Academic literature on the topic 'Hematopoietic stem cell niche'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'Hematopoietic stem cell niche.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "Hematopoietic stem cell niche"

1

Wattrus, Samuel J., and Leonard I. Zon. "Stem cell safe harbor: the hematopoietic stem cell niche in zebrafish." Blood Advances 2, no. 21 (November 13, 2018): 3063–69. http://dx.doi.org/10.1182/bloodadvances.2018021725.

Full text
Abstract:
Abstract Each stem cell resides in a highly specialized anatomic location known as the niche that protects and regulates stem cell function. The importance of the niche in hematopoiesis has long been appreciated in transplantation, but without methods to observe activity in vivo, the components and mechanisms of the hematopoietic niche have remained incompletely understood. Zebrafish have emerged over the past few decades as an answer to this. Use of zebrafish to study the hematopoietic niche has enabled discovery of novel cell–cell interactions, as well as chemical and genetic regulators of hematopoietic stem cells. Mastery of niche components may improve therapeutic efforts to direct differentiation of hematopoietic stem cells from pluripotent cells, sustain stem cells in culture, or improve stem cell transplant.
APA, Harvard, Vancouver, ISO, and other styles
2

Ribeiro-Filho, Antonio Carlos, Débora Levy, Jorge Luis Maria Ruiz, Marluce da Cunha Mantovani, and Sérgio Paulo Bydlowski. "Traditional and Advanced Cell Cultures in Hematopoietic Stem Cell Studies." Cells 8, no. 12 (December 12, 2019): 1628. http://dx.doi.org/10.3390/cells8121628.

Full text
Abstract:
Hematopoiesis is the main function of bone marrow. Human hematopoietic stem and progenitor cells reside in the bone marrow microenvironment, making it a hotspot for the development of hematopoietic diseases. Numerous alterations that correspond to disease progression have been identified in the bone marrow stem cell niche. Complex interactions between the bone marrow microenvironment and hematopoietic stem cells determine the balance between the proliferation, differentiation and homeostasis of the stem cell compartment. Changes in this tightly regulated network can provoke malignant transformation. However, our understanding of human hematopoiesis and the associated niche biology remains limited due to accessibility to human material and the limits of in vitro culture models. Traditional culture systems for human hematopoietic studies lack microenvironment niches, spatial marrow gradients, and dense cellularity, rendering them incapable of effectively translating marrow physiology ex vivo. This review will discuss the importance of 2D and 3D culture as a physiologically relevant system for understanding normal and abnormal hematopoiesis.
APA, Harvard, Vancouver, ISO, and other styles
3

Kandarakov, Oleg, Alexander Belyavsky, and Ekaterina Semenova. "Bone Marrow Niches of Hematopoietic Stem and Progenitor Cells." International Journal of Molecular Sciences 23, no. 8 (April 18, 2022): 4462. http://dx.doi.org/10.3390/ijms23084462.

Full text
Abstract:
The mammalian hematopoietic system is remarkably efficient in meeting an organism’s vital needs, yet is highly sensitive and exquisitely regulated. Much of the organismal control over hematopoiesis comes from the regulation of hematopoietic stem cells (HSCs) by specific microenvironments called niches in bone marrow (BM), where HSCs reside. The experimental studies of the last two decades using the most sophisticated and advanced techniques have provided important data on the identity of the niche cells controlling HSCs functions and some mechanisms underlying niche-HSC interactions. In this review we discuss various aspects of organization and functioning of the HSC cell niche in bone marrow. In particular, we review the anatomy of BM niches, various cell types composing the niche, niches for more differentiated cells, metabolism of HSCs in relation to the niche, niche aging, leukemic transformation of the niche, and the current state of HSC niche modeling in vitro.
APA, Harvard, Vancouver, ISO, and other styles
4

Goldman, Devorah C., Alexis S. Bailey, Dana L. Pfaffle, Azzah Al Masri, Jan L. Christian, and William H. Fleming. "BMP4 regulates the hematopoietic stem cell niche." Blood 114, no. 20 (November 12, 2009): 4393–401. http://dx.doi.org/10.1182/blood-2009-02-206433.

Full text
Abstract:
Abstract Bone morphogenetic protein 4 (BMP4) is required for mesoderm commitment to the hematopoietic lineage during early embryogenesis. However, deletion of BMP4 is early embryonically lethal and its functional role in definitive hematopoiesis is unknown. Consequently, we used a BMP4 hypomorph to investigate the role of BMP4 in regulating hematopoietic stem cell (HSC) function and maintaining steady-state hematopoiesis in the adult. Reporter gene expression shows that Bmp4 is expressed in cells associated with the hematopoietic microenvironment including osteoblasts, endothelial cells, and megakaryocytes. Although resting hematopoiesis is normal in a BMP4-deficient background, the number of c-Kit+, Sca-1+, Lineage− cells is significantly reduced. Serial transplantation studies reveal that BMP4-deficient recipients have a microenvironmental defect that reduces the repopulating activity of wild-type HSCs. This defect is even more pronounced in a parabiosis model that demonstrates a profound reduction in wild-type hematopoietic cells within the bone marrow of BMP4-deficient recipients. Furthermore, wild-type HSCs that successfully engraft into the BMP4-deficient bone marrow show a marked decrease in functional stem cell activity when tested in a competitive repopulation assay. Taken together, these findings indicate BMP4 is a critical component of the hematopoietic microenvironment that regulates both HSC number and function.
APA, Harvard, Vancouver, ISO, and other styles
5

Park, Dongsu. "The hematopoietic stem cell niche." Frontiers in Bioscience 17, no. 1 (2012): 30. http://dx.doi.org/10.2741/3913.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Frenette, Paul S., Simón Méndez-Ferrer, Daniel Lucas-Alcaraz, Michela Batista, Sergio Lira, Tatyana V. Michurina, and Grigori N. Enikolopov. "The Hematopoietic Stem Cell Niche." Blood 114, no. 22 (November 20, 2009): SCI—49—SCI—49. http://dx.doi.org/10.1182/blood.v114.22.sci-49.sci-49.

