16,380 results on '"BONE marrow"'
Search Results
2. GPR56 in GVL: marker or mechanism?
- Author
-
Iness AN and Bachireddy P
- Subjects
- Humans, T-Lymphocytes, Bone Marrow, Leukemia, Myeloid, Acute
- Published
- 2024
- Full Text
- View/download PDF
3. Targeting cancer-associated fibroblasts in the bone marrow prevents resistance to CART-cell therapy in multiple myeloma
- Author
-
Reona Sakemura, Mehrdad Hefazi, Elizabeth L. Siegler, Michelle J. Cox, Daniel P. Larson, Michael J. Hansen, Claudia Manriquez Roman, Kendall J. Schick, Ismail Can, Erin E. Tapper, Paulina Horvei, Mohamad M. Adada, Evandro D. Bezerra, Lionel Aurelien Kankeu Fonkoua, Michael W. Ruff, Wendy K. Nevala, Denise K. Walters, Sameer A. Parikh, Yi Lin, Diane F. Jelinek, Neil E. Kay, P. Leif Bergsagel, and Saad S. Kenderian
- Subjects
Immunology ,Cell- and Tissue-Based Therapy ,virus diseases ,Cell Biology ,Hematology ,Fibroblasts ,Immunotherapy, Adoptive ,Biochemistry ,Cancer-Associated Fibroblasts ,Bone Marrow ,immune system diseases ,mental disorders ,Tumor Microenvironment ,Humans ,Multiple Myeloma - Abstract
Pivotal clinical trials of B-cell maturation antigen-targeted chimeric antigen receptor T (CART)-cell therapy in patients with relapsed/refractory multiple myeloma (MM) resulted in remarkable initial responses, which led to a recent US Food and Drug Administration approval. Despite the success of this therapy, durable remissions continue to be low, and the predominant mechanism of resistance is loss of CART cells and inhibition by the tumor microenvironment (TME). MM is characterized by an immunosuppressive TME with an abundance of cancer-associated fibroblasts (CAFs). Using MM models, we studied the impact of CAFs on CART-cell efficacy and developed strategies to overcome CART-cell inhibition. We showed that CAFs inhibit CART-cell antitumor activity and promote MM progression. CAFs express molecules such as fibroblast activation protein and signaling lymphocyte activation molecule family-7, which are attractive immunotherapy targets. To overcome CAF-induced CART-cell inhibition, CART cells were generated targeting both MM cells and CAFs. This dual-targeting CART-cell strategy significantly improved the effector functions of CART cells. We show for the first time that dual targeting of both malignant plasma cells and the CAFs within the TME is a novel strategy to overcome resistance to CART-cell therapy in MM.
- Published
- 2022
- Full Text
- View/download PDF
4. Fetal vs adult megakaryopoiesis
- Author
-
Patricia Davenport, Zhi-Jian Liu, and Martha Sola-Visner
- Subjects
Adult ,Fetus ,Bone Marrow ,Megakaryopoiesis and Platelet Production ,Immunology ,Infant, Newborn ,Humans ,Cell Biology ,Hematology ,Megakaryocytes ,Biochemistry ,Megakaryocyte Progenitor Cells ,Thrombopoiesis - Abstract
Fetal and neonatal megakaryocyte progenitors are hyperproliferative compared with adult progenitors and generate a large number of small, low-ploidy megakaryocytes. Historically, these developmental differences have been interpreted as “immaturity.” However, more recent studies have demonstrated that the small, low-ploidy fetal and neonatal megakaryocytes have all the characteristics of adult polyploid megakaryocytes, including the presence of granules, a well-developed demarcation membrane system, and proplatelet formation. Thus, rather than immaturity, the features of fetal and neonatal megakaryopoiesis reflect a developmentally unique uncoupling of proliferation, polyploidization, and cytoplasmic maturation, which allows fetuses and neonates to populate their rapidly expanding bone marrow and blood volume. At the molecular level, the features of fetal and neonatal megakaryopoiesis are the result of a complex interplay of developmentally regulated pathways and environmental signals from the different hematopoietic niches. Over the past few years, studies have challenged traditional paradigms about the origin of the megakaryocyte lineage in both fetal and adult life, and the application of single-cell RNA sequencing has led to a better characterization of embryonic, fetal, and adult megakaryocytes. In particular, a growing body of data suggests that at all stages of development, the various functions of megakaryocytes are not fulfilled by the megakaryocyte population as a whole, but rather by distinct megakaryocyte subpopulations with dedicated roles. Finally, recent studies have provided novel insights into the mechanisms underlying developmental disorders of megakaryopoiesis, which either uniquely affect fetuses and neonates or have different clinical presentations in neonatal compared with adult life.
- Published
- 2022
- Full Text
- View/download PDF
5. Don’t you forget about me(gakaryocytes)
- Author
-
Isabelle C Becker, Joseph E. Italiano, and Julia Tilburg
- Subjects
Blood Platelets ,urogenital system ,Immunology ,Cell ,Cell Biology ,Hematology ,Biology ,Hematopoietic Stem Cells ,Biochemistry ,Thrombopoiesis ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Bone Marrow ,Precursor cell ,Megakaryopoiesis and Platelet Production ,medicine ,Platelet ,Bone marrow ,Stem cell ,Megakaryocytes ,Platelet factor 4 ,Transforming growth factor - Abstract
Platelets (small, anucleate cell fragments) derive from large precursor cells, megakaryocytes (MKs), that reside in the bone marrow. MKs emerge from hematopoietic stem cells in a complex differentiation process that involves cytoplasmic maturation, including the formation of the demarcation membrane system, and polyploidization. The main function of MKs is the generation of platelets, which predominantly occurs through the release of long, microtubule-rich proplatelets into vessel sinusoids. However, the idea of a 1-dimensional role of MKs as platelet precursors is currently being questioned because of advances in high-resolution microscopy and single-cell omics. On the one hand, recent findings suggest that proplatelet formation from bone marrow–derived MKs is not the only mechanism of platelet production, but that it may also occur through budding of the plasma membrane and in distant organs such as lung or liver. On the other hand, novel evidence suggests that MKs not only maintain physiological platelet levels but further contribute to bone marrow homeostasis through the release of extracellular vesicles or cytokines, such as transforming growth factor β1 or platelet factor 4. The notion of multitasking MKs was reinforced in recent studies by using single-cell RNA sequencing approaches on MKs derived from adult and fetal bone marrow and lungs, leading to the identification of different MK subsets that appeared to exhibit immunomodulatory or secretory roles. In the following article, novel insights into the mechanisms leading to proplatelet formation in vitro and in vivo will be reviewed and the hypothesis of MKs as immunoregulatory cells will be critically discussed.
- Published
- 2022
- Full Text
- View/download PDF
6. The journey of neutropoiesis: how complex landscapes in bone marrow guide continuous neutrophil lineage determination
- Author
-
Celine Overbeeke, Tamar Tak, and Leo Koenderman
- Subjects
Bone Marrow ,Neutrophils ,Immunology ,Homeostasis ,Cell Differentiation ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Biochemistry - Abstract
Neutrophils are the most abundant white blood cell, and they differentiate in homeostasis in the bone marrow from hematopoietic stem cells (HSCs) via multiple intermediate progenitor cells into mature cells that enter the circulation. Recent findings support a continuous model of differentiation in the bone marrow of heterogeneous HSCs and progenitor populations. Cell fate decisions at the levels of proliferation and differentiation are enforced through expression of lineage-determining transcription factors and their interactions, which are influenced by intrinsic (intracellular) and extrinsic (extracellular) mechanisms. Neutrophil homeostasis is subjected to positive-feedback loops, stemming from the gut microbiome, as well as negative-feedback loops resulting from the clearance of apoptotic neutrophils by mature macrophages. Finally, the cellular kinetics regarding the replenishing of the mature neutrophil pool is discussed in light of recent contradictory data.
- Published
- 2022
- Full Text
- View/download PDF
7. Endothelial cell-derived angiopoietin-like protein 2 supports hematopoietic stem cell activities in bone marrow niches
- Author
-
Zhuo Yu, Wenqian Yang, Xiaoxiao He, Chiqi Chen, Wenrui Li, Limin Zhao, Ligen Liu, Junling Liu, Li Xie, Yaping Zhang, and Junke Zheng
- Subjects
Mice ,Bone Marrow ,Immunology ,Animals ,Endothelial Cells ,Bone Marrow Cells ,Cell Biology ,Hematology ,Stem Cell Niche ,Hematopoietic Stem Cells ,Biochemistry ,Angiopoietin-Like Protein 2 - Abstract
Bone marrow niche cells have been reported to fine-tune hematopoietic stem cell (HSC) stemness via direct interaction or secreted components. Nevertheless, how niche cells control HSC activities remains largely unknown. We previously showed that angiopoietin-like protein 2 (ANGPTL2) can support the ex vivo expansion of HSCs by binding to human leukocyte immunoglobulin-like receptor B2. However, how ANGPTL2 from specific niche cell types regulates HSC activities under physiological conditions is still not clear. Herein, we generated an Angptl2-flox/flox transgenic mouse line and conditionally deleted Angptl2 expression in several niche cells, including Cdh5+ or Tie2+ endothelial cells, Prx1+ mesenchymal stem cells, and Pf4+ megakaryocytes, to evaluate its role in the regulation of HSC fate. Interestingly, we demonstrated that only endothelial cell-derived ANGPTL2 and not ANGPTL2 from other niche cell types plays important roles in supporting repopulation capacity, quiescent status, and niche localization. Mechanistically, ANGPTL2 enhances peroxisome-proliferator-activated receptor D (PPARD) expression to transactivate G0s2 to sustain the perinuclear localization of nucleolin to prevent HSCs from entering the cell cycle. These findings reveal that endothelial cell-derived ANGPTL2 serves as a critical niche component to maintain HSC stemness, which may benefit the understanding of stem cell biology in bone marrow niches and the development of a unique strategy for the ex vivo expansion of HSCs.
- Published
- 2022
- Full Text
- View/download PDF
8. Donor bone marrow–derived macrophage MHC II drives neuroinflammation and altered behavior during chronic GVHD in mice
- Author
-
Dylan Carter-Cusack, Jana Vukovic, Rachael C Adams, Bruce R. Blazar, Glen M. Boyle, Andreas Möller, Kelli P. A. MacDonald, Genesis T Llanes, Rebecca L Johnston, Samreen Shaikh, Gregory A. Quaife-Ryan, and Lambros T. Koufariotis
- Subjects
Myeloid ,Immunology ,Population ,Graft vs Host Disease ,Biochemistry ,Mice ,medicine ,Animals ,Macrophage ,education ,Neuroinflammation ,Bone Marrow Transplantation ,education.field_of_study ,Microglia ,business.industry ,Macrophages ,Histocompatibility Antigens Class II ,Cell Biology ,Hematology ,Transplantation ,medicine.anatomical_structure ,Chronic Disease ,Neuroinflammatory Diseases ,Female ,Bone marrow ,business ,CD8 - Abstract
Graft-versus-host disease (GVHD) remains the leading cause of nonrelapse mortality after allogeneic stem cell transplantation for hematological malignancies. Manifestations of GVHD in the central nervous system (CNS) present as neurocognitive dysfunction in up to 60% of patients; however, the mechanisms driving chronic GVHD (cGVHD) in the CNS are yet to be elucidated. Our studies of murine cGVHD revealed behavioral deficits associated with broad neuroinflammation and persistent Ifng upregulation. By flow cytometry, we observed a proportional shift in the donor-derived T-cell population in the cGVHD brain from early CD8 dominance to later CD4 sequestration. RNA sequencing of the hippocampus identified perturbations to structural and functional synapse-related gene expression, together with the upregulation of genes associated with interferon-γ responses and antigen presentation. Neuroinflammation in the cortex of mice and humans during acute GVHD was recently shown to be mediated by resident microglia-derived tumor necrosis factor. In contrast, infiltration of proinflammatory major histocompatibility complex (MHC) class II+ donor bone marrow (BM)–derived macrophages (BMDMs) was identified as a distinguishing feature of CNS cGVHD. Donor BMDMs, which composed up to 50% of the CNS myeloid population, exhibited a transcriptional signature distinct from resident microglia. Recipients of MHC class II knockout BM grafts exhibited attenuated neuroinflammation and behavior comparable to controls, suggestive of a critical role of donor BMDM MHC class II expression in CNS cGVHD. Our identification of disease mediators distinct from those in the acute phase indicates the necessity to pursue alternative therapeutic targets for late-stage neurological manifestations.
