombosis is at the heart of cardiovascular complications observed in specific diseases. A heightened thrombosis risk above that in general population in diseases such as myelofibrosis and chronic kidney disease implicates disease-specific mediators of thrombosis. This relative lack of information regarding the mechanisms of thrombosis in specific organ pathologies hitherto has remained limited. Evolving literature implicates some soluble factors in the blood of patients with discrete disorders, inflicting fundamental changes in the components of thrombosis. In this era of precision medicine, integrating these disease-specific factors in a comprehensive thrombotic risk assessment of patients is imperative in guiding therapeutic decisions. A complex network of mechanisms regulates each organ pathology and resultant thrombotic phenotypes. This review surveys different effectors of thrombogenicity associated with two pathologically fibrotic organs used as model systems, the bone marrow and kidney, as well as focuses attention to a common inducer of fibrosis and thrombosis, lysyl oxidase.
Collapse
Affiliation(s)
- Orly Leiva
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Roelof H Bekendam
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Brenda D Garcia
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States.,Department of Medicine, Mount Auburn Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Cristal Thompson
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States.,Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Alan Cantor
- Children's Hospital Boston, Boston, Massachusetts, United States
| | - Vipul Chitalia
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States.,Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States.,VA Boston Healthcare System, Boston, Massachusetts, United States
| | - Katya Ravid
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States.,Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States
| |
Collapse
|
43
|
Noetzli LJ, French SL, Machlus KR. New Insights Into the Differentiation of Megakaryocytes From Hematopoietic Progenitors. Arterioscler Thromb Vasc Biol 2019; 39:1288-1300. [PMID: 31043076 PMCID: PMC6594866 DOI: 10.1161/atvbaha.119.312129] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/22/2019] [Indexed: 02/07/2023]
Abstract
Megakaryocytes are hematopoietic cells, which are responsible for the production of blood platelets. The traditional view of megakaryopoiesis describes the cellular journey from hematopoietic stem cells, through a hierarchical series of progenitor cells, ultimately to a mature megakaryocyte. Once mature, the megakaryocyte then undergoes a terminal maturation process involving multiple rounds of endomitosis and cytoplasmic restructuring to allow platelet formation. However, recent studies have begun to redefine this hierarchy and shed new light on alternative routes by which hematopoietic stem cells are differentiated into megakaryocytes. In particular, the origin of megakaryocytes, including the existence and hierarchy of megakaryocyte progenitors, has been redefined, as new studies are suggesting that hematopoietic stem cells originate as megakaryocyte-primed and can bypass traditional lineage checkpoints. Overall, it is becoming evident that megakaryopoiesis does not only occur as a stepwise process, but is dynamic and adaptive to biological needs. In this review, we will reexamine the canonical dogmas of megakaryopoiesis and provide an updated framework for interpreting the roles of traditional pathways in the context of new megakaryocyte biology. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Leila J Noetzli
- Division of Hematology, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Shauna L French
- Division of Hematology, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Kellie R Machlus
- Division of Hematology, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
44
|
Mechanism for oral tumor cell lysyl oxidase like-2 in cancer development: synergy with PDGF-AB. Oncogenesis 2019; 8:34. [PMID: 31086173 PMCID: PMC6513832 DOI: 10.1038/s41389-019-0144-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/04/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular lysyl oxidases (LOX and LOXL1–LOXL4) are critical for collagen biosynthesis. LOXL2 is a marker of poor survival in oral squamous cell cancer. We investigated mechanisms by which tumor cell secreted LOXL2 targets proximal mesenchymal cells to enhance tumor growth and metastasis. This study identified the first molecular mechanism for LOXL2 in the promotion of cancer via its enzymatic modification of a non-collagenous substrate in the context of paracrine signaling between tumor cells and resident fibroblasts. The role and mechanism of active LOXL2 in promoting oral cancer was evaluated and employed a novel LOXL2 small molecule inhibitor, PSX-S1C, administered to immunodeficient, and syngeneic immunocompetent orthotopic oral cancer mouse models. Tumor growth, histopathology, and metastases were monitored. In vitro mechanistic studies with conditioned tumor cell medium treatment of normal human oral fibroblasts were carried out in the presence and absence of the LOXL2 inhibitor to identify signaling mechanisms promoted by LOXL2 activity. Inhibition of LOXL2 attenuated cancer growth and lymph node metastases in the orthotopic tongue mouse models. Immunohistochemistry data indicated that LOXL2 expression in and around tumors was decreased in mice treated with the inhibitor. Inhibition of LOXL2 activity by administration of PXS-S1C to mice reduced tumor cell proliferation, accompanied by changes in morphology and in the expression of epithelial to mesenchymal transition markers. In vitro studies identified PDGFRβ as a direct substrate for LOXL2, and indicated that LOXL2 and PDGF-AB together secreted by tumor cells optimally activated PDGFRβ in fibroblasts to promote proliferation and the tendency toward fibrosis via ERK activation, but not AKT. Optimal fibroblast proliferation in vitro required LOXL2 activity, while tumor cell proliferation did not. Thus, tumor cell-derived LOXL2 in the microenvironment directly targets neighboring resident cells to promote a permissive local niche, in addition to its known role in collagen maturation.