Full text
Abstract:
Abstract Abstract SCI-49 The concept of stem cell niche, proposed by Schofield 30 years ago, refers to the ability of the microenvironment to regulate stem cell fate. The niche provides critical signals allowing hematopoietic stem cells (HSC) to survive, and if so, whether to remain in or to leave the niche (mobilization), or whether to remain quiescent or divide. Some of these signals originate locally from the niche cell(s) but others are coming from afar. For example, we have found that signals from the sympathetic nervous system (SNS) promote the release of HSCs from the bone marrow (BM) niche. Under steady-state conditions, HSC egress in blood is orchestrated in a circadian manner where the fluctuations of circulating HSCs/progenitors are matched with antiphase oscillations in the expression of Cxcl12 mRNA in the BM microenvironment. These oscillations are entrained in the brain by the molecular clock through the local delivery of norepinephrine by SNS nerve terminals in the BM, and transmitted specifically by the β3 adrenergic receptor (Adrβ3) expressed on CXCL12-producing stromal cells, thereby leading to the cyclical degradation of the Sp1 transcription factor. In humans, the circadian release of HSC is inverted compared to rodents and may influence the stem cell yield even when mobilization is enforced by granulocyte colony-stimulating factor (G-CSF), suggesting the potential benefit to harvest HSCs in the clinic at the optimal circadian time. Given the coupling of nervous signals with the stem cell niche, we would expect that the stromal cell forming the niche would be intimately associated with nerve fibers. We have recently found using transgenic mice expressing the green fluorescent protein (GFP) under the Nestin promoter elements (Nes-Gfp), that GFP+ cells (referred to as Nestin+) form a HSC niche in the marrow. Nestin+ cells comprise a relatively small subset (0.08 ± 0.01%) of total BM nucleated cells that is anatomically and functionally associated with the vast majority of CD150+ CD48- Lin- HSCs near blood vessels and SNS fibers of the BM. Nestin+ niche cells express high levels of core genes regulating HSC retention (Cxcl12, Kit ligand, Vcam-1, Angiopoietin-1), and these genes are downregulated by mobilization induced by G-CSF or administration of Adrβ3 agonists. We have identified putative Nestin+ niche cells as bona fide mesenchymal stem cells (MSCs) since they can be propagated as single clonal spheres capable of self-renewal, dramatic in vivo expansion, and multipotency to form osteoblasts, adipocytes, and chondocytes. These data argue for a unique bone marrow niche formed by the pairing of the two rare stem cells, mesenchymal and hematopoietic, that exist in the marrow. Co-authors: Simón Méndez-Ferrer, Ph.D., Daniel Lucas, Ph.D., Michela Batista, Ph.D., Sergio A. Lira, M.D., Mount Sinai School of Medicine, New York, NY; Tatyana V. Michurina, Ph.D., Grigori N. Enikolopov Ph.D., Cold Spring Harbor Laboratory, Cold Spring Harbor, NY Disclosures Frenette: Glycomimetic: Research Funding.
APA, Harvard, Vancouver, ISO, and other styles
7

Boulais, Philip E., and Paul S. Frenette. "Making sense of hematopoietic stem cell niches." Blood 125, no. 17 (April 23, 2015): 2621–29. http://dx.doi.org/10.1182/blood-2014-09-570192.

Full text
Abstract:
Abstract The hematopoietic stem cell (HSC) niche commonly refers to the pairing of hematopoietic and mesenchymal cell populations that regulate HSC self-renewal, differentiation, and proliferation. Anatomic localization of the niche is a dynamic unit from the developmental stage that allows proliferating HSCs to expand before they reach the bone marrow where they adopt a quiescent phenotype that protects their integrity and functions. Recent studies have sought to clarify the complexity behind the HSC niche by assessing the contributions of specific cell populations to HSC maintenance. In particular, perivascular microenvironments in the bone marrow confer distinct vascular niches that regulate HSC quiescence and the supply of lineage-committed progenitors. Here, we review recent data on the cellular constituents and molecular mechanisms involved in the communication between HSCs and putative niches.
APA, Harvard, Vancouver, ISO, and other styles
8

Fielding, Claire, and Simón Méndez-Ferrer. "Neuronal regulation of bone marrow stem cell niches." F1000Research 9 (June 16, 2020): 614. http://dx.doi.org/10.12688/f1000research.22554.1.

Full text
Abstract:
The bone marrow (BM) is the primary site of postnatal hematopoiesis and hematopoietic stem cell (HSC) maintenance. The BM HSC niche is an essential microenvironment which evolves and responds to the physiological demands of HSCs. It is responsible for orchestrating the fate of HSCs and tightly regulates the processes that occur in the BM, including self-renewal, quiescence, engraftment, and lineage differentiation. However, the BM HSC niche is disturbed following hematological stress such as hematological malignancies, ionizing radiation, and chemotherapy, causing the cellular composition to alter and remodeling to occur. Consequently, hematopoietic recovery has been the focus of many recent studies and elucidating these mechanisms has great biological and clinical relevance, namely to exploit these mechanisms as a therapeutic treatment for hematopoietic malignancies and improve regeneration following BM injury. The sympathetic nervous system innervates the BM niche and regulates the migration of HSCs in and out of the BM under steady state. However, recent studies have investigated how sympathetic innervation and signaling are dysregulated under stress and the subsequent effect they have on hematopoiesis. Here, we provide an overview of distinct BM niches and how they contribute to HSC regulatory processes with a particular focus on neuronal regulation of HSCs under steady state and stress hematopoiesis.
APA, Harvard, Vancouver, ISO, and other styles
9

Chan, Charles, Ching-Cheng Chen, Daniel L. Kraft, Cynthia Luppen, Jae-Beom Kim, Anthony DeBoer, Kevin Wei Wei, and Irving L. Weissman. "Identification and Isolation of the Hematopoietic Stem Cell Niche Initiating Cell Population." Blood 112, no. 11 (November 16, 2008): 3574. http://dx.doi.org/10.1182/blood.v112.11.3574.3574.

Full text
Abstract:
Abstract Introduction: Identification and understanding of the cells and processes that can generate, sustain and influence the HSC niche and hematopoiesis are critical for the development of a more comprehensive knowledge of normal hematopoiesis, stem cell homing, trafficking, differentiation and hematopoietic pathology. Growth and renewal in many tissues are initiated by stem cells, supported by the microenvironment (niche) in which they reside. While recent work has begun to describe functional interactions between stem cells and their niches, little is known about the formation of stem cell niches. Methods & Results: We established a functional, in vivo assay (via implantation of cells under the renal capsule) to isolate the determinants of hematopoietic stem cell (HSC) niche formation and activity. Using this novel assay, we show that a population of progenitor cells (CD45−Tie2-aV+CD105+Thy1.1−; CD105+Thy1−) sorted from 15.5 dpc fetal limbs and transplanted under the adult mouse renal capsule recruit host-derived vasculatures in a VEGF dependent manner, produce donor-derived ectopic bones through endochondral ossification, and generate a marrow cavity populated by host-derived long term reconstituting HSC (LT-HSC). In contrast, CD45−Tie2-aV+CD105+Thy1a+ (CD105+Thy1+) progenitors form bone that does not contain a marrow cavity. While analyzing these and other sorted populations, we did not observe any instances where niche was present without bone, suggesting that skeletal progenitors are necessary for initiating an HSC niche but osteoblasts alone cannot initiate and support niche activity. Suppression of factors important for HSC maintenance, such as steel factor (SLF), in progenitor populations prior to transplant did not alter their ability to initiate and support an HSC niche. On the other hand, suppression of factors involved in endochondral ossification, such as osterix and VEGF, inhibited niche generation. Furthermore, CD105+Thy1− progenitor populations derived from regions of the fetal mandible or calvaria that do not undergo endochondral ossification form only bone without marrow in our assay. Conclusions: In addition to identifying the limb-derived skeletal progenitor capable of endochondral ossification involved and the basic mechanisms of HSC niche initiation, our study provides a functional framework by which future studies on HSC-niche interactions at the cellular level can be carried out.
APA, Harvard, Vancouver, ISO, and other styles
10

