Stromal Microenvironment of Human Fetal Hematopoiesis: In vitro Morphologic Studies

Pathobiology ◽  
1994 ◽  
Vol 62 (2) ◽  
pp. 99-103 ◽  
Author(s):  
Colin A.I. Bethel ◽  
Terri Steinkirchner ◽  
Esmail D. Zanjani ◽  
Alan W. Flake
2019 ◽  
Vol 20 (10) ◽  
pp. 2496 ◽  
Author(s):  
Barbara Fazi ◽  
Carla Proserpio ◽  
Silvia Galardi ◽  
Francesca Annesi ◽  
Mattia Cola ◽  
...  

Glioblastoma (GBM) is a primary brain tumor whose prognosis is inevitably dismal, leading patients to death in about 15 months from diagnosis. Tumor cells in the mass of the neoplasm are in continuous exchange with cells of the stromal microenvironment, through the production of soluble molecules, among which chemokines play prominent roles. CXCL14 is a chemokine with a pro-tumor role in breast and prostate carcinoma, where it is secreted by cancer associated fibroblasts, and contributes to tumor growth and invasion. We previously observed that CXCL14 expression is higher in GBM tissues than in healthy white matter. Here, we study the effects of exogenously supplemented CXCL14 on key tumorigenic properties of human GBM cell lines. We show that CXCL14 enhances the migration ability and the proliferation of U87MG and LN229 GBM cell lines. None of these effects was affected by the use of AMD3100, an inhibitor of CXCR4 receptor, suggesting that the observed CXCL14 effects are not mediated by this receptor. We also provide evidence that CXCL14 enhances the sphere-forming ability of glioblastoma stem cells, considered the initiating cells, and is responsible for tumor onset, growth and recurrence. In support of our in vitro results, we present data from several GBM expression datasets, demonstrating that CXCL14 expression is inversely correlated with overall survival, that it is enriched at the leading edge of the tumors and in infiltrating tumor areas, and it characterizes mesenchymal and NON G-CIMP tumors, known to have a particularly bad prognosis. Overall, our results point to CXCL14 as a protumorigenic chemokine in GBM.


2020 ◽  
Vol 21 (14) ◽  
pp. 4993 ◽  
Author(s):  
Raphael Mohr ◽  
Burcin Özdirik ◽  
Jana Knorr ◽  
Alexander Wree ◽  
Münevver Demir ◽  
...  

Cholangiocarcinoma (CCA) comprises a heterogeneous group of primary liver tumors. They emerge from different hepatic (progenitor) cell populations, typically via sporadic mutations. Chronic biliary inflammation, as seen in primary sclerosing cholangitis (PSC), may trigger CCA development. Although several efforts were made in the last decade to better understand the complex processes of biliary carcinogenesis, it was only recently that new therapeutic advances have been achieved. Animal models are a crucial bridge between in vitro findings on molecular or genetic alterations, pathophysiological understanding, and new therapeutic strategies for the clinic. Nevertheless, it is inherently difficult to recapitulate simultaneously the stromal microenvironment (e.g., immune-competent cells, cholestasis, inflammation, PSC-like changes, fibrosis) and the tumor biology (e.g., mutational burden, local growth, and metastatic spread) in an animal model, so that it would reflect the full clinical reality of CCA. In this review, we highlight available data on animal models for CCA. We discuss if and how these models reflect human disease and whether they can serve as a tool for understanding the pathogenesis, or for predicting a treatment response in patients. In addition, open issues for future developments will be discussed.


2020 ◽  
Vol 21 (12) ◽  
pp. 4199
Author(s):  
Metka Novak ◽  
Miha Koprivnikar Krajnc ◽  
Barbara Hrastar ◽  
Barbara Breznik ◽  
Bernarda Majc ◽  
...  

The chemokine CCL5/RANTES is a versatile inflammatory mediator, which interacts with the receptor CCR5, promoting cancer cell interactions within the tumor microenvironment. Glioblastoma is a highly invasive tumor, in which CCL5 expression correlates with shorter patient survival. Using immunohistochemistry, we identified CCL5 and CCR5 in a series of glioblastoma samples and cells, including glioblastoma stem cells. CCL5 and CCR5 gene expression were significantly higher in a cohort of 38 glioblastoma samples, compared to low-grade glioma and non-cancerous tissues. The in vitro invasion of patients-derived primary glioblastoma cells and glioblastoma stem cells was dependent on CCL5-induced CCR5 signaling and is strongly inhibited by the small molecule CCR5 antagonist maraviroc. Invasion of these cells, which was enhanced when co-cultured with mesenchymal stem cells (MSCs), was inhibited by maraviroc, suggesting that MSCs release CCR5 ligands. In support of this model, we detected CCL5 and CCR5 in MSC monocultures and glioblastoma-associated MSC in tissue sections. We also found CCR5 expressing macrophages were in close proximity to glioblastoma cells. In conclusion, autocrine and paracrine cross-talk in glioblastoma and, in particular, glioblastoma stem cells with its stromal microenvironment, involves CCR5 and CCL5, contributing to glioblastoma invasion, suggesting the CCL5/CCR5 axis as a potential therapeutic target that can be targeted with repositioned drug maraviroc.