- Published
- 2022
- Full Text
- View/download PDF
9. Clonal hematopoiesis, myeloid disorders and BAX-mutated myelopoiesis in patients receiving venetoclax for CLL
- Author
-
Ashish Panigrahi, Constantine S. Tam, Piers Blombery, Xiangting Chen, John F. Seymour, Dennis A. Carney, Andrew H. Wei, Thomas E Lew, Mary Ann Anderson, Andrew W. Roberts, Ella R. Thompson, David C. S. Huang, Tamia Nguyen, David Westerman, Michael A. Dengler, Victor S Lin, Sasanka M. Handunnetti, and Jerry M. Adams
- Subjects
Male ,medicine.medical_specialty ,Myeloid ,Chronic lymphocytic leukemia ,Immunology ,Neutropenia ,Biochemistry ,chemistry.chemical_compound ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Aged ,bcl-2-Associated X Protein ,Aged, 80 and over ,Myelopoiesis ,Sulfonamides ,Myeloproliferative Disorders ,Hematology ,business.industry ,Venetoclax ,Myeloid leukemia ,Neoplasms, Second Primary ,Cell Biology ,Middle Aged ,Bridged Bicyclo Compounds, Heterocyclic ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,medicine.anatomical_structure ,chemistry ,Hematologic Neoplasms ,Mutation ,Cancer research ,Female ,Bone marrow ,business ,Vidarabine - Abstract
The BCL2 inhibitor venetoclax has established therapeutic roles in chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML). As BCL2 is an important determinant of survival of both myeloid progenitor and B cells, we investigated whether clinical and molecular abnormalities arise in the myeloid compartment during long-term continuous venetoclax treatment of CLL in 89 patients (87 with relapsed/refractory CLL). Over a median follow-up of 75 (range 21-98) months, persistent cytopenias (≥1 of neutropenia, thrombocytopenia, anemia) lasting ≥4 months and unrelated to CLL occurred in 25 patients (28%). Of these patients, 20 (80%) displayed clonal hematopoiesis, including 10 with therapy-related myeloid neoplasms (t-MNs). t-MNs occurred exclusively in patients previously exposed to fludarabine-alkylator combination therapy with a cumulative 5-year incidence of 10.4% after venetoclax initiation, consistent with rates reported for patients exposed to fludarabine-alkylator combination therapy without venetoclax. To determine whether the altered myelopoiesis reflected the acquisition of mutations, we analyzed samples from patients with no or minimal bone marrow CLL burden (n = 41). Mutations in the apoptosis effector BAX were identified in 32% (13/41). In cellular assays, C-terminal BAX mutants abrogated outer mitochondrial membrane localization of BAX and engendered resistance to venetoclax killing. BAX-mutated clonal hematopoiesis occurred independently of prior fludarabine-alkylator combination therapy exposure and was not associated with t-MNs. Single-cell sequencing revealed clonal co-occurrence of mutations in BAX with DNMT3A or ASXL1. We also observed simultaneous BCL2 mutations within CLL cells and BAX mutations in the myeloid compartment of the same patients, indicating lineage-specific adaptation to venetoclax therapy.
- Published
- 2022
- Full Text
- View/download PDF
10. Autophagy in mesenchymal progenitors protects mice against bone marrow failure after severe intermittent stress
- Author
-
Dirk Strunk, Terry P Yamaguchi, Sandra Romero Marquez, Rouzanna Istvanffy, Michèle C. Buck, Jennifer Rivière, Katharina Brandstetter, Franziska Hettler, Matthias Kieslinger, Akiko Shimamura, Florian Bassermann, Mehmet Sacma, Hartmut Geiger, Erik Hameister, Jürgen Ruland, Christina Schreck, Heinrich Leonhardt, Theresa Landspersky, Robert A.J. Oostendorp, Judith S. Hecker, Kasiani C. Myers, Matthias Schiemann, Marilena Götz, Romina Ludwig, Martin Wolf, and Katharina Götze
- Subjects
Stress fiber ,Immunology ,Biology ,Biochemistry ,Wnt-5a Protein ,Mice ,Autophagy ,medicine ,Animals ,Humans ,Progenitor cell ,Cells, Cultured ,Progenitor ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Hematology ,Bone Marrow Failure Disorders ,Hematopoietic Stem Cells ,Hematopoiesis ,Cell biology ,Oxidative Stress ,Haematopoiesis ,medicine.anatomical_structure ,Blood Commentary ,Bone marrow ,Stem cell - Abstract
The cellular mechanisms required to ensure homeostasis of the hematopoietic niche and the ability of this niche to support hematopoiesis upon stress remain elusive. We here identify Wnt5a in Osterix+ mesenchymal progenitor and stem cells (MSPCs) as a critical factor for niche-dependent hematopoiesis. Mice lacking Wnt5a in MSPCs suffer from stress-related bone marrow (BM) failure and increased mortality. Niche cells devoid of Wnt5a show defective actin stress fiber orientation due to an elevated activity of the small GTPase CDC42. This results in incorrect positioning of autophagosomes and lysosomes, thus reducing autophagy and increasing oxidative stress. In MSPCs from patients from BM failure states which share features of peripheral cytopenia and hypocellular BM, we find similar defects in actin stress fiber orientation, reduced and incorrect colocalization of autophagosomes and lysosomes, and CDC42 activation. Strikingly, a short pharmacological intervention to attenuate elevated CDC42 activation in vivo in mice prevents defective actin-anchored autophagy in MSPCs, salvages hematopoiesis and protects against lethal cytopenia upon stress. In summary, our study identifies Wnt5a as a restriction factor for niche homeostasis by affecting CDC42-regulated actin stress-fiber orientation and autophagy upon stress. Our data further imply a critical role for autophagy in MSPCs for adequate support of hematopoiesis by the niche upon stress and in human diseases characterized by peripheral cytopenias and hypocellular BM.
- Published
- 2022
- Full Text
- View/download PDF
11. The bone marrow niche from the inside out: how megakaryocytes are shaped by and shape hematopoiesis
- Author
-
Maria N. Barrachina, Andrew P Stone, and Thais Ferreira Nascimento
- Subjects
Myeloid ,Immunology ,Review Article ,Cell Biology ,Hematology ,Biology ,Hematopoietic Stem Cells ,Biochemistry ,Hematopoiesis ,Cell biology ,Haematopoiesis ,Paracrine signalling ,medicine.anatomical_structure ,Bone Marrow ,medicine ,Animals ,Humans ,Bone marrow ,Stem Cell Niche ,Stem cell ,Autocrine signalling ,Megakaryocytes ,Progenitor ,Megakaryopoiesis - Abstract
Megakaryocytes (MKs), the largest of the hematopoietic cells, are responsible for producing platelets by extending and depositing long proplatelet extensions into the bloodstream. The traditional view of megakaryopoiesis describes the cellular journey from hematopoietic stem cells (HSCs) along the myeloid branch of hematopoiesis. However, recent studies suggest that MKs can be generated from multiple pathways, some of which do not require transit through multipotent or bipotent MK-erythroid progenitor stages in steady-state and emergency conditions. Growing evidence suggests that these emergency conditions are due to stress-induced molecular changes in the bone marrow (BM) microenvironment, also called the BM niche. These changes can result from insults that affect the BM cellular composition, microenvironment, architecture, or a combination of these factors. In this review, we explore MK development, focusing on recent studies showing that MKs can be generated from multiple divergent pathways. We highlight how the BM niche may encourage and alter these processes using different mechanisms of communication, such as direct cell-to-cell contact, secreted molecules (autocrine and paracrine signaling), and the release of cellular components (eg, extracellular vesicles). We also explore how MKs can actively build and shape the surrounding BM niche.
- Published
- 2022
- Full Text
- View/download PDF
12. Frequency and prognostic impact of blood-circulating tumor mast cells in mastocytosis
- Author
-
Andrea Mayado, Iván Álvarez-Twose, María Jara-Acevedo, Ana Gabriela Henriques, Javier I. Muñoz-González, Laura Sánchez-Muñoz, Alba Pérez-Pons, Alberto Orfao, Carolina Caldas, Lidia Torres-Rivera, Andrés C. García-Montero, and Almudena Matito
- Subjects
Adult ,Male ,medicine.medical_specialty ,Clinical Trials and Observations ,Immunology ,CD34 ,Antigens, CD34 ,Biochemistry ,Gastroenterology ,Flow cytometry ,Young Adult ,Internal medicine ,medicine ,Humans ,Disseminated disease ,Mast Cells ,Systemic mastocytosis ,Aged ,Aged, 80 and over ,Myeloid Neoplasia ,medicine.diagnostic_test ,Cutaneous Mastocytosis ,business.industry ,Cell Biology ,Hematology ,Middle Aged ,Neoplastic Cells, Circulating ,Prognosis ,medicine.disease ,medicine.anatomical_structure ,International Prognostic Scoring System ,Biomarker (medicine) ,Female ,Bone marrow ,business ,Mastocytosis - Abstract
Circulating tumor mast cells (CTMCs) have been identified in the blood of a small number of patients with advanced systemic mastocytosis (SM). However, data are limited about their frequency and prognostic impact in patients with MC activation syndrome (MCAS), cutaneous mastocytosis (CM) and nonadvanced SM. We investigated the presence of CTMCs and MC-committed CD34+ precursors in the blood of 214 patients with MCAS, CM, or SM using highly sensitive next-generation flow cytometry. CTMCs were detected at progressively lower counts in almost all patients with advanced SM (96%) and smoldering SM (SSM; 100%), nearly half of the patients (45%) with indolent SM (ISM), and a few patients (7%) with bone marrow (BM) mastocytosis but were systematically absent in patients with CM and MCAS (P < .0001). In contrast to CTMC counts, the number of MC-committed CD34+ precursors progressively decreased from MCAS, CM, and BM mastocytosis to ISM, SSM, and advanced SM (P < .0001). Clinically, the presence (and number) of CTMCs in blood of patients with SM in general and nonadvanced SM (ISM and BM mastocytosis) in particular was associated with more adverse features of the disease, poorer-risk prognostic subgroups as defined by the International Prognostic Scoring System for advanced SM (P < .0001) and the Global Prognostic Score for mastocytosis (P < .0001), and a significantly shortened progression-free survival (P < .0001) and overall survival (P = .01). On the basis of our results, CTMCs emerge as a novel candidate biomarker of disseminated disease in SM that is strongly associated with advanced SM and poorer prognosis in patients with ISM.