Esmaeli-Azad, Babak, Anand S. Srivastava, Cybele Frederico, Geraldo Martinez, Satoshi Yasukawa, and Ewa Carrier. "Artificial Hematopoietic Stem Cell Niche Sustains Growth and Differentiation of Human ES-Derived Early Hematopoietic Progenitors." Blood 110, no. 11 (November 16, 2007): 1415. http://dx.doi.org/10.1182/blood.v110.11.1415.1415.

Full text
Abstract:
Abstract Using a novel Microplate Biomaterial Microarray (MBM™) technology, we have created an artificial hematopoietic stem cell niche that can sustain growth and differentiation of human embryonic stem cells-derived (hES) early hematopoietic progenitors. This hydrogel based ex-vivo niche allows uploading of human embryonal stem cells, human mesenchymal stem cells (MSC), genes (bcl-2 preventing apoptosis and HoxB4 enhancing hematopoiesis) and extracellular matrices to support growth and differentiation of human ES cells. These experiments were done using NIH-approved hES cell lines H1 and H9. Serum-free, feeder-free culture conditions were established and early hematopoietic progenitors grown using SCF, TPO, VEGF and IL-3 with high efficiency. At day 3–5 dual CD34+/CD31+ progenitors were identified, while on day 7–8 CD34+ hematopoietic progenitors were isolated, which formed typical hematopoietic colonies. These progenitors expressed genes related to early hematopoiesis, such as TAL1/SCL, FLT1, GATA2, GATA1, EPOR and TPOR. The early dual endothelio-hematopoietic progenitor (hemangioblast) expressed PECAM-1 and CD34 and showed typical blast-like morphology. Based on mathematical simulations, various micro-niches were designed to establish optimal differentiation conditions for this progenitor using IL-3, IL-6, TPO, EPO, VEGF, SFC, Flt-3 ligand and various extracellular matrices. Specific micro-niches were created for generation of CFU-E, BFU-E, CFU-GM, CFU-GEMM, CFU-M, CFU-G, and CFU-MK progenitors from human ES-derived hemangioblast. Kinetic uploading of TPO, EPO, SCF and VEGF created a niche-sustaining growth of ES-derived hemangioblast with high efficiency and low apoptosis rate. These niches used pulse -delivery of anti-apoptotic bcl-2 gene and hematopoiesis-enhancing Hoxb4 gene. The model of artifical niche sustaing growth and differentiation of human ES-derived hemangioblast was established. In the future, this system will allow optimized and upscaled generation of early hematopoietic progenitors from human ES cells, as a first step towards clinical applications of human embryonic stem cells. Figure Figure
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "Hematopoietic stem cell niche"

1

Franke, Katja. "Adhesion and Single Cell Tracking of Hematopoietic Stem Cells on Extracellular Matrices." Doctoral thesis, Saechsische Landesbibliothek- Staats- und Universitaetsbibliothek Dresden, 2011. http://nbn-resolving.de/urn:nbn:de:bsz:14-qucosa-77290.

Full text
Abstract:
The local microenvironment of hematopoietic stem cells (HSCs) in the bone marrow -referred to as stem cell niche- is thought to regulate the balance of stem cell maintenance and differentiation by a complex interplay of extrinsic signals including spatial constraints, extracellular matrix (ECM) components and cell-cell interactions. To dissect the role of niche ECM components, a set of well-defined matrix biomolecular coatings including fibronectin, laminin, collagen IV, tropocollagen I, heparin, heparan sulphate, hyaluronic acid and co-fibrils of collagen I with heparin or hyaluronic acid were prepared and analyzed with respect to adhesive interactions of human CD133+ HSCs in vitro. ECM molecule dependent adhesion areas as well as fractions of adherent HSCs were assessed by reflection interference contrast microscopy and differential interference contrast microscopy. HSCs, so far mostly classified as suspension cells, exhibited intense adhesive interactions with fibronectin, laminin, collagen IV, heparin, heparan sulphate, and collagen I based co-fibrils. An integrin mediated adhesion on fibronectin and a L-selectin mediated adhesion on heparin pointed to specific interactions based on different adhesion mechanisms. As a consequence of HSC adhesion to molecules of the vascular and the endosteal regions, both regions were confirmed as possible stem cell niches and adhesive signals were suggested as potential regulators of stem cell fate. Furthermore, the impact of a spatially organized ECM on the HSC behavior was analyzed by single cell tracking. These studies required the development of engineered three-dimensional, ECM coated microcavities with the option for single cell tracking. A semi-automated cell-tracking tool was established to accelerate data access from time-lapse image sequences. From this analysis it was possible to reveal the genealogy, localization, morphology and migration of single HSCs over a time period of 4 days. A decreased cycling frequency was observed depending on the HSC localization in the spatially constraining microcavities. Besides the revealed impact of spatial constraints on HSC fate, the newly engineered ECM-coated microcavity setup and the semi-automated cell tracking tool provide new options to study the cell fate in engineered microenvironments at single cell level for other cell types ex vivo
Die lokale Mikroumgebung von Blutstammzellen (BSZ) im Knochenmark, bezeichnet als Stammzellnische, reguliert das Gleichgewicht von Stammzellerhaltung und -differenzierung durch ein komplexes Zusammenspiel von extrinsischen Signalen wie räumliche Beschränkungen, Komponenten der extrazellulären Matrix (EZM) und Zell-Zell Wechselwirkungen. Um die Rolle der EZM-Komponenten zu analysieren, wurden definierte Beschichtungen von Fibronektin, Laminin, Kollagen IV, monomerem Kollagen I, Heparin, Heparan Sulphat, Hyaluronsäure und Co-Fibrillen aus Kollagen I und Heparin oder Hyaluronsäure hergestellt und in vitro bezüglich der adhäsiven Wechselwirkungen von humanen CD133+ BSZ untersucht. Die Adhäsionsflächen und der Anteil adhärenter Zellen wurden in Abhängigkeit von der EZM-Beschichtung mittels Reflexions- Interferenz-Kontrast-Mikroskopie und Differentieller Interferenz Kontrast Mikroskopie bestimmt. BSZ, bisher als Suspensionszellen definiert, zeigten intensive adhäsive Wechselwirkungen mit Fibronektin, Laminin, Kollagen IV, Heparin, Heparan Sulphat und den Co-Fibrillen. Eine Integrin abhängige Adhäsion auf Fibronektin und eine L-Selektin abhängige Adhäsion auf Heparin, wiesen auf spezifische Wechselwirkungen hin, die auf unterschiedlichen Mechanismen basieren. Aufgrund der Adhäsion von BSZ sowohl zu Molekülen der vaskulären als auch der endostealen Knochenmarkregion, wurden beide Bereiche als mögliche Stammzellnische bestätigt. Adhäsive Signale sind potentielle Regulatoren der Stammzellentwicklung. Im Weiteren wurde der Einfluss einer räumlich beschränkenden EZM auf das Verhalten der BSZ durch Einzelzellverfolgung untersucht. Diese Studien erforderten die Entwicklung von dreidimensionalen EZM-beschichteten Mikrokavitäten, die das Verfolgen einzelner Zellen ermöglichten. Es wurde ein halbautomatischer Algorithmus für die Zellverfolgung etabliert, um die Datengenerierung von den Zeitreihenaufnahmen zu beschleunigen. Die Analysen ermöglichten Aussagen über die Genealogie, Lokalisierung, Morphologie und Migration einzelner BSZ während einer Analysenzeit von 4 Tagen. Eine verringerte Zellteilungsaktivität wurde in Abhängigkeit von der BSZ Lokalisierung innerhalb der räumlich einschränkenden Mikrokavitäten festgestellt. Neben diesen Erkenntnissen bieten die entwickelten Mikrokavitäten und die etablierte Einzelzellverfolgung neue Möglichkeiten auch andere Zelltypen auf Einzelzellniveau ex vivo zu untersuchen
APA, Harvard, Vancouver, ISO, and other styles
2