2021 ◽  
Vol 14 (3) ◽  
pp. 448-455
Author(s):  
Xian-Ning Liu ◽  
◽  
Yun Chen ◽  
Yao Wang ◽  
◽  
...  

Corneal stroma-derived mesenchymal stem cells (CS-MSCs) are mainly distributed in the anterior part of the corneal stroma near the corneal limbal stem cells (LSCs). CS-MSCs are stem cells with self-renewal and multidirectional differentiation potential. A large amount of data confirmed that CS-MSCs can be induced to differentiate into functional keratocytes in vitro, which is the motive force for maintaining corneal transparency and producing a normal corneal stroma. CS-MSCs are also an important component of the limbal microenvironment. Furthermore, they are of great significance in the reconstruction of ocular surface tissue and tissue engineering for active biocornea construction. In this paper, the localization and biological characteristics of CS-MSCs, the use of CS-MSCs to reconstruct a tissue-engineered active biocornea, and the repair of the limbal and matrix microenvironment by CS-MSCs are reviewed, and their application prospects are discussed.


Blood ◽  
1989 ◽  
Vol 74 (1) ◽  
pp. 139-144
Author(s):  
NG Abraham ◽  
D Bucher ◽  
U Niranjan ◽  
AC Brown ◽  
JD Lutton ◽  
...  

Azidothymidine (AZT) is a useful drug in management of AIDS. Nevertheless, its hematologic toxicity such as anemia and neutropenia present further complications to an already compromised hematopoietic state in patients. We studied the effects of AZT on human and murine bone marrow (BM) colony growth as determined by assays of CFU-E, BFU-E, CFU-GM, and fibroblastoid stromal (CFU-Fb) colonies. Cultures were grown in methylcellulose with growth factors and scored after three- to 14-day incubation. In general, murine marrow cultures were more sensitive to AZT as compared with human marrow. Furthermore, interindividual variation in toxicity to AZT was observed between marrow samples; 1 mumol/L AZT inhibited murine CFU-E, BFU-E, and CFU-GM by 98% to 100%, whereas human marrow was inhibited by 52%, 87%, and 65%, respectively. Lower concentrations of AZT (0.1 mumol/L) inhibited murine erythroid colony growth by 85% to 90%, whereas human growth was inhibited by only 39% to 52%. Myeloid colony inhibition was similar for human and murine systems. CFU-Fb growth was markedly suppressed (75%) by 1 mumol/L AZT. Hemin, at a concentration of 10 mumol/L, overcame some of the inhibitory effects of 1 to 0.1 mumol/L AZT without hindering antiviral activity. Inhibition of human CFU-E growth was completely overcome with hemin, whereas CFU-GM growth was recovered to 66% to 74% of control. A similar but less pronounced effect was observed for BFU-E. Furthermore, hemin does not decrease AZT's effects of HIV antigen content in vitro. We conclude that anemia and neutropenia, occurring as a result of AZT, may not be as pronounced in the presence of hemin. Furthermore, CFU-Fb was significantly reduced in the presence of low concentrations of AZT. This may indicate a major target site for BM toxicity since the stromal microenvironment may be responsible for maintaining short- and long-term hematopoiesis.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3602-3602
Author(s):  
Surya S Kotha ◽  
Kiet T Phong ◽  
Amie Adams ◽  
Brian Hayes ◽  
Meredith A Roberts ◽  
...  