- Published
- 2022
- Full Text
- View/download PDF
13. Murine AGM single-cell profiling identifies a continuum of hemogenic endothelium differentiation marked by ACE
- Author
-
Georges Lacaud, Pierre Savatier, Renaud Mevel, Syed Murtuza Baker, Irene Aksoy, Wen Hao Neo, Rahima Patel, Valerie Kouskoff, Magnus Rattray, Laura Fontenille, Roshana Thambyrajah, Nam Do Khoa, Muhammad Zaki Hidayatullah Fadlullah, and Michael Lie-A-Ling
- Subjects
Hemangioblasts ,Immunology ,Biology ,Biochemistry ,Mice ,chemistry.chemical_compound ,Dorsal aorta ,medicine ,Animals ,Zebrafish ,Hemogenic endothelium ,Hematopoietic Stem Cells/cytology ,Manchester Cancer Research Centre ,ResearchInstitutes_Networks_Beacons/mcrc ,Mesonephros/cytology ,Mesenchymal stem cell ,Cell Differentiation ,Cell Biology ,Hematology ,Embryo, Mammalian ,Hematopoietic Stem Cells ,Hematopoiesis ,Cell biology ,Transplantation ,Haematopoiesis ,Embryo, Mammalian/cytology ,medicine.anatomical_structure ,RUNX1 ,chemistry ,Mesonephros ,Bone marrow ,Hemangioblasts/cytology ,Single-Cell Analysis ,Stem cell ,Transcriptome - Abstract
In vitro generation and expansion of hematopoietic stem cells (HSCs) holds great promise for the treatment of any ailment that relies on bone marrow or blood transplantation. To achieve this, it is essential to resolve the molecular and cellular pathways that govern HSC formation in the embryo. HSCs first emerge in the aorta-gonad-mesonephros (AGM) region, where a rare subset of endothelial cells, hemogenic endothelium (HE), undergoes an endothelial-to-hematopoietic transition (EHT). Here, we present full-length single-cell RNA sequencing (scRNA-seq) of the EHT process with a focus on HE and dorsal aorta niche cells. By using Runx1b and Gfi1/1b transgenic reporter mouse models to isolate HE, we uncovered that the pre-HE to HE continuum is specifically marked by angiotensin-I converting enzyme (ACE) expression. We established that HE cells begin to enter the cell cycle near the time of EHT initiation when their morphology still resembles endothelial cells. We further demonstrated that RUNX1 AGM niche cells consist of vascular smooth muscle cells and PDGFRa+ mesenchymal cells and can functionally support hematopoiesis. Overall, our study provides new insights into HE differentiation toward HSC and the role of AGM RUNX1+ niche cells in this process. Our expansive scRNA-seq datasets represents a powerful resource to investigate these processes further.
- Published
- 2022
- Full Text
- View/download PDF
14. IL-1 mediates microbiome-induced inflammaging of hematopoietic stem cells in mice
- Author
-
Fatima Al-Shahrour, Larisa V. Kovtonyuk, Emma Slack, Hitoshi Takizawa, Markus G. Manz, Eva-Maria Manz, Patrick M. Helbling, César Nombela-Arrieta, Francisco Caiado, Steffen Boettcher, and Santiago García-Martín
- Subjects
Aging ,Myeloid ,Interleukin-1beta ,Immunology ,Population ,Biology ,Biochemistry ,Mice ,Immune system ,Interleukin-1alpha ,medicine ,Animals ,education ,Cellular Senescence ,Inflammation ,Mice, Knockout ,education.field_of_study ,Microbiota ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Hematopoiesis ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Cancer research ,Bone marrow ,Stem cell - Abstract
Aging is associated with impaired hematopoietic and immune function caused in part by decreased fitness in the hematopoietic stem cell (HSC) population and an increased myeloid differentiation bias. The reasons for this aging-associated HSC impairment are incompletely understood. Here we demonstrate that older specific pathogen free (SPF) wild-type (WT) mice in contrast to young SPF mice produce more interleukin-1a and interleukin-1b (IL-1a/b) in steady-state bone marrow (BM), with most of the IL-1a/b being derived from myeloid BM cells. Furthermore, blood from steady-state older SPF WT mice contains higher levels of microbe-associated molecular patterns, specifically TLR4 and TLR8 ligands. In addition, BM myeloid cells from older mice produce more IL-1b in vitro, and older mice show higher and more durable IL-1a/b responses upon stimulation with lipopolysaccharide in vivo. To test whether HSC aging is driven by IL-1a/b, we evaluated HSCs from IL-1 receptor 1 (IL-1R1) knockout (KO) mice. Indeed, older HSCs from IL-1R1KO mice show significantly mitigated aging-associated inflammatory signatures. Moreover, HSCs from older IL-1R1KO and from germ-free mice maintain unbiased lymphomyeloid hematopoietic differentiation upon transplantation, thus resembling this functionality of young HSCs. Importantly, in vivo antibiotic suppression of microbiota or pharmacologic blockade of IL-1 signaling in older WT mice was similarly sufficient to reverse myeloid-biased output of their HSC populations. Collectively, our data define the microbiome/IL-1/IL-1R1 axis as a key, self-sustaining and also therapeutically partially reversible driver of HSC inflammaging.
- Published
- 2022
- Full Text
- View/download PDF
15. HLFexpression defines the human hematopoietic stem cell state
- Author
-
Tara MacRae, Deanne Gracias, Nadine Mayotte, Sophie Corneau, Elisa Tomellini, Guy Sauvageau, Jalila Chagraoui, Isabel Boivin, Bernhard Lehnertz, and Aurelie Durand
- Subjects
education.field_of_study ,Immunology ,Population ,Gene Expression ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Biology ,Hematopoietic Stem Cells ,Biochemistry ,Hepatic Leukemia Factor ,Hematopoiesis ,Cell biology ,Blood cell ,Transplantation ,Haematopoiesis ,Basic-Leucine Zipper Transcription Factors ,medicine.anatomical_structure ,medicine ,Humans ,Bone marrow ,Single-Cell Analysis ,Stem cell ,Transcriptome ,education - Abstract
Hematopoietic stem cells (HSCs) sustain blood cell homeostasis throughout life and can regenerate all blood lineages after transplantation. Despite this clear functional definition, highly enriched isolation of human HSCs can currently only be achieved through combinatorial assessment of multiple surface antigens. Although several transgenic HSC reporter mouse strains have been described, no analogous approach to prospectively isolate human HSCs has been reported. To identify genes with the most selective expression in human HSCs, we profiled population and single-cell transcriptomes of unexpanded and ex vivo cultured cord blood–derived hematopoietic stem and progenitor cells as well as peripheral blood, adult bone marrow, and fetal liver. On the basis of these analyses, we propose the master transcription factor HLF (hepatic leukemia factor) as one of the most specific HSC marker genes. To directly track its expression in human hematopoietic cells, we developed a genomic HLF reporter strategy, capable of selectively labeling the most immature blood cells on the basis of a single engineered parameter. Most importantly, HLF-expressing cells comprise all stem cell activity in culture and in vivo during serial transplantation. Taken together, these results experimentally establish HLF as a defining gene of the human HSC state and outline a new approach to continuously mark these cells with high fidelity.
- Published
- 2021
- Full Text
- View/download PDF
16. Murine bone marrow mesenchymal stromal cells have reduced hematopoietic maintenance ability in sickle cell disease
- Author
-
Mahmudur Rahman, Yunfeng Liu, Alice Tang, Avital Mendelson, David Shi, Helen Zhang, Patricia A. Shi, Karina Yazdanbakhsh, Weili Bao, Xiuli An, Ana Nicolle Strat, and Deepa Manwani
- Subjects
Male ,Genetic enhancement ,Immunology ,Mice, Transgenic ,Inflammation ,Anemia, Sickle Cell ,medicine.disease_cause ,Hemolysis ,Biochemistry ,Mice ,hemic and lymphatic diseases ,medicine ,Animals ,Blood Transfusion ,business.industry ,Mesenchymal stem cell ,Hematopoietic Stem Cell Transplantation ,Hematopoietic stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Oxidative Stress ,Haematopoiesis ,medicine.anatomical_structure ,Cancer research ,Female ,Bone marrow ,Stem cell ,medicine.symptom ,Transcriptome ,business ,Oxidative stress - Abstract
Sickle cell disease (SCD) is characterized by hemolytic anemia, which can trigger oxidative stress, inflammation, and tissue injury that contribute to disease complications. Bone marrow mesenchymal stromal cells (MSCs) tightly regulate hematopoietic stem cell (HSC) homeostasis in health and disease, but their functionality in SCD remains unclear. We identified for the first time that murine SCD MSCs have altered gene signatures, reduced stem cell properties, and increased oxidative stress, due in part to hemolysis. Murine SCD MSCs had lower HSC maintenance ability in vitro and in vivo, as manifested by increased HSC mobilization and decreased HSC engraftment after transplant. Activation of Toll-like receptor-4 through p65 in MSCs further contributed to MSC dysfunction. Transfusions led to an improved MSC and HSC oxidative state in SCD mice. Improving the regulation between MSCs and HSCs has vital implications for enhancing clinical HSC transplantation and gene therapy outcomes and for identification of new molecular targets for alleviating SCD complications.
- Published
- 2021
- Full Text
- View/download PDF
17. Nonclassical manifestations of acute GVHD
- Author
-
Robert Zeiser and Takanori Teshima
- Subjects
Central Nervous System ,Male ,Immunology ,Graft vs Host Disease ,Context (language use) ,Disease ,Kidney ,Biochemistry ,Bone Marrow ,immune system diseases ,Testis ,medicine ,Animals ,Humans ,Transplantation, Homologous ,Lung ,business.industry ,Ovary ,Hematopoietic Stem Cell Transplantation ,Disease Management ,Cell Biology ,Hematology ,Rash ,Transplantation ,Diarrhea ,surgical procedures, operative ,medicine.anatomical_structure ,Acute Disease ,Female ,Bone marrow ,medicine.symptom ,Complication ,business - Abstract
Acute graft-versus-host disease (GVHD) is a major life-threatening complication after allogeneic hematopoietic cell transplantation (allo-HCT). The classical target organs of acute GVHD include the intestines, liver, and skin. The damage of these organs is relatively easy to detect for the clinician as diarrhea, increased bilirubin, and rash. However, there is increasing evidence that other organs, where the acute damage is less apparent or more difficult to distinguish from drug toxicity, such as the central nervous system, lungs, ovaries and testis, thymus, bone marrow, and kidney, can be target organs of acute GVHD. Here, we review current evidence for nonclassical manifestations of acute GVHD in rodent models and in patients and discuss them in the context of novel emerging therapies for GVHD. A better understanding of the involvement of nonclassical GVHD target organs may help to improve patient outcomes after allo-HCT.
- Published
- 2021
- Full Text
- View/download PDF
18. Megakaryocytes in the lung: guests or ghosts?
- Author
-
Malara A and Balduini A
- Subjects
- Thrombopoiesis, Erythrocyte Membrane, Lung, Bone Marrow, Megakaryocytes
- Published
- 2024
- Full Text
- View/download PDF
19. The bone marrow is the primary site of thrombopoiesis.
- Author
-
Asquith NL, Carminita E, Camacho V, Rodriguez-Romera A, Stegner D, Freire D, Becker IC, Machlus KR, Khan AO, and Italiano JE
- Subjects
- Mice, Humans, Animals, Blood Platelets, Megakaryocytes, Spleen, Bone Marrow, Thrombopoiesis genetics
- Abstract
Abstract: Megakaryocytes (MKs) generate thousands of platelets over their lifespan. The roles of platelets in infection and inflammation has guided an interest to the study of extramedullary thrombopoiesis and therefore MKs have been increasingly reported within the spleen and lung. However, the relative abundance of MKs in these organs compared to the bone marrow and the scale of their contribution to the platelet pool in a steady state remain controversial. We investigated the relative abundance of MKs in the adult murine bone marrow, spleen, and lung using whole-mount light-sheet and quantitative histological imaging, flow cytometry, intravital imaging, and an assessment of single-cell RNA sequencing (scRNA-seq) repositories. Flow cytometry revealed significantly higher numbers of hematopoietic stem and progenitor cells and MKs in the murine bone marrow than in spleens or perfused lungs. Two-photon intravital and light-sheet microscopy, as well as quantitative histological imaging, confirmed these findings. Moreover, ex vivo cultured MKs from the bone marrow subjected to static or microfluidic platelet production assays had a higher capacity for proplatelet formation than MKs from other organs. Analysis of previously published murine and human scRNA-seq data sets revealed that only a marginal fraction of MK-like cells can be found within the lung and most likely only marginally contribute to platelet production in the steady state., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
20. Germline loss-of-function SAMD9 and SAMD9L alterations in adult myelodysplastic syndromes.
- Author
-
Yasunobu Nagata, Satoshi Narumi, Yihong Guan, Przychodzen, Bartlomiej P., Hirsch, Cassandra M., Hideki Makishima, Hirohito Shima, Mai Aly, Pastor, Victor, Kuzmanovic, Teodora, Radivoyevitch, Tomas, Adema, Vera, Awada, Hassan, Kenichi Yoshida, Li, Samuel, Sole, Francesc, Hanna, Rabi, Jha, Babal K., LaFramboise, Thomas, and Seishi Ogawa
- Subjects
- *
INFANTS , *THROMBOCYTOPENIA , *MEGAKARYOCYTES , *MYELODYSPLASTIC syndromes , *BONE marrow - Abstract
The article presents a case study of a 9 month-old infant with familial thrombocytopenia with a SAMD9L variant, marrow normocellularity, and the absence of megakaryocytes. Topics discussed include to assess the effects of rare variants identified in adults with myelodysplastic syndromes (MDSs) and bone marrow failure (BMFs); tested the growth of HEK293 cells with inducible expression of SAMD9 or SAMD9L proteins; and mentions analyzation of copy number alterations (CNAs).