Liu, Wei. "Rational targeting of Cdc42 in hematopoietic stem cell mobilization and engraftment." University of Cincinnati / OhioLINK, 2011. http://rave.ohiolink.edu/etdc/view?acc_num=ucin1303845649.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Kräter, Martin. "Bone marrow niche-mimetics modulate hematopoietic stem cell function via adhesion signaling in vitro." Doctoral thesis, Saechsische Landesbibliothek- Staats- und Universitaetsbibliothek Dresden, 2017. http://nbn-resolving.de/urn:nbn:de:bsz:14-qucosa-230268.

Full text
Abstract:
As graft source for lymphoma or leukemia treatment, hematopoietic stem and progenitor cells (HSPCs) have been the focus of translational medicine for decades. HSPCs are defined by their self-renewing capacity and their ability to give rise to all mature blood cells. They are found anchored to a specialized microenvironment in the bone marrow (BM) called the hematopoietic niche. HSPCs can be enriched by sorting them based on the presence of the surface antigen CD34 before clinical or tissue engineering use. As these cells represent a minority in most graft sources and the amount of applicable cells is limited, ex vivo expansion-cultures were established using cytokine cocktails or small molecules. However, in vitro culture of HSPCs as suspension-cultures result in heterogeneous cell populations with undefined cellular identities. In the BM niche, HSPCs are not exclusively maintained by cytokines but also by cell-matrix adhesions mediated by integrins (ITGs). Thus, β1 and β2 ITGs were found to promote initial contact of HSPCs with mesenchymal stromal cells (MSCs) and ITGβ3 expression was shown to be a marker for long-term repopulating HSPCs in vivo. Consequently, ex vivo remodeling of the BM niche using co-cultures of HSPCs and niche cells like MSCs came into spotlight and was proven to be a promising tool for stem cell expansion. However, in clinical and research applications, direct contact of two cell populations necessitates HSPC post-culture purification. To address these problems, we established a novel culture method for remodeling the BM extra cellular stroma in vitro wherein we used decellularized extracellular matrix (ECM) scaffolds derived from immortalized mesenchymal stromal cells (SCP-1). Such scaffolds were found to be highly reproducible and served as in vitro niche for HSPCs by being more effective for the expansion of CD34+ cells, compared to classical suspension cultures. ECMs were shown to consist of multiple proteins including fibronectins, collagens, and a major niche chemokine responsible for BM homing and retention of HSPCs in vivo, namely, stromal derived factor 1 (SDF-1). SDF-1 is known to be secreted by MSCs and is anchored to matrix proteins. This reveals that ECM scaffolds produced by SCP-1 cells are a naïve reconstructed microenvironment. When CD34+ cells were seeded, only around 20% of the cells adhered to the provided ECM scaffold. These cells recognized SDF-1 via C-X-C chemokine receptor type 4 (CXCR-4), as shown by laser scanning confocal microscopy. Thus, adhesive sides as they are present in the BM niche are provided. However, CD34+ cells isolated from G-CSF mobilized peripheral blood of healthy donors were found to be heterogenous with respect to adhesion capacity. Nonetheless, it was similar to HSPC co-cultures with SCP-1 cells as feeder layer. Therefore, we separated and analyzed two cell fractions, the adherent (AT-cells) and the non- adherent supernatant (SN-cells) cells. Other signals provided by the BM extracellular stroma to HSPCs are physical cues that control HSPC fate. HSPCs sense these physical features through focal contacts and accordingly remodel their morphological and biomechanical properties. Using real-time deformability cytometry (RT-DC) to uncover biomechanical phenotypes of freshly isolated HSPCs, SN-cells, AT-cells, and classical suspension cultured HSPCs in plastic culture dishes (PCD) were analyzed. We found freshly isolated cells to be less deformable and small. AT-cells displayed actin polymerization to stress fibers, and exhibited a stiffer mechanical phenotype compared to PCD-cultured or SN-cells. This might constitute the first hint of functional adaptation. Integrins are known to establish mechanosensing focal contacts. Thus, we analyzed ITG surface expression and identified ITGαIIb, ITGαV, and ITGβ3 to be enriched on AT-cells compared to freshly isolated cells or SN-cells. Active integrins need to form heterodimers consisting of one α- and one β subunit. Interestingly, the identified ITGs exclusively interact with each other to form RGD peptide receptors. RGD is a tripeptide consisting of the amino acids arginine, glycine, and aspartic acid and was identified as an adhesion sequence within fibronectin and other extracellular proteins. Consequently, we could confirm an important role for ITGαVβ3 in HSPC- ECM interaction with respect to adhesion and migration. However, we also identified ITGβ3 expression on a subset of CD34+ cells either freshly isolated or ECM cultured cells, as a marker for erythrocyte differentiation. These findings demonstrate that, in vitro, the ECM compartment acts as a regulator of HSPC fate and portray mechanical recognition as a potent driver of differentiation. In this context, targeted modulation of ECM scaffolds could enhance cell-ECM interactions and accelerate stem cell expansion or differentiation. These modulations could also provide further insights into HSPC-niche regulation. We demonstrate that ECMs derived from osteogenic differentiated SCP-1 cells increase HSPC expansion but do not lead to increased cell adhesion. As ECM adhesion preliminary alters HSPC function, we aimed at developing ECM scaffolds with increased adhesion capacity. Using lentiviral transduction, we generated a stable knock down of fibulin-1 in SCP-1 cells. Fibulin-1 is an ECM protein known to form anti-adhesion sites with fibronectin. However, we failed to increase adherent cell numbers or enhance HSPC expansion in the fibulin-1 knock down ECMs. Taken together, SCP-1 cell-derived ECM protein scaffolds provide an in vitro niche for HSPCs capable of stem cell expansion. Integrin mediated signaling altered the biomechanical and functional properties of HSPCs and hints at suspension cultures as being inappropriate to study the physiological aspects of HSPCs. Targeted modulation of ECM scaffolds could theoretically generate suitable ex vivo environments with the capacity to gain detailed insight into HSPC regulation within their niche. This will enhance the functionality of new biomaterials and will lead to improved regenerative therapies like BM transplantation.
APA, Harvard, Vancouver, ISO, and other styles
4