Abstract Hematopoietic cells dynamically interact with their surrounding microenvironment during their residence, maturation and differentiation. Individual marrow components have been isolated and studied in 2D in vitro cultures, yet their functional contributions to a complete niche are not fully understood. In vivo studies in mouse models are complex, and the inaccessibility of marrow architecture has precluded systematic analysis of each component. Here, we employ an in vitro 3D microfluidic vascular system to study the effect of microenvironmental cues on cell trafficking in an engineered hematopoietic niche. Our system allows for control of 3D geometric cues, hydrodynamic flow, multi-cellular compositions, and cellular matrix remodeling by combining soft lithography and injection modeling in type I collagen gel (Zheng et al. PNAS 2012). Endothelial cells perfused through the embedded microfluidic network form a confluent, patent endothelium within the collagen. Incorporating hematopoietic cells and stromal cells into the extravascular collagen space allows us to visualize how cells interact with vasculature during culture. First, we developed a marrow stromal microenvironment to understand how stromal cells modify the marrow microvascular environment, by incorporating two different human marrow-derived stromal fibroblast cell lines (HS27a and HS5) in the extravascular space surrounding the vessels (Fig 1A). HS27a and HS5 cells created distinct vascular microenvironments by secreting divergent cytokine profiles: stem cell niche-associated and inflammatory cytokines, respectively. Both stromal lines modified the vascular phenotype by reducing endothelial expression of vWF and junctional proteins. In particular, co-culture with HS5 increased expression of inflammatory markers on the endothelium. Next, we evaluated the function of this modified vasculature on hematopoietic stem cell trafficking through the vessels. When CD34+ cells alone were perfused through the microvessels, they adhered on the vessel wall and migrated into the matrix. The HS27a and HS5-induced microenvironment did not significantly change trafficking behavior (Fig 1B, C). When monocytes were perfused through the microvessels, they also adhered on the vessel wall and migrated into the matrix. Monocytes are a critical part of the marrow space, and are known to modify the endothelium and stromal microenvironment. When CD34+ cells were perfused through the vessels 24 hours after monocytes, crosstalk shifted trafficking patterns of both cell types and led to increased adhesion and migration within the HS5- and HS27a-modified vessels (Fig 1D). To explore the competent marrow niche in vitro, we further examined hematopoietic cell trafficking from the extravascular space into the circulation by embedding cells from fresh human bone marrow screens within the collagen matrix. We found this cell fraction contained a mixed population of hematopoietic and stromal cells, and could be cultured in the microvascular niche for at least two weeks. Scanning electron microscopy showed various types of marrow cells residing in both the abluminal and luminal side of the endothelium (Fig 1E-G). Throughout culture with continuous vascular perfusion, we collected the media flow-through and identified different hematopoietic cell populations released from the matrix into the circulation over the course of two weeks. Specifically, we identified CD34+ hematopoietic progenitor cells along with megakaryocyte, erythroid, lymphoid, and myeloid lineage cells by flow cytometry analysis. In summary, we developed an in vitro 3D microvascular marrow niche and gained insight into hematopoietic cell trafficking between the stroma and the circulation. By guiding the interplay of heterogeneous cell populations, we have demonstrated the capacity to define distinct microenvironment spaces. This platform shows promise for long term culture of a whole marrow population and for the ex vivo generation of hematopoietic cells. Further development of this 3D marrow niche will allow us to better understand the complexities mediating stem cell trafficking, residence, proliferation, mobilization, and differentiation in both health and disease. Disclosures No relevant conflicts of inteqerest to declare.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. 7026-7026 ◽  
Author(s):  
Michael Richard Grunwald ◽  
Mark J. Levis

7026 Background: FLT3 ligand (FL) is a hematopoietic growth factor expressed in many tissues. AML patients who are administered myeloablative therapy exhibit a marked and transient rise in plasma FL concentrations. Furthermore, the presence of high concentrations (1000 pg/mL) of FL impedes the efficacy of FLT3 tyrosine kinase inhibitors in vitro. However, the behavior of FL concentrations throughout the course of AML treatment remains unknown. This pilot study was undertaken to track the relationship between AML therapy and FL levels over time. Methods: Ten AML patients were enrolled in an IRB-approved procurement protocol. Blood samples were collected at weekly intervals for one year, and plasma was isolated by centrifugation. Plasma FL and stem cell factor (SCF) concentrations were measured by ELISA. Results: We observed four distinct patterns in FL fluctuations. First, in all cases where induction or consolidation chemotherapy resulted in an aplastic bone marrow (nine patients), FL concentrations rose markedly and consistently to levels >1000 pg/mL following the administration of chemotherapy. Second, in three of four patients whose leukemia was refractory to induction chemotherapy, FL concentrations remained below 500 pg/mL during induction. Third, in two patients receiving the FLT3 TKI sorafenib, FL concentrations did not rise above 500 pg/mL while on this medication. Fourth, in one patient receiving the hypomethylating agent 5-azacitidine, FL concentrations remained below 100 pg/mL throughout the course of therapy. SCF concentrations did not vary throughout the course of chemotherapy. Conclusions: An “FL surge” was seen when cytotoxic chemotherapy resulted in aplasia. This FL surge was not seen with sorafenib or 5-azacitidine. In addition, the FL surge was attenuated in three patients whose leukemia was refractory to chemotherapy. These observations give rise to two new hypotheses regarding FL: 1) It is possible to maintain lower FL levels with targeted agents than with chemotherapy; and 2) Residual leukemia appears to inhibit the FL surge, providing indirect evidence of cross-talk between leukemia and the stromal microenvironment. This inhibition may be the explanation for why AML patients develop pancytopenia early in relapse.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 324-324 ◽  
Author(s):  
Nunzia Montuori ◽  
Patrizia Ricci ◽  
Bianca Serio ◽  
Valeria Visconte ◽  
Claudio La Penna ◽  
...  