- Published
- 2018
- Full Text
- View/download PDF
21. Measurable residual disease monitoring by NGS before allogeneic hematopoietic cell transplantation in AML.
- Author
-
Thol, Felicitas, Gabdoulline, Razif, Liebich, Alessandro, Klement, Piroska, Schiller, Johannes, Kandziora, Christian, Hambach, Lothar, Stadler, Michael, Koenecke, Christian, Flintrop, Madita, Pankratz, Mira, Wichmann, Martin, Neziri, Blerina, Büttner, Konstantin, Heida, Bennet, Klesse, Sabrina, Chaturvedi, Anuhar, Kloos, Arnold, Göhring, Gudrun, and Schlegelberger, Brigitte
- Subjects
- *
ACUTE myeloid leukemia , *POLYMERASE chain reaction , *GENETIC mutation , *HEMATOPOIETIC stem cell transplantation , *BONE marrow - Abstract
Molecular measurable residual disease (MRD) assessment is not established in approximately 60% of acute myeloid leukemia (AML) patients because of the lack of suitable markers for quantitative real-time polymerase chain reaction. To overcome this limitation, we established an error-corrected next-generation sequencing (NGS) MRD approach that can be applied to any somatic gene mutation. The clinical significance of this approach was evaluated in 116 AML patients undergoing allogeneic hematopoietic cell transplantation (alloHCT) in complete morphologic remission (CR). Targeted resequencing at the time of diagnosis identified a suitable mutation in 93% of the patients, covering 24 different genes. MRD was measured in CR samples from peripheral blood or bone marrow before alloHCT and identified 12 patients with persistence of an ancestral clone (variant allele frequency [VAF] >5%). The remaining 96 patients formed the final cohort of which 45% were MRD+ (median VAF, 0.33%; range, 0.016%-4.91%). In competing risk analysis, cumulative incidence of relapse (CIR) was higher in MRD+ than in MRD- patients (hazard ratio [HR], 5.58; P < .001; 5-year CIR, 66% vs 17%), whereas nonrelapse mortality was not significantly different (HR, 0.60; P = .47). In multivariate analysis, MRD positivity was an independent negative predictor of CIR (HR, 5.68; P < .001), in addition to FLT3-ITD and NPM1 mutation status at the time of diagnosis, and of overall survival (HR, 3.0; P = .004), in addition to conditioning regimen and TP53 and KRAS mutation status. In conclusion, NGS-based MRD is widely applicable to AML patients, is highly predictive of relapse and survival, and may help refine transplantation and posttransplantation management in AML patients. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
22. Extravascular coagulation in hematopoietic stem and progenitor cell regulation.
- Author
-
Nguyen, T. Son, Lapidot, Tsvee, and Ruf, Wolfram
- Subjects
- *
HEMATOPOIETIC stem cells , *BLOOD coagulation factors , *CELLULAR control mechanisms , *BONE marrow , *ENDOTHELIAL cells - Abstract
The hemostatic system plays pivotal roles in injury repair, innate immunity, and adaptation to inflammatory challenges. We review the evidence that these vascular-protective mechanisms have nontraditional roles in hematopoietic stem cell (HSC) maintenance in their physiological bone marrow (BM) niches at steady-state and under stress. Expression of coagulation factors and the extrinsic coagulation initiator tissue factor by osteoblasts, tissue-resident macrophages, and megakaryocytes suggests that endosteal and vascular HSC niches are functionally regulated by extravascular coagulation. The anticoagulant endothelial protein C receptor (EPCR; Procr) is highly expressed by primitive BM HSCs and endothelial cells. EPCR is associated with its major ligand, activated protein C (aPC), in proximity to thrombomodulin-positive blood vessels, enforcing HSC integrin a4 adhesion and chemotherapy resistance in the context of CXCL12-CXCR4 niche retention signals. Protease-activated receptor 1--biased signaling by EPCR-aPC also maintains HSC retention, whereas thrombin signaling activates HSC motility and BM egress. Furthermore, HSC mobilization under stress is enhanced by the fibrinolytic and complement cascades that target HSCs and their BM niches. In addition, coagulation, fibrinolysis, and HSC-derived progeny, including megakaryocytes, synergize to reestablish functional perivascular HSC niches during BM stress. Therapeutic restoration of the anticoagulant pathway has preclinical efficacy in reversing BM failure following radiation injury, but questions remain about how antithrombotic therapy influences extravascular coagulation in HSC maintenance and hematopoiesis. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
23. Reactivation of γ-globin in adult β-YAC mice after ex vivo and in vivo hematopoietic stem cell genome editing.
- Author
-
Chang Li, Psatha, Nikoletta, Sova, Pavel, Sucheol Gil, Hongjie Wang, Jiho Kim, Kulkarni, Chandana, Valensisi, Cristina, Hawkins, R. David, Stamatoyannopoulos, George, and Lieber, André
- Subjects
- *
HEMATOPOIETIC stem cells , *BONE marrow , *STEM cell research , *GENETIC engineering , *HEMATOLOGY - Abstract
Disorders involving β-globin gene mutations, primarily β-thalassemia and sickle cell disease, represent a major target for hematopoietic stem/progenitor cell (HSPC) gene therapy. This includes CRISPR/Cas9-mediated genome editing approaches in adult CD341 cells aimed toward the reactivation of fetal γ-globin expression in red blood cells. Because models involving erythroid differentiation of CD341 cells have limitations in assessing γ-globin reactivation, we focused on human b-globin locus-transgenic (β-YAC) mice. We used a helper-dependent human CD46-targeting adenovirus vector expressing CRISPR/Cas9 (HDAd-HBG-CRISPR) to disrupt a repressor binding region within the γ-globin promoter. We transduced HSPCs from β-YAC/human CD46-transgenic mice ex vivo and subsequently transplanted them into irradiated recipients. Furthermore, we used an in vivo HSPC transduction approach that involves HSPC mobilization and the intravenous injection of HDAd-HBG-CRISPR into β-YAC/CD46-transgenicmice. In both models, we demonstrated efficient target site disruption, resulting in a pronounced switch from human β- to γ-globin expression in red blood cells of adult mice that was maintained after secondary transplantation of HSPCs. In long-term follow-up studies, we did not detect hematological abnormalities, indicating that HBG promoter editing does not negatively affect hematopoiesis. This is the first study that shows successful in vivo HSPC genome editing by CRISPR/Cas9. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
24. Plerixafor effectively mobilizes CD56bright NK cells in blood, providing an allograft predicted to protect against GVHD.
- Author
-
Wong, Peggy P. C., Kariminia, Amina, Jones, David, Eaves, Connie J., Foley, Ronan, Ivison, Sabine, Couban, Stephen, and Schultz, Kirk R.
- Subjects
- *
GRAFT versus host disease , *BONE marrow - Published
- 2018
- Full Text
- View/download PDF
25. A single-cell hematopoietic landscape resolves 8 lineage trajectories and defects in Kit mutant mice.
- Author
-
Dahlin, Joakim S., Hamey, Fiona K., Pijuan-Sala, Blanca, Shepherd, Mairi, Lau, Winnie W. Y., Nestorowa, Sonia, Weinreb, Caleb, Wolock, Samuel, Hannah, Rebecca, Diamanti, Evangelia, Kent, David G., Göttgens, Berthold, and Wilson, Nicola K.
- Subjects
- *
HEMATOPOIETIC stem cells , *CELL proliferation , *GENE expression , *BONE marrow , *EOSINOPHILIA - Abstract
Hematopoietic stem and progenitor cells (HSPCs) maintain the adult blood system, and their dysregulation causes a multitude of diseases. However, the differentiation journeys toward specific hematopoietic lineages remain ill defined, and system-wide disease interpretation remains challenging. Here, we have profiled 44 802 mouse bone marrow HSPCs using single-cell RNA sequencing to provide a comprehensive transcriptional landscape with entry points to 8 different blood lineages (lymphoid, megakaryocyte, erythroid, neutrophil, monocyte, eosinophil, mast cell, and basophil progenitors). We identified a common basophil/mast cell bone marrow progenitor and characterized its molecular profile at the single-cell level. Transcriptional profiling of 13 815 HSPCs from the c-Kit mutant (W41/W41) mouse model revealed the absence of a distinct mast cell lineage entry point, together with global shifts in cell type abundance. Proliferative defects were accompanied by reduced Myc expression. Potential compensatory processes included upregulation of the integrated stress response pathway and downregulation of proapoptotic gene expression in erythroid progenitors, thus providing a template of how large-scale single-cell transcriptomic studies can bridge between molecular phenotypes and quantitative population changes. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
26. Donor telomere length and causes of death after unrelated hematopoietic cell transplantation in patients with marrow failure.
- Author
-
Gadalla, Shahinaz M., Aubert, Geraldine, Tao Wang, Haagenson, Michael, Spellman, Stephen R., Lingxiao Wang, Katki, Hormuzd A., Savage, Sharon A., and Lee, Stephanie J.
- Subjects
- *
TELOMERES , *HEMATOPOIETIC stem cell transplantation , *T cells , *MYELODYSPLASTIC syndromes , *BONE marrow - Abstract
Previous studies have suggested that longer donor leukocyte telomere length (TL) is associated with improved survival after hematopoietic cell transplantation (HCT) in severe aplastic anemia (SAA). This study aimed to determine whether cell-specific lymphocyte TL is associated with certain post-HCT causes of death. We used flow cytometry and fluorescence in situ hybridization to measure TL in donor total lymphocytes and subsets: naïve enriched T cells (CD45RA+CD20-), memory enriched T cells (CD45RA-CD20-), natural killer (NK) fully differentiated T cells (CD45RA+CD57+), and B cells (CD45RA+CD20+). Competing risk survival regression was used for cause-specific death analyses. Clinical data and biospecimens were available from the Center for International Blood and Marrow Transplant Research database and biorepository. The study included 197 patients who underwent unrelated-donor HCT for SAA between 1988 and 2004. The median age at HCT was 15 years (range, 0.5-40 years), and the median follow-up was 5 years (range, <1 month to 20.7 years). Longer donor TL in all cell subsets was associated with lower risk of all-cause mortality (P < .01). In cause-specific mortality analyses, longer TL in B cells (hazard ratio [HR], 0.63; 95% confidence interval [CI], 0.46-0.87; P=.006) and possibly NK fully differentiated T cells (HR, 0.7; 95% CI, 0.51 to 0.97; P = .03) was associated with lower risk of infection-related death. Donor TL in other tested lymphocyte subsets was not statistically significantly associated with death resulting from graft-versus-host disease or graft failure (P > .05). However, a trend toward excess risk of graft-versus-host mortality was noted (HR for total lymphocyte TL, 1.26; P = .15). In conclusion, longer donor TL was associated with reduced rate of infection-related deaths after HCT for SAA. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
27. The identification of fibrosis-driving myofibroblast precursors reveals new therapeutic avenues in myelofibrosis.
- Author
-
Kramann, Rafael and Schneider, Rebekka K.