Kokkaliaris, Konstantinos [Verfasser], and Heinrich [Akademischer Betreuer] Leonhardt. "Identification of novel niche molecules controlling hematopoietic stem cell behavior / Konstantinos Kokkaliaris ; Betreuer: Heinrich Leonhardt." München : Universitätsbibliothek der Ludwig-Maximilians-Universität, 2016. http://d-nb.info/1115144944/34.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Shimoto, Manabu. "Numerous niches for hematopoietic stem cells remain empty during homeostasis." Kyoto University, 2017. http://hdl.handle.net/2433/226772.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Sugimura, Ryohichi. "Non-canonical Wnt signaling maintains hematopoietic stem cell through Flamingo and Frizzled8 interaction in the niche." Thesis, Open University, 2012. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.580673.

Full text
Abstract:
Wnt signaling is involved in self-renewal and maintenance ofhematopoietic stem cells (HSCs); however, the particular role of non-canonical Wnt signaling in regulating HSCs in vivo is largely unknown. Here I show Flamingo and Frizzled8, members of non-canonical Wnt signaling, both express in and functionally maintain quiescent long-term HSCs. Flamingo regulates Frizzled8 distribution at the interface between HSCs and N-cadherin + osteoblasts (N-cad+OBs that enrich osteoprogenitors) in the niche. I further show that N-cad+OBs predominantly express non-canonical Wnt ligands and inhibitors of canonical Wnt signaling under homeostasis. This non-canonical Wnt signaling is attenuated prior to activation of HSCs. In the activated HSCs, however, canonical Wnt signaling is enhanced. Mechanistically, non-canonical Wnt signaling mediated by Frizzled8 suppresses the Ca2+-NF AT- IFNy pathway and antagonizes canonical Wnt signaling in HSCs. My findings demonstrate that non-canonical Wnt signaling maintains quiescent long- term HSCs through Flamingo and Frizzled8 interaction in the niche.
APA, Harvard, Vancouver, ISO, and other styles
7

Hazen, Amy L. "Inositol phospholipid and tyrosine phosphorylation signaling in the biology of hematopoietic stem cells." [Tampa, Fla] : University of South Florida, 2009. http://digital.lib.usf.edu/?e14.2829.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Ordemann, Rainer, Duohui Jing, Ana-Violeta Fonseca, Nael Alakel, Fernando A. Fierro, Katrin Muller, Martin Bornhauser, Gerhard Ehninger, and Denis Corbeil. "Hematopoietic stem cells in co-culture with mesenchymal stromal cells - modeling the niche compartments in vitro." Saechsische Landesbibliothek- Staats- und Universitaetsbibliothek Dresden, 2016. http://nbn-resolving.de/urn:nbn:de:bsz:14-qucosa-177403.

Full text
Abstract:
Background Hematopoietic stem cells located in the bone marrow interact with a specific microenvironment referred to as the stem cell niche. Data derived from ex vivo co-culture systems using mesenchymal stromal cells as a feeder cell layer suggest that cell-to-cell contact has a significant impact on the expansion, migratory potential and ‘stemness’ of hematopoietic stem cells. Here we investigated in detail the spatial relationship between hematopoietic stem cells and mesenchymal stromal cells during ex vivo expansion. Design and Methods In the co-culture system, we defined three distinct localizations of hematopoietic stem cells relative to the mesenchymal stromal cell layer: (i) those in supernatant (non-adherent cells); (ii) those adhering to the surface of mesenchymal stromal cells (phase-bright cells) and (iii) those beneath the mesenchymal stromal cells (phase-dim cells). Cell cycle, proliferation, cell division and immunophenotype of these three cell fractions were evaluated from day 1 to 7. Results Phase-bright cells contained the highest proportion of cycling progenitors during co-culture. In contrast, phase-dim cells divided much more slowly and retained a more immature phenotype compared to the other cell fractions. The phase-dim compartment was soon enriched for CD34+/CD38− cells. Migration beneath the mesenchymal stromal cell layer could be hampered by inhibiting integrin β1 or CXCR4. Conclusions Our data suggest that the mesenchymal stromal cell surface is the predominant site of proliferation of hematopoietic stem cells, whereas the compartment beneath the mesenchymal stromal cell layer seems to mimic the stem cell niche for more immature cells. The SDF-1/CXCR4 interaction and integrin-mediated cell adhesion play important roles in the distribution of hematopoietic stem cells in the co-culture system.
APA, Harvard, Vancouver, ISO, and other styles
9

Ordemann, Rainer, Duohui Jing, Ana-Violeta Fonseca, Nael Alakel, Fernando A. Fierro, Katrin Muller, Martin Bornhauser, Gerhard Ehninger, and Denis Corbeil. "Hematopoietic stem cells in co-culture with mesenchymal stromal cells - modeling the niche compartments in vitro." Ferrata Storti Foundation, 2010. https://tud.qucosa.de/id/qucosa%3A28891.