Collapse
|
45
|
Abstract
Myelofibrosis (MF) is a myeloproliferative neoplasm that is pathologically characterized by bone marrow myeloproliferation, reticulin and collagen fibrosis, and extramedullary hematopoiesis. Constitutive activation of the Janus associated kinase (JAK)-signal transducers and activators of transcription signaling pathway with resultant elevation in pro-inflammatory cytokine levels is the pathogenic hallmark of MF. JAK inhibitors, namely ruxolitinib, have been successful in alleviating symptoms and reducing splenomegaly, but therapy-related myelosuppression has led to the further development of highly selective JAK2 inhibitors. Additionally, ruxolitinib does not appear to affect the malignant hematopoietic clone substantially, evidenced by lack of molecular remissions, bone marrow histopathologic responses, and a proportion of treated patients developing progressive disease and leukemic transformation while receiving therapy. A number of other pharmacotherapeutic strategies are currently being explored in the clinic. Non-JAK inhibitor strategies being evaluated in MF include non-JAK signaling pathway inhibitors, epigenetic-directed therapies, immune-modulating agents, anti-fibrotic agents, and telomerase inhibitors. This review highlights the current landscape of MF pharmacotherapy and explores therapeutic advances underway.
Collapse
Affiliation(s)
- Douglas Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1079, New York, NY, 10029, USA
| | - Bridget Marcellino
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1079, New York, NY, 10029, USA
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1079, New York, NY, 10029, USA.
| |
Collapse
|
46
|
Zhang J, Zhuang R, Zhang X, Hu W, Cheng K, Jiang D, Shen S, Zhang Y, Ding Y, Zhang Y. CD226 is involved in megakaryocyte activation and early-stage differentiation. Mol Immunol 2019; 107:123-131. [DOI: 10.1016/j.molimm.2019.01.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 01/21/2019] [Indexed: 12/11/2022]
|
47
|
Megakaryocyte Contribution to Bone Marrow Fibrosis: many Arrows in the Quiver. Mediterr J Hematol Infect Dis 2018; 10:e2018068. [PMID: 30416700 PMCID: PMC6223581 DOI: 10.4084/mjhid.2018.068] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/23/2018] [Indexed: 01/14/2023] Open
Abstract
In Primary Myelofibrosis (PMF), megakaryocyte dysplasia/hyperplasia determines the release of inflammatory cytokines that, in turn, stimulate stromal cells and induce bone marrow fibrosis. The pathogenic mechanism and the cells responsible for progression to bone marrow fibrosis in PMF are not completely understood. This review article aims to provide an overview of the crucial role of megakaryocytes in myelofibrosis by discussing the role and the altered secretion of megakaryocyte-derived soluble factors, enzymes and extracellular matrices that are known to induce bone marrow fibrosis.