- Subjects
- *
FIBROSIS , *MYOFIBROBLASTS , *MYELOFIBROSIS , *STROMAL cells , *BONE marrow , *THERAPEUTICS - Abstract
Myofibroblasts are fibrosis-driving cells and are well characterized in solid organ fibrosis, but their role and cellular origin in bone marrow fibrosis remains obscure. Recent work has demonstrated that Gli1+ and LepR+ mesenchymal stromal cells (MSCs) are progenitors of fibrosis-causing myofibroblasts in the bone marrow. Genetic ablation of Gli1+ MSCs or pharmacologic targeting of hedgehog (Hh)-Gli signaling ameliorated fibrosis in mouse models of myelofibrosis (MF). Moreover, pharmacologic or genetic intervention in platelet-derived growth factor receptor a (Pdgfra) signaling in Lepr+ stromal cells suppressed their expansion and ameliorated MF. Improved understanding of cellular and molecular mechanisms in the hematopoietic stem cell niche that govern the transition of MSCs to myofibroblasts and myofibroblast expansion in MF has led to new paradigms in the pathogenesis and treatment of MF. Here, we highlight the central role of malignant hematopoietic clone-derived megakaryocytes in reprogramming the hematopoietic stem cell niche in MF with potential detrimental consequences for hematopoietic reconstitution after allogenic stem cell transplantation, so far the only therapeutic approach in MF considered to be curative. We and others have reported that targeting Hh-Gli signaling is a therapeutic strategy in solid organ fibrosis. Data indicate that targeting Gli proteins directly inhibits Gli1+ cell proliferation and myofibroblast differentiation, which results in reduced fibrosis severity and improved organ function. Although canonical Hh inhibition (eg, smoothened [Smo] inhibition) failed to improve pulmonary fibrosis, kidney fibrosis, or MF, the direct inhibition of Gli proteins ameliorated fibrosis. Therefore, targeting Gli proteins directly might be an interesting and novel therapeutic approach in MF. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
28. Stable lines and clones of long-term proliferating normal, genetically unmodified murine common lymphoid progenitors.
- Author
-
Kawano, Yohei, Petkau, Georg, Stehle, Christina, Durek, Pawel, Heinz, Gitta Anne, Tanimoto, Kousuke, Karasuyama, Hajime, Mashreghi, Mir-Farzin, Romagnani, Chiara, and Melchers, Fritz
- Subjects
- *
LYMPHOCYTIC leukemia , *MOUSE leukemia , *PROGENITOR cells , *CELL lines , *BONE marrow , *CELLULAR therapy - Abstract
Common lymphoid progenitors (CLPs) differentiate to T and B lymphocytes, dendritic cells, natural killer cells, and innate lymphoid cells. Here, we describe culture conditions that, for the first time, allow the establishment of lymphoid-restricted, but uncommitted, long-term proliferating CLP cell lines and clones from a small pool of these cells from normal mouse bone marrow, without any genetic manipulation. Cells from more than half of the cultured CLP clones could be induced to differentiate to T, B, natural killer, dendritic, and myeloid cells in vitro. Cultured, transplanted CLPs transiently populate the host and differentiate to all lymphoid subsets, and to myeloid cells in vivo. This simple method to obtain robust numbers of cultured noncommitted CLPs will allow studies of cell-intrinsic and environmentally controlled lymphoid differentiation programs. If this method can be applied to human CLPs, it will provide new opportunities for cell therapy of patients in need of myeloid-lymphoid reconstitution. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
29. The interplay of leukemia cells and the bone marrow microenvironment.
- Author
-
Duarte, Delfim, Hawkins, Edwin D., and Lo Celso, Cristina
- Subjects
- *
CANCER cells , *STROMAL cells , *DISEASE progression , *BONE marrow , *MYELOID leukemia - Abstract
The interplay of cancer cells and surrounding stroma is critical in disease progression. This is particularly evident in hematological malignancies that infiltrate the bone marrow and peripheral lymphoid organs. Despite clear evidence for the existence of these interactions, the precise repercussions on the growth of leukemic cells are poorly understood. Recent development of novel imaging technology and preclinical disease models has advanced our comprehension of leukemia-microenvironment crosstalk and has potential implications for development of novel treatment options. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
30. mTOR masters monocyte development in bone marrow by decreasing the inhibition of STAT5 on IRF8.
- Author
-
Yang Zhao, Xiaofei Shen, Ning Na, Zhulang Chu, Huiting Su, Shanshan Chao, Lu Shi, Yanan Xu, Lianfeng Zhang, Bingyi Shi, and Yong Zhao
- Subjects
- *
MONOCYTES , *MACROPHAGES , *BONE marrow , *WOUND healing , *PROGENITOR cells - Abstract
Monocytes and macrophages play a key role in defending pathogens, removing the dead cells or cell debris, and wound healing. The mammalian target of rapamycin (mTOR) inhibitor rapamycin (RPM) is widely used in clinics to treat patients with organ transplantation or tumors. The role of mTOR in monocyte/macrophage development remains to be clarified. Here we found that mTOR intrinsically controls monocyte/macrophage development, as evidenced by the decreased percentages and cell numbers of CD11b+F4/80+ cells resulting from mTOR inhibition in SCID mice, mTOR-deficient mice, and mixed chimera mice, and the in vitro colony formation and monocyte/macrophage induction assays. However, Lyzs-mTOR knockout mice displayed normal levels of monocytes/macrophages, indicating that mTOR is not essential for the survival and maturation of monocytes/macrophages. Further studies showed that mTOR deficiency significantly reduced macrophage colony-stimulating factor receptor CD115 expression at the transcriptional and translational levels. The molecular mechanism studies indicate that the impaired monocyte/macrophage development caused by mTOR deficiency is mainly a result of the overactivated STAT5 and subsequent downregulation of IRF8, but not the altered cell metabolism and autophagy. Therefore, our work identifies that mTOR is an intrinsic master for monocyte/macrophage development at the early stages through regulating STAT5-IRF8-dependent CD115-expressing pathway. Long-term usage of RPM may cause a defect of myeloid progenitors in bone marrow. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
31. Measurable residual disease detection by high-throughput sequencing improves risk stratification for pediatric B-ALL.
- Author
-
Wood, Brent, Wu, David, Crossley, Beryl, Yunfeng Dai, Williamson, David, Gawad, Charles, Borowitz, Michael J., Devidas, Meenakshi, Maloney, Kelly W., Larsen, Eric, Winick, Naomi, Raetz, Elizabeth, Carroll, William L., Hunger, Stephen P., Loh, Mignon L., Robins, Harlan, and Kirsch, Ilan
- Subjects
- *
LYMPHOBLASTIC leukemia in children , *CANCER chemotherapy , *BONE marrow , *FLOW cytometry , *T-cell receptor genes , *LEUKEMIA treatment - Abstract
Early response to induction chemotherapy is an important prognostic factor in B-lymphoblastic leukemia (B-ALL). Here, we compare high-throughput sequencing (HTS) of IGH and TRG genes vs flow cytometry (FC) for measurable residual disease (MRD) detection at the end of induction chemotherapy in pediatric patients with newly diagnosed B-ALL. Six hundred nineteen paired pretreatment and end-of-induction bone marrow samples from Children's Oncology Group studies AALL0331 (clinicaltrials.gov #NCT00103285) (standard risk [SR]; with MRD by FC at any level) and AALL0232 (clinicaltrials.gov #NCT00075725) (high risk; with day 29 MRD <0.1% by FC) were evaluated by HTS and FC for event-free (EFS) and overall survival (OS). HTS and FC showed similar 5-year EFS and OS for MRDpositive and -negative patients using an MRD threshold of 0.01%. However, there was a high discordant rate with HTS identifying 55 (38.7%) more patients MRD positive at this threshold. These discrepant patients have worse outcomes than FC MRD-negative patients. In addition, the increased analytic sensitivity of HTS permitted identification of 19.9% of SR patients without MRD at any detectable level who had excellent 5-year EFS (98.1%) and OS (100%). The higher analytic sensitivity and lower false-negative rate of HTS improves upon FC for MRD detection in pediatric B-ALL by identifying a novel subset of patients at end of induction who are essentially cured using current chemotherapy and identifying MRD at 0.01% in up to one-third of patients who are missed at the same threshold by FC. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
32. Atorvastatin enhances bone marrow endothelial cell function in corticosteroid-resistant immune thrombocytopenia patients.
- Author
-
Yuan Kong, Xie-Na Cao, Xiao-Hui Zhang, Min-Min Shi, Yue-Yun Lai, Yu Wang, Lan-Ping Xu, Ying-Jun Chang, and Xiao-Jun Huang
- Subjects
- *
BONE marrow , *ATORVASTATIN , *STATINS (Cardiovascular agents) , *ANTILIPEMIC agents , *HEMATOPOIESIS , *IDIOPATHIC thrombocytopenic purpura , *PROGENITOR cells - Abstract
The pathogenesis of corticosteroid-resistant immune thrombocytopenia (ITP), a clinically challenging condition in which patients exhibit either no response to corticosteroids or are corticosteroid-dependent, remains poorly understood. Murine studies suggest that bone marrow (BM) endothelial progenitor cells (EPCs) play a crucial role in regulating megakaryocytopoiesis. However, little is known regarding the number and function of BM EPCs or how to improve impaired BM EPCs in corticosteroid-resistant ITP patients. In the current case-control study, we evaluated whether the BM EPCs in corticosteroid-resistant ITP differed from those in corticosteroid-sensitive ITP. Moreover, whether atorvastatin could enhance the number and function of BM EPCs derived from corticosteroid-resistant ITP patients was investigated in vitro and in vivo. Reduced and dysfunctional BM EPCs, characterized by decreased capacities of migration and angiogenesis as well as higher levels of reactive oxygen species and apoptosis, were observed in corticosteroid-resistant ITP patients. In vitro treatment with atorvastatin quantitatively and functionally improved BM EPCs derived from corticosteroid-resistant ITP patients by downregulating the p38 MAPK pathway and upregulating the Akt pathway, and rescued the impaired BM EPCs to support megakaryocytopoiesis. Subsequently, a pilot cohort study showed that atorvastatin was safe and effective in corticosteroid-resistant ITP patients. Taken together, these results indicate that reduced and dysfunctional BM EPCs play a role in the pathogenesis of corticosteroid-resistant ITP, and the impaired BM EPCs could be improved by atorvastatin both in vitro and in vivo. Although requiring further validation, our data indicate that atorvastatin represents a promising therapeutic approach for repairing impaired BM EPCs in corticosteroid-resistant ITP patients. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
33. The impact of aging on primate hematopoiesis as interrogated by clonal tracking.
- Author
-
Kyung-Rok Yu, Espinoza, Diego A., Chuanfeng Wu, Truitt, Lauren, Tae-Hoon Shin, Chen, Shirley, Xing Fan, Yabe, Idalia M., Panch, Sandhya, Hong, So Gun, Koelle, Samson, Rong Lu, Bonifacino, Aylin, Krouse, Allen, Metzger, Mark, Donahue, Robert E., and Dunbar, Cynthia E.
- Subjects
- *
HEMATOPOIETIC stem cells , *HEMATOPOIESIS , *STEM cell culture , *BONE marrow , *PRIMATES - Abstract
Age-associated changes in hematopoietic stem and progenitor cells (HSPCs) have been carefully documented in mouse models but poorly characterized in primates and humans. To investigate clinically relevant aspects of hematopoietic aging, we compared the clonal output of thousands of genetically barcoded HSPCs in aged vs young macaques after autologous transplantation. Aged macaques showed delayed emergence of output from multipotent (MP) clones, with persistence of lineage-biased clones for many months after engraftment. In contrast to murine aging models reporting persistence of myeloid-biased HSPCs, aged macaques demonstrated persistent output from both B-cell and myeloid-biased clones. Clonal expansions of MP, myeloid-biased, and B-biased clones occurred in aged macaques, providing a potential model for human clonal hematopoiesis of indeterminate prognosis. These results suggest that long-term MP HSPC output is impaired in aged macaques, resulting in differences in the kinetics and lineage reconstitution patterns between young and aged primates in an autologous transplantation setting. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
34. Syndecan-1 promotes Wnt/β-catenin signaling in multiple myeloma by presenting Wnts and R-spondins.
- Author
-
Ren, Zemin, van Andel, Harmen, de Lau, Wim, Hartholt, Robin B., Maurice, Madelon M., Clevers, Hans, Kersten, Marie José, Spaargaren, Marcel, and Pals, Steven T.