Full text
Abstract:
Background Hematopoietic stem cells located in the bone marrow interact with a specific microenvironment referred to as the stem cell niche. Data derived from ex vivo co-culture systems using mesenchymal stromal cells as a feeder cell layer suggest that cell-to-cell contact has a significant impact on the expansion, migratory potential and ‘stemness’ of hematopoietic stem cells. Here we investigated in detail the spatial relationship between hematopoietic stem cells and mesenchymal stromal cells during ex vivo expansion. Design and Methods In the co-culture system, we defined three distinct localizations of hematopoietic stem cells relative to the mesenchymal stromal cell layer: (i) those in supernatant (non-adherent cells); (ii) those adhering to the surface of mesenchymal stromal cells (phase-bright cells) and (iii) those beneath the mesenchymal stromal cells (phase-dim cells). Cell cycle, proliferation, cell division and immunophenotype of these three cell fractions were evaluated from day 1 to 7. Results Phase-bright cells contained the highest proportion of cycling progenitors during co-culture. In contrast, phase-dim cells divided much more slowly and retained a more immature phenotype compared to the other cell fractions. The phase-dim compartment was soon enriched for CD34+/CD38− cells. Migration beneath the mesenchymal stromal cell layer could be hampered by inhibiting integrin β1 or CXCR4. Conclusions Our data suggest that the mesenchymal stromal cell surface is the predominant site of proliferation of hematopoietic stem cells, whereas the compartment beneath the mesenchymal stromal cell layer seems to mimic the stem cell niche for more immature cells. The SDF-1/CXCR4 interaction and integrin-mediated cell adhesion play important roles in the distribution of hematopoietic stem cells in the co-culture system.
APA, Harvard, Vancouver, ISO, and other styles
10

Buglass, Surahanil Katrin. "Regulating stem cell fate within microenvironmental niches." Thesis, University of Oxford, 2014. http://ora.ox.ac.uk/objects/uuid:75f9498c-30f0-4983-84b2-dd58f2ccf52b.

Full text
Abstract:
Improving the repopulation potential of human umbilical cord blood (UCB) haemopoietic stem cells (HSCs) remains a paramount goal in HSC transplantation (HSCT) therapy. This implies enhancing the homing and engraftment potential of UCB-CD34+CD133+ cells to the bone marrow (BM). Although an array of molecules continues to be identified as ‘key’ homing molecules, the molecular mechanisms controlling HSC homing are still not fully understood. The regulatory implications of hypoxia in the BM, with the concomitant stabilisation of hypoxia inducible transcription factor-1α (HIF-1α), are becoming more apparent, yet at the commencement of this thesis no study had explored whether hypoxia induced signalling can be adopted to regulate the homing and engraftment of transplanted HSCs. The aim of this DPhil project was thus to investigate whether hypoxic conditions as detected in the BM influence the adhesion of UBC-CD133+ cells to osteoblasts, BM stromal cells and BM endothelial cells-60 (BMEC-60), as well as their transmigration towards chemokine SDF-1α across BMEC-60. Increasing the exposure of UCB-CD133+ cells to 1.5% O2 doubled the percentage of transmigrating cells (p<0.05), and while hypoxia stimulated UCB-CD133+ cells preferentially adhered to IL-1β stimulated BMEC-60, their adhesion to non-stimulated (BMEC-60) was significantly improved (p<0.001). To help unravel the underlying molecular mechanisms, we attempted to examine the potential involvement of hypoxia regulated scaffolding protein HEF-1/NEDD9/Cas-L (HEF-1) in the increased percentage of migrating UCB-CD133+ cells after hypoxia pre-conditioning. The role of HEF-1 in HSCs is unexplored, and its multifunctional contribution in a variety of processes including cell migration, attachment and invasion make HEF-1 a prime candidate as a contributing homing molecule. After identifying a suitable short-hairpin RNA (shRNA) sequence to knockdown HEF-1, generating lentiviral (LV)-particles in house and optimising transduction protocols, HEF-1 knockdown was achieved in haemopoietic model cell lines KG-1 and KG-1A (KG-1/KG-1A–HEF1). Significantly decreased KG-1A–HEF1 cell adhesion to non-stimulated BMEC-60 was detected. Together, these studies provide a promising platform to further explore the role of HEF-1 in hypoxia induced UCB-CD133+ cell transmigration towards the key homing molecule SDF-1α.
APA, Harvard, Vancouver, ISO, and other styles

Books on the topic "Hematopoietic stem cell niche"

1

Turksen, Kursad, ed. Stem Cell Niche. Totowa, NJ: Humana Press, 2013. http://dx.doi.org/10.1007/978-1-62703-508-8.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Turksen, Kursad, ed. Stem Cell Niche. New York, NY: Springer New York, 2019. http://dx.doi.org/10.1007/978-1-4939-9508-0.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

J, Forman Stephen, Blume Karl G, and Thomas E. Donnall, eds. Hematopoietic cell transplantation. 2nd ed. Oxford: Blackwell Science, 1999.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
4

Klug, Christopher A., and Craig T. Jordan. Hematopoietic Stem Cell Protocols. New Jersey: Humana Press, 2001. http://dx.doi.org/10.1385/159259140x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Soiffer, Robert J., ed. Hematopoietic Stem Cell Transplantation. Totowa, NJ: Humana Press, 2008. http://dx.doi.org/10.1007/978-1-59745-438-4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Bishop, Michael R., ed. Hematopoietic Stem Cell Transplantation. Boston, MA: Springer US, 2009. http://dx.doi.org/10.1007/978-0-387-78580-6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Bunting, Kevin D., ed. Hematopoietic Stem Cell Protocols. Totowa, NJ: Humana Press, 2008. http://dx.doi.org/10.1007/978-1-59745-182-6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Bunting, Kevin D., and Cheng-Kui Qu, eds. Hematopoietic Stem Cell Protocols. New York, NY: Springer New York, 2014. http://dx.doi.org/10.1007/978-1-4939-1133-2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Godin, Isabelle, and Ana Cumano. Hematopoietic Stem Cell Development. Boston, MA: Springer US, 2006. http://dx.doi.org/10.1007/978-0-387-33535-3.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Kondo, Motonari, ed. Hematopoietic Stem Cell Biology. Totowa, NJ: Humana Press, 2010. http://dx.doi.org/10.1007/978-1-60327-347-3.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Book chapters on the topic "Hematopoietic stem cell niche"