Abstract The urokinase-type plasminogen activator receptor (uPAR) is a cell-surface receptor involved in cell adhesion and migration. uPAR binds urokinase (uPA) and vitronectin (VN) and interacts with integrins and chemotaxis receptors. Soluble forms of uPAR (suPAR) have been detected in human plasma and urine. A cleaved form of suPAR (c-suPAR), lacking the N-terminal domain and exposing the sequence SRSRY (aa 88–92), stimulates cell migration by activating fMLP receptors. We recently demonstrated uPAR involvement in G-CSF-induced CD34+ hematopoietic stem cell (HSC) mobilization. We also demonstrated that c-suPAR could induce mobilization of hematopoietic stem/progenitor cells in mice. Since HSC mobilization and homing to bone marrow (BM) are mirror image processes which utilize the same mediators and similar signaling pathways, we investigated whether uPAR and its ligands could play a role in regulating CD34+ HSC interactions with the BM stroma, thus also contributing to HSC homing and engraftment to the BM. We found expression of uPA and VN in cultures of human BM stroma cells. Interestingly, stroma cells also produced suPAR and high amounts of c-suPAR, exposing the chemotactic SRSRY sequence. The role of the different soluble forms of uPAR produced by stroma cells in regulating HSC interactions with the BM microenvironment was analyzed by long term cultures (LTC) of BM and G-CSF mobilized CD34+ HSCs, in the presence of suPAR or the uPAR-derived uPAR84–95 peptide, corresponding to the active site of c-suPAR. Both suPAR and the uPAR84–95 peptide increased the number of adherent and released clonogenic progenitors from LTC of BM and G-CSF mobilized HSCs. To elucidate the mechanism of suPAR and c-suPAR effects on CD34+ HSC interactions with the stromal microenvironment, in vitro adhesion and proliferation assays were performed on CD34+ KG1 cells. suPAR treatment determined a significant increase in CD34+ KG1 cell adhesion whereas c-suPAR increased cell proliferation. Taken together, our results indicate that BM stroma produces soluble forms of uPAR that regulate CD34+ HSC interactions with BM microenvironment, their local proliferation and trafficking from and to BM.


Blood ◽  
2007 ◽  
Vol 109 (10) ◽  
pp. 4237-4244 ◽  
Author(s):  
Tarvinder S. Dhanjal ◽  
Caroline Pendaries ◽  
Ewan A. Ross ◽  
Mark K. Larson ◽  
Majd B. Protty ◽  
...  

Abstract During thrombopoiesis, maturing megakaryocytes (MKs) migrate within the complex bone marrow stromal microenvironment from the proliferative osteoblastic niche to the capillary-rich vascular niche where proplatelet formation and platelet release occurs. This physiologic process involves proliferation, differentiation, migration, and maturation of MKs before platelet production occurs. In this study, we report a role for the glycoprotein PECAM-1 in thrombopoiesis. We show that following induced thrombocytopenia, recovery of the peripheral platelet count is impaired in PECAM-1–deficient mice. Whereas MK maturation, proplatelet formation, and platelet production under in vitro conditions were unaffected, we identified a migration defect in PECAM-1–deficient MKs in response to a gradient of stromal cell–derived factor 1 (SDF1), a major chemokine regulating MK migration within the bone marrow. This defect could be explained by defective PECAM-1−/− MK polarization of the SDF1 receptor CXCR4 and an increase in adhesion to immobilized bone marrow matrix proteins that can be explained by an increase in integrin activation. The defect of migration and polarization was confirmed in vivo with demonstration of altered spatial localization of MKs within the bone marrow in PECAM-1–deficient mice, following immune-induced thrombocytopenia. This study identifies a novel role for PECAM-1 in regulating MK migration and thrombopoiesis.


2012 ◽  
Vol 2012 ◽  
pp. 1-13 ◽  
Author(s):  
Guang-Jer Wu

METCAM, an integral membrane cell adhesion molecule (CAM) in theIg-like gene superfamily, is capable of performing typical functions ofCAMs, such as mediating cell-cell and cell-extracellular interactions, crosstalk with intracellular signaling pathways, and modulating social behaviors of cells.METCAMis expressed in about nine normal cells/tissues. Aberrant expression ofMETCAMhas been associated with the progression of several epithelial tumors. Furtherin vitroandin vivostudies show thatMETCAMplays a dual role in the progression of different tumors. It can promote the malignant progression of several tumors. On the other hand, it can suppress the malignant progression of other tumors. We suggest that the role ofMETCAMin the progression of different cancer types may be modulated by different intrinsic factors present in different cancer cells and also in different stromal microenvironment. Many possible mechanisms mediated by thisCAMduring early tumor development and metastasis are suggested.


Sign in / Sign up

Export Citation Format

Share Document