- Subjects
- *
MULTIPLE myeloma , *PLASMA cells , *BONE marrow , *CELL proliferation , *GENETIC mutation - Abstract
Multiple myeloma (MM) is characterized by the expansion of malignant plasma cells in the bone marrow (BM). Most MMs display aberrant Wnt/β-catenin signaling, which drives proliferation; however, they lack oncogenic Wnt pathway mutations, suggesting activation by autocrine Wnt ligands and/or paracrine Wnts from the BM microenvironment. Expression of the heparan sulfate (HS) proteoglycan syndecan-1 is a hallmark of MM. Syndecan-1 is a critical player in the complex reciprocal interaction between MM cells and their BM niche, mediating growth factor/cytokine binding and signaling by its HS chains. Here, by means of CRISPR/Cas9-mediated knockout and doxycycline-inducible short hairpin RNA-mediated knockdown of EXT1, a critical enzyme for HS polymerization, we demonstrate that the HS chains decorating syndecan-1 mediate aberrant Wnt pathway activation in MM. HS-deficient MM cells exhibited strongly decreased autocrine Wnt/β-catenin pathway activity and reduced Wnt pathway-dependent proliferation. In addition, we demonstrate that Wnts bind to the HS side chains of syndecan-1 and that this binding contributes to paracrine Wnt pathway activation through the Wnt receptor Frizzled (Fzd). Furthermore, in an HS-dependent fashion, syndecan-1 also binds osteoblast-produced R-spondin, which represses Fzd degradation by activation of LGR4, an R-spondin receptor aberrantly expressed on MM cells. Costimulation with R-spondin and its binding to HS chains decorating syndecan-1 are indispensable for optimal stimulation of Wnt signaling in MM. Taken together, our results identify syndecan-1 as a crucial component of the Wnt signalosome in MM cells, binding Wnts and R-spondins to promote aberrant Wnt/β-catenin signaling and cell growth, and suggest HS and its biosynthetic enzymes as potential targets in the treatment of MM. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
35. Specific targeting of the KRAS mutational landscape in myeloma as a tool to unveil the elicited antitumor activity
- Author
-
Rossella Ribolla, Joana Hauser, Marco Presta, Chiara Cattaneo, Roberto Ronca, Cosetta Ravelli, Arianna Giacomini, Federica Maccarinelli, Aldo M. Roccaro, Anna Staniszewska, Stefania Mitola, Alexey S. Revenko, Michele Moschetta, Philip Martin, Antonio Sacco, Niccolo Bolli, Bachisio Ziccheddu, Angelo Belotti, Giada Bianchi, Alessandra Tucci, Cinzia Federico, Helen Ambrose, Hongbo Cai, Robert A. Macleod, Lyndsey Hanson, Valentina Palermo, Giuseppe Rossi, Brandon Willis, Sarah Ross, Claire Rooney, Katia Todoerti, Vanessa Favasuli, Antonino Neri, and Sophie E. Willis
- Subjects
endocrine system diseases ,Somatic cell ,Immunology ,Antineoplastic Agents ,Context (language use) ,Mice, SCID ,Biology ,medicine.disease_cause ,Biochemistry ,Proto-Oncogene Proteins p21(ras) ,Small Molecule Libraries ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,Molecular Targeted Therapy ,neoplasms ,Gene ,Multiple myeloma ,Mutation ,Lymphoid Neoplasia ,Cell Biology ,Hematology ,Oligonucleotides, Antisense ,medicine.disease ,digestive system diseases ,respiratory tract diseases ,medicine.anatomical_structure ,Cancer research ,Bone marrow ,KRAS ,Neoplasm Recurrence, Local ,Multiple Myeloma - Abstract
Alterations in KRAS have been identified as the most recurring somatic variants in the multiple myeloma (MM) mutational landscape. Combining DNA and RNA sequencing, we studied 756 patients and observed KRAS as the most frequently mutated gene in patients at diagnosis; in addition, we demonstrated the persistence or de novo occurrence of the KRAS aberration at disease relapse. Small-molecule inhibitors targeting KRAS have been developed; however, they are selective for tumors carrying the KRASG12C mutation. Therefore, there is still a need to develop novel therapeutic approaches to target the KRAS mutational events found in other tumor types, including MM. We used AZD4785, a potent and selective antisense oligonucleotide that selectively targets and downregulates all KRAS isoforms, as a tool to dissect the functional sequelae secondary to KRAS silencing in MM within the context of the bone marrow niche and demonstrated its ability to significantly silence KRAS, leading to inhibition of MM tumor growth, both in vitro and in vivo, and confirming KRAS as a driver and therapeutic target in MM.
- Published
- 2021
- Full Text
- View/download PDF
36. Diversity, localization, and (patho)physiology of mature lymphocyte populations in the bone marrow
- Author
-
Christian M. Schürch, Martijn A. Nolte, and Chiara Caraccio
- Subjects
Immunology ,Graft vs Host Disease ,Bone Marrow Cells ,Biology ,Biochemistry ,Mice ,Immune system ,Cell Movement ,medicine ,Animals ,Humans ,Lymphocytes ,Aplastic anemia ,Innate lymphoid cell ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Allografts ,Hematopoietic Stem Cells ,medicine.disease ,Natural killer T cell ,Thrombocytopenia ,Immunity, Innate ,Hematopoiesis ,Haematopoiesis ,medicine.anatomical_structure ,Hematologic Neoplasms ,Mature Lymphocyte ,Bone marrow ,Stem cell - Abstract
The bone marrow (BM) is responsible for generating and maintaining lifelong output of blood and immune cells. In addition to its key hematopoietic function, the BM acts as an important lymphoid organ, hosting a large variety of mature lymphocyte populations, including B cells, T cells, natural killer T cells, and innate lymphoid cells. Many of these cell types are thought to visit the BM only transiently, but for others, like plasma cells and memory T cells, the BM provides supportive niches that promote their long-term survival. Interestingly, accumulating evidence points toward an important role for mature lymphocytes in the regulation of hematopoietic stem cells (HSCs) and hematopoiesis in health and disease. In this review, we describe the diversity, migration, localization, and function of mature lymphocyte populations in murine and human BM, focusing on their role in immunity and hematopoiesis. We also address how various BM lymphocyte subsets contribute to the development of aplastic anemia and immune thrombocytopenia, illustrating the complexity of these BM disorders and the underlying similarities and differences in their disease pathophysiology. Finally, we summarize the interactions between mature lymphocytes and BM resident cells in HSC transplantation and graft-versus-host disease. A better understanding of the mechanisms by which mature lymphocyte populations regulate BM function will likely improve future therapies for patients with benign and malignant hematologic disorders.
- Published
- 2021
- Full Text
- View/download PDF
37. CD44 loss of function sensitizes AML cells to the BCL-2 inhibitor venetoclax by decreasing CXCL12-driven survival cues
- Author
-
Tanja Nicole Hartmann, Karolin Streule, Carsten Müller-Tidow, Romina Judith Walter, Richard Greil, Leonel Munoz-Sagredo, Patricia Muschong, Miguel L. Concha, Julia Christine Gutjahr, Elisabeth Bayer, Xiaobing Yu, and Véronique Orian-Rousseau
- Subjects
Male ,Homeobox protein NANOG ,Cell Survival ,Immunology ,Biochemistry ,chemistry.chemical_compound ,SOX2 ,Loss of Function Mutation ,Tumor Cells, Cultured ,Humans ,Medicine ,Sulfonamides ,biology ,Venetoclax ,business.industry ,CD44 ,Myeloid leukemia ,Cell Biology ,Hematology ,Bridged Bicyclo Compounds, Heterocyclic ,Minimal residual disease ,Chemokine CXCL12 ,Leukemia, Myeloid, Acute ,Hyaluronan Receptors ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,chemistry ,embryonic structures ,Cancer research ,biology.protein ,Female ,Bone marrow ,Stem cell ,business - Abstract
Acute myeloid leukemia (AML) has a poor prognosis under the current standard of care. In recent years, venetoclax, a BCL-2 inhibitor, was approved to treat patients who are ineligible for intensive induction chemotherapy. However, complete remission rates with venetoclax-based therapies are hampered by minimal residual disease (MRD) in a proportion of patients, leading to relapse. MRD is a result of leukemic stem cells being retained in bone marrow protective environments; activation of the CXCL12-CXCR4 pathway was shown to be relevant to this process. An important role is also played by cell adhesion molecules such as CD44, which has been shown to be crucial for the development of AML. Here we show that CD44 is involved in CXCL12 promotion of resistance to venetoclax-induced apoptosis in human AML cell lines and AML patient samples, which could be abrogated by CD44 knock down, knockout, or blocking with an anti-CD44 antibody. Split-Venus bimolecular fluorescence complementation showed that CD44 and CXCR4 physically associate at the cell membrane upon CXCL12 induction. In the venetoclax-resistant OCI-AML3 cell line, CXCL12 promoted an increase in the proportion of cells expressing high levels of embryonic stem cell core transcription factors (ESC-TFs: Sox2, Oct4, Nanog) abrogated by CD44 knockdown. This ESC-TF–expressing subpopulation which could be selected by venetoclax treatment, exhibited a basally enhanced resistance to apoptosis and expressed higher levels of CD44. Finally, we developed a novel AML xenograft model in zebrafish, which showed that CD44 knockout sensitizes OCI-AML3 cells to venetoclax treatment in vivo. Our study shows that CD44 is a potential molecular target for sensitizing AML cells to venetoclax-based therapies.
- Published
- 2021
- Full Text
- View/download PDF
38. Hematologic disorder–associated Cxcr4 gain-of-function mutation leads to uncontrolled extrafollicular immune response
- Author
-
Nagham Alouche, Niclas Setterblad, Amélie Bonaud, Karl Balabanian, Etienne Crickx, Valeria Bisio, Mélanie Khamyath, David H. McDermott, Matthieu Mahévas, Nicolas Dulphy, Philip M. Murphy, Marion Espéli, Vincent Rondeau, Rim Hussein-Agha, and Julie Nguyen
- Subjects
Receptors, CXCR4 ,Immunobiology and Immunotherapy ,Plasma Cells ,Immunology ,Mice, Transgenic ,Biology ,Biochemistry ,CXCR4 ,Hypogammaglobulinemia ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Humans ,Mechanistic target of rapamycin ,030304 developmental biology ,Myelokathexis ,0303 health sciences ,TOR Serine-Threonine Kinases ,Cell Biology ,Hematology ,medicine.disease ,Hematologic Diseases ,3. Good health ,medicine.anatomical_structure ,Gain of Function Mutation ,biology.protein ,Bone marrow ,Antibody ,WHIM syndrome ,Signal Transduction ,030215 immunology - Abstract
The extrafollicular immune response is essential to generate a rapid but transient wave of protective antibodies during infection. Despite its importance, the molecular mechanisms controlling this first response are poorly understood. Here, we demonstrate that enhanced Cxcr4 signaling caused by defective receptor desensitization leads to exacerbated extrafollicular B-cell response. Using a mouse model bearing a gain-of-function mutation of Cxcr4 described in 2 human hematologic disorders, warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome and Waldenström macroglobulinemia, we demonstrated that mutant B cells exhibited enhanced mechanistic target of rapamycin signaling, cycled more, and differentiated more potently into plasma cells than wild-type B cells after Toll-like receptor (TLR) stimulation. Moreover, Cxcr4 gain of function promoted enhanced homing and persistence of immature plasma cells in the bone marrow, a phenomenon recapitulated in WHIM syndrome patient samples. This translated in increased and more sustained production of antibodies after T-independent immunization in Cxcr4 mutant mice. Thus, our results establish that fine-tuning of Cxcr4 signaling is essential to limit the strength and length of the extrafollicular immune response.