Collapse
|
48
|
Shen Y, Jing D, Hao J, Tang G, Yang P, Zhao Z. The Effect of β-Aminopropionitrile on Skeletal Micromorphology and Osteogenesis. Calcif Tissue Int 2018; 103:411-421. [PMID: 29916126 DOI: 10.1007/s00223-018-0430-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/03/2018] [Indexed: 12/16/2022]
Abstract
Collagen cross-linking, as a form of collagen post-translational modification, plays a crucial role in maintaining bone mechanical properties as well as in regulating cell biological functions. Shifts in cross-links profile are found apparently correlated to kinds of skeletal pathology and diseases, whereas little is known about the relationship between collagen cross-links and osteogenesis. Here, we hypothesized that the inhibition of collagen cross-links could impair skeletal microstructure and inhibit osteogenesis. A mouse model of collagen cross-linking defects has been established using subcutaneous injection of 350 mg/kg β-aminopropionitrile (BAPN) daily for 4 weeks, and same dose of phosphate buffered saline (PBS) served as control group. The analysis of bone microstructural parameters revealed a significant decrease of bone volume fraction (BV/TV) and trabecular thickness (Tb.Th), and increase of bone surface ratio (BS/BV), structure model index (SMI) as well as trabecular separation (Tb.Sp) in the experimental group (p < 0.05), whereas there was no difference observed in bone mineral density (BMD). Histological staining displayed that the BAPN treatment caused thinner trabeculae and decrease of collagen content in proximal tibiae. The analysis of osteogenesis PCR (Polymerase Chain Reaction) array reflected that BAPN remarkably influenced the expression of Alpl, Bglap, Bgn, Bmp5, Col10a1, Col1a1, Col1a2, Col5a1, Itga2b, and Serpinh1. The results of immunohistochemistry displayed a significant reduction in the mean optical densities of OCN and COL1 at the presence of BAPN. The overall results of this study suggested that BAPN alters bone microstructure and hinders the expression of osteogenic genes without affecting mineralization processes, indicating the influences of collagen cross-links on osteogenesis may be a potential pathological mechanism in skeletal diseases.
Collapse
Affiliation(s)
- Yu Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd section of Renmin South Road, Chengdu, 610041, China
| | - Dian Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd section of Renmin South Road, Chengdu, 610041, China
| | - Jin Hao
- Harvard School of Dental Medicine, Harvard University, Cambridge, MA, USA
| | - Ge Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd section of Renmin South Road, Chengdu, 610041, China
| | - Pu Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd section of Renmin South Road, Chengdu, 610041, China.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd section of Renmin South Road, Chengdu, 610041, China.
| |
Collapse
|
49
|
Saxena D, Mahjour F, Findlay A, Mously E, Kantarci A, Trackman P. Multiple Functions of Lysyl Oxidase Like-2 in Oral Fibroproliferative Processes. J Dent Res 2018; 97:1277-1284. [PMID: 29787337 PMCID: PMC6151912 DOI: 10.1177/0022034518775971] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Gingival overgrowth is a side effect of certain medications, including calcium channel blockers, cyclosporin A, and phenytoin. Phenytoin-induced gingival overgrowth is fibrotic. Lysyl oxidases are extracellular enzymes that are required for biosynthetic cross-linking of collagens, and members of this enzyme family are upregulated in fibrosis. Previous studies in humans and in a mouse model of phenytoin-induced gingival overgrowth have shown that LOXL2 is elevated in the epithelium and connective tissue in gingival overgrowth tissues and not in normal tissues. Here, using a novel LOXL2 isoform-selective inhibitor and knockdown studies in loss- and gain-of-function studies, we investigated roles for LOXL2 in promoting cultures of human gingival fibroblasts to proliferate and to accumulate collagen. Data indicate that LOXL2 stimulates gingival fibroblast proliferation, likely by a platelet-derived growth factor B receptor-mediated mechanism. Moreover, collagen accumulation was stimulated by LOXL2 enzyme and inhibited by LOXL2 inhibitor or gene knockdown. These studies suggest that LOXL2 could serve as a potential therapeutic target to address oral fibrotic conditions.
Collapse
Affiliation(s)
- D. Saxena
- Department of Molecular and Cell Biology,
Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | - F. Mahjour
- Department of Molecular and Cell Biology,
Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| | | | - E.A. Mously
- Department of Molecular and Cell Biology,
Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
- College of Dentistry, Taibah University,
Medina, Saudi Arabia
| | | | - P.C. Trackman
- Department of Molecular and Cell Biology,
Boston University Henry M. Goldman School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
50
|
Ricard-Blum S, Baffet G, Théret N. Molecular and tissue alterations of collagens in fibrosis. Matrix Biol 2018; 68-69:122-149. [DOI: 10.1016/j.matbio.2018.02.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 02/07/2023]
|