1

Tan, Keai Sinn, Nathalie Brouard, and Daisuke Sugiyama. "Analysis of Hematopoietic Niche in the Mouse Embryo." In Stem Cell Niche, 13–27. New York, NY: Springer New York, 2018. http://dx.doi.org/10.1007/7651_2018_176.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Costa, Marta H. G., Tiago S. Monteiro, Susana Cardoso, Joaquim M. S. Cabral, Frederico Castelo Ferreira, and Cláudia L. da Silva. "Three-Dimensional Co-culture of Human Hematopoietic Stem/Progenitor Cells and Mesenchymal Stem/Stromal Cells in a Biomimetic Hematopoietic Niche Microenvironment." In Stem Cell Niche, 101–19. New York, NY: Springer New York, 2018. http://dx.doi.org/10.1007/7651_2018_181.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Iwasaki, Hiroko, and Toshio Suda. "Hematopoietic Stem Cells and Their Niche." In Hematopoietic Stem Cell Biology, 37–55. Totowa, NJ: Humana Press, 2009. http://dx.doi.org/10.1007/978-1-60327-347-3_2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Chitteti, Brahmananda Reddy, Monique Bethel, Sherry L. Voytik-Harbin, Melissa A. Kacena, and Edward F. Srour. "In Vitro Construction of 2D and 3D Simulations of the Murine Hematopoietic Niche." In Stem Cell Niche, 43–56. Totowa, NJ: Humana Press, 2013. http://dx.doi.org/10.1007/978-1-62703-508-8_5.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Sugiyama, Daisuke, and Tatsuya Sasaki. "Isolation of Embryonic Hematopoietic Niche Cells by Flow Cytometry and Laser Capture Microdissection." In Stem Cell Niche, 57–65. Totowa, NJ: Humana Press, 2013. http://dx.doi.org/10.1007/978-1-62703-508-8_6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Park, Sun H., Matthew R. Eber, Russell S. Taichman, and Yusuke Shiozawa. "Determining Competitive Potential of Bone Metastatic Cancer Cells in the Murine Hematopoietic Stem Cell Niche." In Stem Cell Niche, 141–50. New York, NY: Springer New York, 2018. http://dx.doi.org/10.1007/7651_2018_178.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Hoang, Van T., Isabel Hoffmann, Karina Borowski, Abraham Zepeda-Moreno, Dan Ran, Eike C. Buss, Patrick Wuchter, Volker Eckstein, and Anthony D. Ho. "Identification and Separation of Normal Hematopoietic Stem Cells and Leukemia Stem Cells from Patients with Acute Myeloid Leukemia." In Stem Cell Niche, 217–30. Totowa, NJ: Humana Press, 2013. http://dx.doi.org/10.1007/978-1-62703-508-8_19.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Mise-Omata, Setsuko, Takahiro S. Doi, Kazuhiro Aoki, and Yuichi Obata. "Impact of Radiation on Hematopoietic Niche." In Stem Cell Biology and Regenerative Medicine, 147–60. Cham: Springer International Publishing, 2015. http://dx.doi.org/10.1007/978-3-319-21702-4_6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Childress, Paul J., Marta B. Alvarez, Brahmananda R. Chitteti, Melissa A. Kacena, and Edward F. Srour. "The Hematopoietic Stem Cell Niche: Cell-Cell Interactions and Quiescence." In Stem Cell Biology and Regenerative Medicine, 1–22. Cham: Springer International Publishing, 2015. http://dx.doi.org/10.1007/978-3-319-21702-4_1.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Gottwald, Eric, Cordula Nies, Patrick Wuchter, Rainer Saffrich, Roman Truckenmüller, and Stefan Giselbrecht. "A Microcavity Array-Based 3D Model System of the Hematopoietic Stem Cell Niche." In Stem Cell Mobilization, 85–95. New York, NY: Springer New York, 2019. http://dx.doi.org/10.1007/978-1-4939-9574-5_7.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Conference papers on the topic "Hematopoietic stem cell niche"

1

Giles, Amber J., Meera Murgai, Yorleny Vicioso, Steven Highfil, Crystal Mackall, Leonard Wexler, David Lyden, and Rosandra Natasha Kaplan. "Abstract 3058: Bone marrow hematopoietic stem cell niche activation and mobilization fosters the metastatic niche." In Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA. American Association for Cancer Research, 2014. http://dx.doi.org/10.1158/1538-7445.am2014-3058.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Giles, Amber J., Meera Murgai, Yorleny Vicioso, Steven Highfill, Miki Kasai, Linda Vahdat, Leonard Wexler, Crystal Mackall, David Lyden, and Rosandra Kaplan. "Abstract 4725: Hematopoietic stem cell niche activation and progenitor mobilization mediate cancer-associated immunosuppression and metastasis." In Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA. American Association for Cancer Research, 2015. http://dx.doi.org/10.1158/1538-7445.am2015-4725.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Alonso, Salvador, Meng Su, Richard Jones, and Gabriel Ghiaur. "Abstract 4842: The stem cell niche detoxifies chemotherapy and protects malignant hematopoietic cells via expression of cytochrome P450 enzymes." In Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA. American Association for Cancer Research, 2014. http://dx.doi.org/10.1158/1538-7445.am2014-4842.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Park, ES, YJ Chung, and PD Aplan. "PO-020 Discrepancy in efficacy of disulfiram between NUP98-PHF23 fusion acute myelogenous leukaemia cell line andin vivomouse model: sharing normal hematopoietic stem cells niche." In Abstracts of the 25th Biennial Congress of the European Association for Cancer Research, Amsterdam, The Netherlands, 30 June – 3 July 2018. BMJ Publishing Group Ltd, 2018. http://dx.doi.org/10.1136/esmoopen-2018-eacr25.555.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Luo, Biquan, Ben S. Lam, Sung Hyung Lee, Shiuan Wey, Hui Zhou, Miao Wang, Si-Yi Chen, Gregor B. Adams, and Amy S. Lee. "Abstract LB-49: The endoplasmic reticulum chaperone protein GRP94 is required for maintaining hematopoietic stem cell interactions with the adult bone marrow niche." In Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL. American Association for Cancer Research, 2011. http://dx.doi.org/10.1158/1538-7445.am2011-lb-49.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Ohsuga, Mieko, Yuma Tada, and Jun Ishikawa. "Interactive environment for hematopoietic stem-cell transplant patients." In 2017 International Conference on Virtual Rehabilitation (ICVR). IEEE, 2017. http://dx.doi.org/10.1109/icvr.2017.8007469.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Bergeron, Anne, Cendrine Godet, Sylvie Chevret, Gwenaël Lorillon, Régis Peffault de Latour, Marie Robin, Patricia Ribaud, Gérard Socié, and Abdellatif Tazi. "Bronchial Disorders After Allogeneic Hematopoietic Stem Cell Transplantation." In American Thoracic Society 2011 International Conference, May 13-18, 2011 • Denver Colorado. American Thoracic Society, 2011. http://dx.doi.org/10.1164/ajrccm-conference.2011.183.1_meetingabstracts.a4666.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Hartigan, Adam, John Westwick, Gabor Jarai, and Cory Hogaboam. "CCR7 Deficiency Enhances Hematopoietic Stem Cell And Myeloid Progenitor Cell Proliferation And Ameliorates Susceptibility To Invasive Pulmonary Aspergillosis Following Hematopoietic Stem Cell Transplantation." In American Thoracic Society 2010 International Conference, May 14-19, 2010 • New Orleans. American Thoracic Society, 2010. http://dx.doi.org/10.1164/ajrccm-conference.2010.181.1_meetingabstracts.a5117.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Simion, Luminita, Georgina Shaw, Florin Zugun Eloae, Paul Botez, Marry Murphy, and Frank Barry. "Stem Cell Niche in Osteoarthritic Joints - Preliminary Results." In 2010 Advanced Technologies for Enhancing Quality of Life (ATEQUAL). IEEE, 2010. http://dx.doi.org/10.1109/atequal.2010.9.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Farhat, R., A. C. Miller, and D. Picker. "Polymicrobial Lung Abscess Complicating Autologous Hematopoietic Stem Cell Transplant." In American Thoracic Society 2019 International Conference, May 17-22, 2019 - Dallas, TX. American Thoracic Society, 2019. http://dx.doi.org/10.1164/ajrccm-conference.2019.199.1_meetingabstracts.a6855.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Reports on the topic "Hematopoietic stem cell niche"