- Published
- 2021
- Full Text
- View/download PDF
39. Potent preclinical activity of HexaBody-DR5/DR5 in relapsed and/or refractory multiple myeloma
- Author
-
Sonja Zweegman, Anne T. Gelderloos, Esther C.W. Breij, Inger S. Nijhof, Tuna Mutis, Hilma J. van der Horst, Marije B. Overdijk, Martine E D Chamuleau, Hematology, CCA - Cancer biology and immunology, AII - Cancer immunology, and Hematology laboratory
- Subjects
0301 basic medicine ,Antineoplastic Agents ,Immunoglobulin G ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Medicine ,Cytotoxic T cell ,Humans ,Lenalidomide ,Multiple myeloma ,Lymphoid Neoplasia ,biology ,business.industry ,Bortezomib ,Antibody-Dependent Cell Cytotoxicity ,Daratumumab ,Hematology ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Bone marrow ,Antibody ,business ,Multiple Myeloma ,medicine.drug - Abstract
Apoptosis induction by death receptor (DR)-specific agonistic antibodies is a potentially effective antitumor therapy. Nonetheless, to date, all conventional DR-targeting antibodies that induce apoptosis via FcγR-dependent DR clustering failed to show clinical efficacy. HexaBody-DR5/DR5 (GEN1029) has been developed to overcome full FcγR dependence. HexaBody-DR5/DR5 is a mixture of 2 noncompeting DR5-specific immunoglobulin G1 (IgG1) antibodies, each with an E430G mutation in the Fc domain. This mutation enhances Fc-Fc interactions, resulting in antibody hexamerization, followed by FcγR-independent clustering of DR5 molecules. This unique combination of dual epitope targeting and increased IgG hexamerization resulted in potent preclinical antitumor activity in various solid cancers. In this study, we explored the preclinical activity of HexaBody-DR5/DR5 in multiple myeloma (MM), because MM cells are known to express DR5. In bone marrow samples from 48 MM patients, HexaBody-DR5/DR5 induced potent cytotoxicity of primary MM cells. Importantly, HexaBody-DR5/DR5 mediated the highest cytotoxic activity in samples from relapsed/refractory MM patients, including those who are refractory to daratumumab. This improved cytotoxic activity was observed only in patients who received their last anti-MM treatment
- Published
- 2021
- Full Text
- View/download PDF
40. miR-15a/16-1 deletion in activated B cells promotes plasma cell and mature B-cell neoplasms
- Author
-
Irene M. Ghobrial, Vignesh Shanmugam, Juerg R. Straubhaar, Tomasz Sewastianik, Petr Jarolim, Mehmet Kemal Samur, Peter S. Dennis, Nikhil C. Munshi, Ruben D. Carrasco, Vinodh Pillai, Mary L. Bouxsein, David M. Dorfman, Jianjun Zhao, Jianli Wang, Helen Tanton, Jianhong Lin, Geraldine S. Pinkus, Daniel J. Brooks, Omar Nadeem, Ying Huang, Davide F. Robbiani, Bogdan Budnik, Meng Jiang, Keith Adler, and Kenneth C. Anderson
- Subjects
Plasma Cells ,Immunology ,Chromosome Disorders ,Plasma cell ,Biology ,Biochemistry ,medicine ,Animals ,Humans ,Neoplasms, Plasma Cell ,Multiple myeloma ,B-Lymphocytes ,Lymphoid Neoplasia ,Chromosomes, Human, Pair 13 ,Germinal center ,Cancer ,Chromosome ,Cell Biology ,Hematology ,medicine.disease ,Molecular biology ,Lymphoma ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,MicroRNAs ,medicine.anatomical_structure ,Multigene Family ,Lymphoma, Large B-Cell, Diffuse ,Bone marrow ,Hyperdiploidy ,Chromosome Deletion ,Multiple Myeloma ,Gene Deletion ,Plasmacytoma - Abstract
Chromosome 13q deletion [del(13q)], harboring the miR-15a/16-1 cluster, is one of the most common genetic alterations in mature B-cell malignancies, which originate from germinal center (GC) and post-GC B cells. Moreover, miR-15a/16 expression is frequently reduced in lymphoma and multiple myeloma (MM) cells without del(13q), suggesting important tumor-suppressor activity. However, the role of miR-15a/16-1 in B-cell activation and initiation of mature B-cell neoplasms remains to be determined. We show that conditional deletion of the miR-15a/16-1 cluster in murine GC B cells induces moderate but widespread molecular and functional changes including an increased number of GC B cells, percentage of dark zone B cells, and maturation into plasma cells. With time, this leads to development of mature B-cell neoplasms resembling human extramedullary plasmacytoma (EP) as well as follicular and diffuse large B-cell lymphomas. The indolent nature and lack of bone marrow involvement of EP in our murine model resembles human primary EP rather than MM that has progressed to extramedullary disease. We corroborate human primary EP having low levels of miR-15a/16 expression, with del(13q) being the most common genetic loss. Additionally, we show that, although the mutational profile of human EP is similar to MM, there are some exceptions such as the low frequency of hyperdiploidy in EP, which could account for different disease presentation. Taken together, our studies highlight the significant role of the miR-15a/16-1 cluster in the regulation of the GC reaction and its fundamental context-dependent tumor-suppression function in plasma cell and B-cell malignancies.
- Published
- 2021
- Full Text
- View/download PDF
41. Syndecan-1 and stromal heparan sulfate proteoglycans: key moderators of plasma cell biology and myeloma pathogenesis
- Author
-
Marcel Spaargaren, Steven T. Pals, and Zemin Ren
- Subjects
Stromal cell ,Immunology ,Plasma Cells ,Biology ,Plasma cell ,Blood Spotlight ,Biochemistry ,Heparan Sulfate Proteoglycans ,Syndecan 1 ,Pathogenesis ,Bone Marrow ,Malignant myeloma ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Multiple myeloma ,Cell Biology ,Hematology ,medicine.disease ,Cell biology ,carbohydrates (lipids) ,medicine.anatomical_structure ,Proteoglycans ,Bone marrow ,Heparitin Sulfate ,Syndecan-1 ,Multiple Myeloma - Abstract
Plasma cells no longer express a B-cell antigen receptor and are hence deprived of signals crucial for survival throughout B-cell development. Instead, normal plasma cells, as well as their malignant myeloma counterparts, heavily rely on communication with the bone marrow (BM) microenvironment for survival. The plasma cell heparan sulfate proteoglycan (HSPG) syndecan-1 (CD138) and HSPGs in the BM microenvironment act as master regulators of this communication by co-opting specific growth and survival factors from the BM niche. This designates syndecan-1/HSPGs and their synthesis machinery as potential treatment targets in multiple myeloma.
- Published
- 2021
42. The characterization of distinct populations of murine skeletal cells that have different roles in B lymphopoiesis
- Author
-
Lenny Straszkowski, Alistair M. Chalk, Julie M. Quach, Louise E. Purton, Samuel C. Lee, Takaharu Kimura, Joy Y. Wu, Diannita Kwang, Gavin Tjin, Alanna C. Green, and Emma Baker
- Subjects
education.field_of_study ,Leptin receptor ,Cell adhesion molecule ,Immunology ,Cell ,Population ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Growth factor receptor ,medicine ,Lymphopoiesis ,Bone marrow ,education - Abstract
Hematopoiesis is extrinsically controlled by cells of the bone marrow microenvironment, including skeletal lineage cells. The identification and subsequent studies of distinct subpopulations of maturing skeletal cells is currently limited because of a lack of methods to isolate these cells. We found that murine Lin–CD31–Sca-1–CD51+ cells can be divided into 4 subpopulations by using flow cytometry based on their expression of the platelet-derived growth factor receptors ⍺ and β (PDGFR⍺ and PDGFRβ). The use of different skeletal lineage reporters confirmed the skeletal origin of the 4 populations. Multiplex immunohistochemistry studies revealed that all 4 populations were localized near the growth plate and trabecular bone and were rarely found near cortical bone regions or in central bone marrow. Functional studies revealed differences in their abundance, colony-forming unit–fibroblast capacity, and potential to differentiate into mineralized osteoblasts or adipocytes in vitro. Furthermore, the 4 populations had distinct gene expression profiles and differential cell surface expression of leptin receptor (LEPR) and vascular cell adhesion molecule 1 (VCAM-1). Interestingly, we discovered that 1 of these 4 different skeletal populations showed the highest expression of genes involved in the extrinsic regulation of B lymphopoiesis. This cell population varied in abundance between distinct hematopoietically active skeletal sites, and significant differences in the proportions of B-lymphocyte precursors were also observed in these distinct skeletal sites. This cell population also supported pre-B lymphopoiesis in culture. Our method of isolating 4 distinct maturing skeletal populations will help elucidate the roles of distinct skeletal niche cells in regulating hematopoiesis and bone.
- Published
- 2021
- Full Text
- View/download PDF
43. Absence of a common founder mutation in patients with cooccurring myelodysplastic syndrome and plasma cell disorder
- Author
-
Monika Klimkowska, Robert Månsson, Seishi Ogawa, Hareth Nahi, Charlotte Gran, Magnus Tobiasson, Iyadh Douagi, and Yasuhito Nannya
- Subjects
Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Myeloid ,Plasma Cells ,Immunology ,Biochemistry ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,Epidemiology ,medicine ,Humans ,Exome sequencing ,Multiple myeloma ,Aged ,Aged, 80 and over ,business.industry ,Incidence (epidemiology) ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Founder Effect ,030104 developmental biology ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,Mutation ,Cohort ,Female ,Bone marrow ,business ,030215 immunology - Abstract
Epidemiological studies have demonstrated an increased incidence of MDS in patients with plasma cell disorder (PCD) i.e. multiple myeloma (MM) or MGUS and several case reports / series of co-occurring MDS and PCD have been published. The underlying pathogenesis for this condition, and if the two diseases share a common genetic lesion remains unknown. Here, we describe a cohort of 27 consecutive patients with co-occurring MDS and MM (n=6), MGUS (n=20) or plasmocytoma (n=1), diagnosed at the Karolinska University Hospital. In 5 patients, the diagnosis of MGUS preceded the diagnosis of MDS , in one patient the MDS diagnosis preceded PCD, and in 21 patients MDS and PCD were diagnosed at the same time. There was a preponderance for lower-risk MDS subgroups with only 3 patients belonging to the IPSS-R high / very high risk groups. Median overall survival for the whole cohort was 44 months. The most common mutations were TET2, SRSF2 and SF3B1. To identify potential common founder clones, we performed whole exome sequencing on isolated bone marrow myeloid-, plasma- and T-cells from 9 patients. In none of the patients, we could detect a common founder mutation and the two diseases have likely emerged from separate clones.
- Published
- 2021
- Full Text
- View/download PDF
44. Gelatinous transformation of bone marrow and concomitant fungal infection in a patient with HIV
- Author
-
Sri Bharathi Kavuri and Kirill A. Lyapichev
- Subjects
Mycoses ,Bone Marrow ,Immunology ,Gelatin ,Humans ,HIV Infections ,Cell Biology ,Hematology ,Biochemistry ,Bone Marrow Diseases - Published
- 2022
45. A landscape of germ line mutations in a cohort of inherited bone marrow failure patients.
- Author
-
Bluteau, Olivier, Sebert, Marie, Leblanc, Thierry, de Latour, Régis Peffault, Quentin, Samuel, Lainey, Elodie, Hernandez, Lucie, Dalle, Jean-Hugues, de Fontbrune, Flore Sicre, Lengline, Etienne, Itzykson, Raphael, Clappier, Emmanuelle, Boissel, Nicolas, Vasquez, Nadia, Costa, Mélanie Da, Masliah-Planchon, Julien, Cuccuini, Wendy, Raimbault, Anna, De Jaegere, Louis, and Adès, Lionel
- Subjects
- *
BONE marrow , *FANCONI'S anemia , *RIBOSOMES , *PANCYTOPENIA , *APLASTIC anemia , *DNA - Abstract
Bone marrow (BM) failure (BMF) in children and young adults is often suspected to be inherited, but in many cases diagnosis remains uncertain. We studied a cohort of 179 patients (from 173 families) with BMF of suspected inherited origin but unresolved diagnosis after medical evaluation and Fanconi anemia exclusion. All patients had cytopenias, and 12.0% presented ≥5% BM blast cells. Median age at genetic evaluation was 11 years; 20.7% of patients were aged ≤2 years and 36.9% were ≥18 years. We analyzed genomic DNA from skin fibroblasts using whole-exome sequencing, and were able to assign a causal or likely causal germ line mutation in 86 patients (48.0%), involving a total of 28 genes. These included genes in familial hematopoietic disorders (GATA2, RUNX1), telomeropathies (TERC, TERT, RTEL1), ribosome disorders (SBDS, DNAJC21, RPL5), and DNA repair deficiency (L/G4). Many patients had an atypical presentation, and the mutated gene was often not clinically suspected. We also found mutations in genes seldom reported in inherited BMF (IBMF), such as SAMD9 and SAMD9L (N = 16 of the 86 patients, 18.6%), MECOM/EVI1 (N = 6, 7.0%), and ERCC6L2 (N = 7, 8.1%), each of which was associated with a distinct natural history; SAMD9 and SAMD9L patients often experienced transient aplasia and monosomy 7, whereas MECOM patients presented early-onset severe aplastic anemia, and ERCC6L2 patients, mild pancytopenia with myelodysplasia. This study broadens the molecular and clinical portrait of IBMF syndromes and sheds light on newly recognized disease entities. Using a high-throughput sequencing screen to implement precision medicine at diagnosis can improve patient management and family counseling. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
46. Cellular kinetics of CTL019 in relapsed/refractory B-cell acute lymphoblastic leukemia and chronic lymphocytic leukemia.
- Author
-
Thudium Mueller, Karen, Maude, Shannon L., Porter, David L., Frey, Noelle, Wood, Patricia, Han, Xia, Waldron, Edward, Chakraborty, Abhijit, Awasthi, Rakesh, Levine, Bruce L., Melenhorst, J. Joseph, Grupp, Stephan A., June, Carl H., and Lacey, Simon F.
- Subjects
- *
LYMPHOBLASTIC leukemia , *LYMPHOCYTIC leukemia , *IMMUNOTHERAPY , *ANTIGEN receptors , *BONE marrow , *FLOW cytometry - Abstract
Tisagenlecleucel (CTL019) is an investigational immunotherapy that involves reprogramming a patient's own T cells with a transgene encoding a chimeric antigen receptor to identify and eliminate CD19-expressing cells. We previously reported that CTL019 achieved impressive clinical efficacy in patients with relapsed/refractory B-cell acute lymphoblastic leukemia (ALL) and chronic lymphocytic leukemia (CLL), including the expansion and persistence of CTL019 cells, which correlates with response to therapy. Here, we performed formal cellular kinetic analyses of CTL019 in a larger cohort of 103 patients treated with CTL019 in 2 different diseases (ALL and CLL). CTL019 was measured in peripheral blood and bone marrow, using quantitative polymerase chain reaction and flow cytometry. CTL019 levels in peripheral blood typically peaked at 10 to 14 days postinfusion and then declined slowly over time. Patients with complete response (CR)/CR with incomplete count recovery had higher levels of CTL019 in peripheral blood, with greater maximal concentration and area under the curve values compared with nonresponding patients (P < .0001 for each). CTL019 transgene levels were measurable up to 780 days in peripheral blood. CTL019 trafficking and persistence were observed in bone marrow and cerebrospinal fluid. CTL019 expansion correlated with severity of cytokine release syndrome (CRS) and preinfusion tumor burden in pediatric ALL. The results described here are the first detailed formal presentation of cellular kinetics across 2 diseases and highlight the importance of the application of in vivo cellular kinetic analyses to characterize clinical efficacy and CRS severity associated with CTL019 therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
47. TP53 mutations identify younger mantle cell lymphoma patients who do not benefit from intensive chemoimmunotherapy.
- Author
-
Eskelund, Christian W., Dahl, Christina, Hansen, Jakob W., Westman, Maj, Kolstad, Arne, Pedersen, Lone B., Montano-Almendras, Carmen P., Husby, Simon, Freiburghaus, Catja, Ek, Sara, Pedersen, Anja, Niemann, Carsten, Räty, Riikka, Brown, Peter, Geisler, Christian H., Andersen, Mette K., Guldberg, Per, Jerkeman, Mats, and Grønbæk, Kirsten
- Subjects
- *
MANTLE cell lymphoma , *LYMPHOMA treatment , *BONE marrow , *CANCER chemotherapy , *STEM cell transplantation , *MORPHOLOGY - Abstract
Despite recent advances in lymphoma treatment, mantle cell lymphoma (MCL) remains incurable, and we are still unable to identify patients who will not benefit from the current standard of care. Here, we explore the prognostic value of recurrent genetic aberrations in diagnostic bone marrow (BM) specimens from 183 younger patients with MCL from the Nordic MCL2 and MCL3 trials, which represent current standard-of-care regimens. In the univariate model, mutations of TP53 (11%) and NOTCH1 (4%), and deletions of TP53 (16%) andCDKN2A(20%),weresignificantly associatedwithinferioroutcomes(togetherwithMIPI, MIPI-c, blastoidmorphology, and Ki67 > 30%); however, inmultivariate analyses, only TP53 mutations (HR, 6.2; P < .0001) retained prognostic impact for overall survival (OS), whereas TP53 mutations (HR, 6.9; P < .0001) andMIPI-c high-risk (HR, 2.6; P5.003) had independent prognostic impact on time to relapse. TP53-mutated cases had a dismal outcome, with a median OS of 1.8 years, and 50% relapsed at 1.0 years, compared to a median OS of 12.7 years for TP53-unmutated cases (P < .0001). TP53 mutations were significantly associated with Ki67 > 30%, blastoid morphology, MIPI high-risk, and inferior responses to both induction- and high-dose chemotherapy. In conclusion, we show that TP53mutations identify a phenotypically distinct and highly aggressive form of MCL with poor or no response to regimens including cytarabine, rituximab, and autologous stem-cell transplant (ASCT). We suggest patients with MCL should be stratified according to TP53 status, and that patients with TP53 mutations should be considered for experimental frontline trials exploring novel agents. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
48. Tropomodulin 1 controls erythroblast enucleation via regulation of F-actin in the enucleosome.
- Author
-
Nowak, Roberta B., Papoin, Julien, Gokhin, David S., Casu, Carla, Rivella, Stefano, Lipton, Jeffrey M., Blanc, Lionel, and Fowler, Velia M.
- Subjects
- *
TROPOMODULIN , *CELL enucleation , *F-actin , *ERYTHROCYTES , *BONE marrow - Abstract
Biogenesis of mammalian red blood cells requires nuclear expulsion by orthochromatic erythoblasts late in terminal differentiation (enucleation), but the mechanism is largely unexplained. Here, we employed high-resolution confocal microscopy to analyze nuclear morphology and F-actin rearrangements during the initiation, progression, and completion of mouse and human erythroblast enucleation in vivo. Mouse erythroblast nuclei acquire a dumbbell-shaped morphology during enucleation, whereas human bone marrow erythroblast nuclei unexpectedly retain their spherical morphology. These morphological differences are linked to differential expression of Lamin isoforms, with primary mouse erythroblasts expressing only Lamin B and primary human erythroblasts only Lamin A/C. We did not consistently identify a continuous F-actin ring at the cell surface constriction in mouse erythroblasts, nor at the membrane protein-sorting boundary in human erythroblasts, which do not have a constriction, arguing against a contractile ring-based nuclear expulsion mechanism. However, both mouse and human erythroblasts contain an F-actin structure at the rear of the translocating nucleus, enriched in tropomodulin 1 (Tmod1) and nonmuscle myosin IIB. We investigated Tmod1 function in mouse and human erythroblasts both in vivo and in vitro and found that absence of Tmod1 leads to enucleation defects in mouse fetal liver erythroblasts, and in CD34+ hematopoietic stem and progenitor cells, with increased F-actin in the structure at the rear of the nucleus. This novel structure, the "enucleosome,"maymediatecommoncytoskeletalmechanismsunderlying erythroblast enucleation, notwithstanding the morphological heterogeneity of enucleation across species. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
49. JAK1/2 and BCL2 inhibitors synergize to counteract bone marrow stromal cell-induced protection of AML.
- Author
-
Karjalainen, Riikka, Pemovska, Tea, Liu, Minxia, Javarappa, Komal K., Majumder, Muntasir M., Yadav, Bhagwan, Tang, Jing, Bychkov, Dmitrii, Parsons, Alun, Suvela, Minna, Aittokallio, Tero, Kallioniemi, Olli, Wennerberg, Krister, Knowles, Jonathan, Heckman, Caroline A., Popa, Mihaela, Safont, Mireia Mayoral, McCormack, Emmet, Gjertsen, Bjørn T., and Tamborero, David
- Subjects
- *
ACUTE myeloid leukemia , *BONE marrow , *STROMAL cells , *CHRONIC lymphocytic leukemia , *PROTEIN-tyrosine kinase inhibitors - Abstract
The bone marrow (BM) provides a protective microenvironment to support the survival of leukemic cells and influence their response to therapeutic agents. In acute myeloid leukemia (AML), the high rate of relapse may in part be a result of the inability of current treatment to effectively overcome the protective influence of the BM niche. To better understand the effect of the BM microenvironment on drug responses in AML, we conducted a comprehensive evaluation of 304 inhibitors, including approved and investigational agents, comparing ex vivo responses of primary AML cells in BM stroma-derived and standard culture conditions. In the stroma-based conditions, the AML patient cells exhibited significantly reduced sensitivity to 12% of the tested compounds, including topoisomerase II, B-cell chronic lymphocytic leukemia/lymphoma 2 (BCL2), and many tyrosine kinase inhibitors (TKIs). The loss of TKI sensitivity was most pronounced in patient samples harboring FLT3 or PDGFRB alterations. In contrast, the stroma-derived conditions enhanced sensitivity to Janus kinase (JAK) inhibitors. Increased cell viability and resistance to specific drug classes in the BM stroma-derived conditions was a result of activation of alternative signaling pathways mediated by factors secreted by BM stromal cells and involved a switch from BCL2 to BCLXL-dependent cell survival. Moreover, the JAK1/2 inhibitor ruxolitinib restored sensitivity to the BCL2 inhibitor venetoclax in AML patient cells ex vivo in different model systems and in vivo in an AML xenograft mouse model. These findings highlight the potential of JAK inhibitors to counteract stroma-induced resistance to BCL2 inhibitors in AML. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
50. DNMT3A and TET2 dominate clonal hematopoiesis and demonstrate benign phenotypes and different genetic predispositions.
- Author
-
Buscarlet, Manuel, Bourgoin, Vincent, Lépine, Guylaine, Mollica, Luigina, Busque, Lambert, Provost, Sylvie, Zada, Yassamin Feroz, Barhdadi, Amina, Dubé, Marie-Pierre, and Szuber, Natasha
- Subjects
- *
HEMATOPOIESIS , *ACUTE myeloid leukemia , *BONE marrow , *MYELODYSPLASTIC syndromes , *GENETICS - Abstract
Age-associated clonal hematopoiesis caused by acquired mutations in myeloid cancer-associated genes is highly prevalent in the normal population. Its etiology, biological impact on hematopoiesis, and oncogenic risk is poorly defined at this time. To gain insight into this phenomenon, we analyzed a cohort of 2530 related and unrelated hematologically normal individuals (ages 55 to 101 years). We used a sensitive genetargeted deep sequencing approach to gain precision on the exact prevalence of driver mutations and the proportions of affected genes. Mutational status was correlated with biologicalparameters. We report a higher overall prevalence of driver mutations (13.7%), which occurredmostly (93%) in DNMT3A or TET2 and were highly age-correlated. Mutation in these 2 genes had some distinctive effects on end points. TET2 mutations weremore age-dependent, associated with amodestneutropenic effect (9%,P5.012), demonstrated familial aggregation, and associated with chronic obstructive pulmonary disease. Mutations in DNMT3A had no impact on blood counts or indices. Mutational burden of both genes correlated with Xinactivation skewing but no significant associationwith age-adjusted telomere length reductionwas documented. The discordance between the high prevalence ofmutations in these 2 genes and their limited biological impact raise the question of the potential role of dysregulated epigenetic modifiers in normal aging hematopoiesis, which may include support to failing hematopoiesis. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.