1

Dorshkind, Kenneth. Effects of Hematopoietic Stem Cell Age on CML Disease Progression. Fort Belvoir, VA: Defense Technical Information Center, March 2006. http://dx.doi.org/10.21236/ada451341.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Chepko, Gloria, and Leena Hilakivi-Clarke. Role of the Stem Cell Niche in Hormone-induced Tumorigenesis in Fetal Mouse Mammary Epithelium. Fort Belvoir, VA: Defense Technical Information Center, August 2006. http://dx.doi.org/10.21236/ada471087.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Liu, Huan, Hui Huang, Jia Guo, Chengyuan Li, Jiandang Zhou, Qifeng Yi, Wei Hua, and Lihong Zeng. Effects of aerobic exercise on fatigue in patients with hematopoietic stem cell transplantation: a meta analysis. INPLASY - International Platform of Registered Systematic Review and Meta-analysis Protocols, May 2021. http://dx.doi.org/10.37766/inplasy2021.5.0110.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Wu, Xiaojin, Xiao Ma, Tiemei Song, Jie Liu, Yi Sun, and Depei Wu. Evidence Map for the Indirect Effects of CMV Infection on Patients with Allogeneic Hematopoietic Stem Cell transplantation. INPLASY - International Platform of Registered Systematic Review and Meta-analysis Protocols, November 2022. http://dx.doi.org/10.37766/inplasy2022.11.0032.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Wang, Xiaoyue, Hui Lu, Zhihao Liang, Liang Wang, and Ji Ma. Ixazomib combined with autologous stem cell transplantation for POEMS syndrome: a case report and meta‑analysis. INPLASY - International Platform of Registered Systematic Review and Meta-analysis Protocols, July 2022. http://dx.doi.org/10.37766/inplasy2022.7.0061.

Full text
Abstract:
Review question / Objective: POEMS syndrome is a rare monoclonal plasma cell proliferation disorder. At present, there is no unified treatment for POEMS syndrome. Here, we describe one case with POEMS syndrome. And we made a meta­analysis to assess the efficacy of treatment strategies in recent ten years. Search strategy: We searched relevant articles in PubMed, Embase and MEDLINE database for the period up to July 2021.The search strategy included the keywords: POEMS, Therapy, Drug Therapy, Biological Therapy, Combined Modality Therapy, Hematopoietic Stem Cell Transplantation, Immunotherapy, Molecular Targeted Therapy, Chemoradiotherapy, Salvage Therapy, Controlled Clinical Trial, Randomized Controlled Trial et al. In addition, we checked all the references of eligible articles that our search retrieved to identify potentially eligible papers.
APA, Harvard, Vancouver, ISO, and other styles
6

Ji, Conghua, Rongchen Dai, Hanting Wu, Qiushuang Li, Shan Liu, Peijie He, Juan Liang, and Qing Guo. Efficacy and safety of hematopoietic stem cell transplantation for hematologic malignancies: A protocol for an overview of systematic reviews and meta-analyses. INPLASY - International Platform of Registered Systematic Review and Meta-analysis Protocols, May 2021. http://dx.doi.org/10.37766/inplasy2021.5.0064.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Yin, Xuewei, Liming Yu, Lingling Yin, Yan Wang, Wei Zheng, Jie Xu, Yueli Liu, et al. Efficacy and safety of tandem autologous hematopoietic stem cell transplantation in the treatment of multiple myeloma: A protocol for systematic review and meta-analysis. INPLASY - International Platform of Registered Systematic Review and Meta-analysis Protocols, February 2022. http://dx.doi.org/10.37766/inplasy2022.2.0007.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Wang, Yan, Zhenzhen Wang, Jie Xu, Yueli Liu, Kui Liu, Xuewei Yin, Xinyu Tang, and Siyuan Cui. Efficacy and safety of haploidentical hematopoietic stem cell transplantation on severe aplastic anemia using busulfan-based myeloablative regimenA protocol for a Bayesian network meta-analysis. INPLASY - International Platform of Registered Systematic Review and Meta-analysis Protocols, January 2022. http://dx.doi.org/10.37766/inplasy2022.1.0116.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Yin, Xuewei, Liming Yu, Lingling Yin, Yan Wang, Wei Zheng, Jie Xu, Yueli Liu, et al. Efficacy and safety of tandem versus single autologous hematopoietic stem cell transplantation for the treatment of multiple myeloma: A protocol for systematic review and meta-analysis. INPLASY - International Platform of Registered Systematic Review and Meta-analysis Protocols, June 2021. http://dx.doi.org/10.37766/inplasy2021.6.0112.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Kungwankiattichai, Smith, Ben Ponvilawa, Claudie Roy, Pattaraporn Tunsing, Florian Kuchenbauer, and Weerapat Owattanapanich. Maintenance with Hypomethylating Agents after Allogeneic Stem Cell Transplantation in Acute Myeloid Leukemia and Myelodysplastic Syndrome: A Systematic Review and Meta-Analysis. INPLASY - International Platform of Registered Systematic Review and Meta-analysis Protocols, November 2021. http://dx.doi.org/10.37766/inplasy2021.11.0078.

Full text
Abstract:
Review question / Objective: P: Patients with AML or MDS after allo-SCT; I: Hypomethylating agents after allo-SCT; C: Observation after allo-SCT; O: Overall survival rates. Condition being studied: Hypomethylating agents (HMAs) seem to have a range of properties favorable to post-allogeneic hematopoietic stem cell transplantation (allo-SCT) maintenance in acute myeloid leukemia (AML) patients. This meta-analysis was performed to review all relevant studies to compare the outcomes of patients undergoing allo-SCT for AML or MDS receiving HMA maintenance therapy with observation only. Information sources: The systematic search of the Embase and MEDLINE databases identified 4,416 articles, from which 512 duplicates were removed. This resulted in 3,904 articles available for title and abstract review. Subsequently, 3,875 articles were excluded as the article type and study design did not fulfill the inclusion criteria, or there was no report on a primary outcome of interest. The remaining 29 articles underwent full-length review and 18 of those were excluded for the aforementioned reasons. Ultimately, the eligibility criteria for our meta-analysis were met by 11 studies: 2 RCTs, 1 prospective cohort study, and 8 retrospective cohort studies.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography