501
|
Song JW, Cavnar SP, Walker AC, Luker KE, Gupta M, Tung YC, Luker GD, Takayama S. Microfluidic endothelium for studying the intravascular adhesion of metastatic breast cancer cells. PLoS One 2009; 4:e5756. [PMID: 19484126 DOI: 10.1371/journal.pone.0005756.g001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 05/05/2009] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND The ability to properly model intravascular steps in metastasis is essential in identifying key physical, cellular, and molecular determinants that can be targeted therapeutically to prevent metastatic disease. Research on the vascular microenvironment has been hindered by challenges in studying this compartment in metastasis under conditions that reproduce in vivo physiology while allowing facile experimental manipulation. METHODOLOGY/PRINCIPAL FINDINGS We present a microfluidic vasculature system to model interactions between circulating breast cancer cells with microvascular endothelium at potential sites of metastasis. The microfluidic vasculature produces spatially-restricted stimulation from the basal side of the endothelium that models both organ-specific localization and polarization of chemokines and many other signaling molecules under variable flow conditions. We used this microfluidic system to produce site-specific stimulation of microvascular endothelium with CXCL12, a chemokine strongly implicated in metastasis. CONCLUSIONS/SIGNIFICANCE When added from the basal side, CXCL12 acts through receptor CXCR4 on endothelium to promote adhesion of circulating breast cancer cells, independent of CXCL12 receptors CXCR4 or CXCR7 on tumor cells. These studies suggest that targeting CXCL12-CXCR4 signaling in endothelium may limit metastases in breast and other cancers and highlight the unique capabilities of our microfluidic device to advance studies of the intravascular microenvironment in metastasis.
Collapse
Affiliation(s)
- Jonathan W Song
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | |
Collapse
|
502
|
Song JW, Cavnar SP, Walker AC, Luker KE, Gupta M, Tung YC, Luker GD, Takayama S. Microfluidic endothelium for studying the intravascular adhesion of metastatic breast cancer cells. PLoS One 2009; 4:e5756. [PMID: 19484126 PMCID: PMC2684591 DOI: 10.1371/journal.pone.0005756] [Citation(s) in RCA: 238] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 05/05/2009] [Indexed: 11/25/2022] Open
Abstract
Background The ability to properly model intravascular steps in metastasis is essential in identifying key physical, cellular, and molecular determinants that can be targeted therapeutically to prevent metastatic disease. Research on the vascular microenvironment has been hindered by challenges in studying this compartment in metastasis under conditions that reproduce in vivo physiology while allowing facile experimental manipulation. Methodology/Principal Findings We present a microfluidic vasculature system to model interactions between circulating breast cancer cells with microvascular endothelium at potential sites of metastasis. The microfluidic vasculature produces spatially-restricted stimulation from the basal side of the endothelium that models both organ-specific localization and polarization of chemokines and many other signaling molecules under variable flow conditions. We used this microfluidic system to produce site-specific stimulation of microvascular endothelium with CXCL12, a chemokine strongly implicated in metastasis. Conclusions/Significance When added from the basal side, CXCL12 acts through receptor CXCR4 on endothelium to promote adhesion of circulating breast cancer cells, independent of CXCL12 receptors CXCR4 or CXCR7 on tumor cells. These studies suggest that targeting CXCL12-CXCR4 signaling in endothelium may limit metastases in breast and other cancers and highlight the unique capabilities of our microfluidic device to advance studies of the intravascular microenvironment in metastasis.
Collapse
Affiliation(s)
- Jonathan W. Song
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Stephen P. Cavnar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ann C. Walker
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kathryn E. Luker
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Mudit Gupta
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yi-Chung Tung
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Gary D. Luker
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (GDL); (ST)
| | - Shuichi Takayama
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (GDL); (ST)
| |
Collapse
|
503
|
RhoA and Rac1 GTPases play major and differential roles in stromal cell-derived factor-1-induced cell adhesion and chemotaxis in multiple myeloma. Blood 2009; 114:619-29. [PMID: 19443661 DOI: 10.1182/blood-2009-01-199281] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The interaction of multiple myeloma (MM) cells with the bone marrow (BM) milieu plays a crucial role in MM pathogenesis. Stromal cell-derived factor-1 (SDF1) regulates homing of MM cells to the BM. In this study, we examined the role of RhoA and Rac1 GTPases in SDF1-induced adhesion and chemotaxis of MM. We found that both RhoA and Rac1 play key roles in SDF1-induced adhesion of MM cells to BM stromal cells, whereas RhoA was involved in chemotaxis and motility. Furthermore, both ROCK and Rac1 inhibitors reduced SDF1-induced polymerization of actin and activation of LIMK, SRC, FAK, and cofilin. Moreover, RhoA and Rac1 reduced homing of MM cells to BM niches. In conclusion, we characterized the role of RhoA and Rac1 GTPases in SDF1-induced adhesion, chemotaxis, and homing of MM cells to the BM, providing the framework for targeting RhoA and Rac1 GTPases as novel MM therapy.
Collapse
|
504
|
Abstract
In this issue of Cell Stem Cell, Hooper et al. (2009) use a combination of immunohistochemistry and flow cytometry to characterize the bone marrow vasculature both before and after injury. The authors demonstrate that recovery of normal hematopoiesis after myelosuppressive insult is dependent upon endothelial VEGFR2.
Collapse
Affiliation(s)
- Rebecca L Porter
- Wilmot Cancer Center and Endocrine Division, University of Rochester School of Medicine, 601 Elmwood Avenue, Box 693, Rochester, NY 14642, USA
| | | |
Collapse
|
505
|
Hooper AT, Butler JM, Nolan DJ, Kranz A, Iida K, Kobayashi M, Kopp HG, Shido K, Petit I, Yanger K, James D, Witte L, Zhu Z, Wu Y, Pytowski B, Rosenwaks Z, Mittal V, Sato TN, Rafii S. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell 2009; 4:263-74. [PMID: 19265665 DOI: 10.1016/j.stem.2009.01.006] [Citation(s) in RCA: 481] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Revised: 05/06/2008] [Accepted: 01/09/2009] [Indexed: 01/12/2023]
Abstract
Myelosuppression damages the bone marrow (BM) vascular niche, but it is unclear how regeneration of bone marrow vessels contributes to engraftment of transplanted hematopoietic stem and progenitor cells (HSPCs) and restoration of hematopoiesis. We found that chemotherapy and sublethal irradiation induced minor regression of BM sinusoidal endothelial cells (SECs), while lethal irradiation induced severe regression of SECs and required BM transplantation (BMT) for regeneration. Within the BM, VEGFR2 expression specifically demarcated a continuous network of arterioles and SECs, with arterioles uniquely expressing Sca1 and SECs uniquely expressing VEGFR3. Conditional deletion of VEGFR2 in adult mice blocked regeneration of SECs in sublethally irradiated animals and prevented hematopoietic reconstitution. Similarly, inhibition of VEGFR2 signaling in lethally irradiated wild-type mice rescued with BMT severely impaired SEC reconstruction and prevented engraftment and reconstitution of HSPCs. Therefore, regeneration of SECs via VEGFR2 signaling is essential for engraftment of HSPCs and restoration of hematopoiesis.
Collapse
Affiliation(s)
- Andrea T Hooper
- Howard Hughes Medical Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
506
|
Juarez JG, Thien M, Dela Pena A, Baraz R, Bradstock KF, Bendall LJ. CXCR4 mediates the homing of B cell progenitor acute lymphoblastic leukaemia cells to the bone marrow via activation of p38MAPK. Br J Haematol 2009; 145:491-9. [DOI: 10.1111/j.1365-2141.2009.07648.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
507
|
Roccaro AM, Sacco A, Chen C, Runnels J, Leleu X, Azab F, Azab AK, Jia X, Ngo HT, Melhem MR, Burwick N, Varticovski L, Novina CD, Rollins BJ, Anderson KC, Ghobrial IM. microRNA expression in the biology, prognosis, and therapy of Waldenström macroglobulinemia. Blood 2009; 113:4391-402. [PMID: 19074725 PMCID: PMC2943754 DOI: 10.1182/blood-2008-09-178228] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Accepted: 11/15/2008] [Indexed: 02/07/2023] Open
Abstract
Multilevel genetic characterization of Waldenström macroglobulinemia (WM) is required to improve our understanding of the underlying molecular changes that lead to the initiation and progression of this disease. We performed microRNA-expression profiling of bone marrow-derived CD19(+) WM cells, compared with their normal cellular counterparts and validated data by quantitative reverse-transcription-polymerase chain reaction (qRT-PCR). We identified a WM-specific microRNA signature characterized by increased expression of microRNA-363*/-206/-494/-155/-184/-542-3p, and decreased expression of microRNA-9* (ANOVA; P < .01). We found that microRNA-155 regulates proliferation and growth of WM cells in vitro and in vivo, by inhibiting MAPK/ERK, PI3/AKT, and NF-kappaB pathways. Potential microRNA-155 target genes were identified using gene-expression profiling and included genes involved in cell-cycle progression, adhesion, and migration. Importantly, increased expression of the 6 miRNAs significantly correlated with a poorer outcome predicted by the International Prognostic Staging System for WM. We further demonstrated that therapeutic agents commonly used in WM alter the levels of the major miRNAs identified, by inducing downmodulation of 5 increased miRNAs and up-modulation of patient-downexpressed miRNA-9*. These data indicate that microRNAs play a pivotal role in the biology of WM; represent important prognostic marker; and provide the basis for the development of new microRNA-based targeted therapies in WM.
Collapse
Affiliation(s)
- Aldo M Roccaro
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
508
|
Abstract
The genetic events that contribute to the pathogenesis of acute myeloid leukemia are among the best characterized of all human malignancies. However, with notable exceptions such as acute promyelocytic leukemia, significant improvements in outcome based on these insights have not been forthcoming. Acute myeloid leukemia is a paradigm of cancer stem (or leukemia initiating) cells with hierarchy analogous to that seen in hematopoiesis. Normal hematopoiesis requires complex bidirectional interactions between the bone marrow microenvironment (or niche) and hematopoietic stem cells (HSCs). These interactions are critical for the maintenance of normal HSC quiescence and perturbations can influence HSC self-renewal. Leukemia stem cells (LSCs), which also possess limitless self-renewal, may hijack these homeostatic mechanisms, take refuge within the sanctuary of the niche during chemotherapy, and consequently contribute to eventual disease relapse. We will discuss the emerging evidence supporting the importance of the bone marrow microenvironment in LSC survival and consider the physiologic interactions of HSCs and the niche that inform our understanding of microenvironment support of LSCs. Finally, we will discuss approaches for the rational development of therapies that target the microenvironment.
Collapse
|
509
|
Abstract
Detailed genomic studies have shown that cytogenetic abnormalities contribute to multiple myeloma (MM) pathogenesis and disease progression. Nevertheless, little is known about the characteristics of MM at the epigenetic level and specifically how microRNAs regulate MM progression in the context of the bone marrow milieu. Therefore, we performed microRNA expression profiling of bone marrow derived CD138(+) MM cells versus their normal cellular counterparts and validated data by qRT-PCR. We identified a MM-specific microRNA signature characterized by down-expression of microRNA-15a/-16 and overexpression of microRNA-222/-221/-382/-181a/-181b (P < .01). We investigated the functional role of microRNA-15a and -16 and showed that they regulate proliferation and growth of MM cells in vitro and in vivo by inhibiting AKT serine/threonine-protein-kinase (AKT3), ribosomal-protein-S6, MAP-kinases, and NF-kappaB-activator MAP3KIP3. Moreover, miRNA-15a and -16 exerted their anti-MM activity even in the context of the bone marrow milieu in vitro and in vivo. These data indicate that microRNAs play a pivotal role in the biology of MM and represent important targets for novel therapies in MM.
Collapse
|
510
|
Altered cellular dynamics and endosteal location of aged early hematopoietic progenitor cells revealed by time-lapse intravital imaging in long bones. Blood 2009; 114:290-8. [PMID: 19357397 DOI: 10.1182/blood-2008-12-195644] [Citation(s) in RCA: 179] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aged hematopoietic stem cells (HSCs) are impaired in supporting hematopoiesis. The molecular and cellular mechanisms of stem cell aging are not well defined. HSCs interact with nonhematopoietic stroma cells in the bone marrow forming the niche. Interactions of hematopoietic cells with the stroma/microenvironment inside bone cavities are central to hematopoiesis as they regulate cell proliferation, self-renewal, and differentiation. We recently hypothesized that one underlying cause of altered hematopoiesis in aging might be due to altered interactions of aged stem cells with the microenvironment/niche. We developed time-lapse 2-photon microscopy and novel image analysis algorithms to quantify the dynamics of young and aged hematopoietic cells inside the marrow of long bones of mice in vivo. We report in this study that aged early hematopoietic progenitor cells (eHPCs) present with increased cell protrusion movement in vivo and localize more distantly to the endosteum compared with young eHPCs. This correlated with reduced adhesion to stroma cells as well as reduced cell polarity upon adhesion of aged eHPCs. These data support a role of altered eHPC dynamics and altered cell polarity, and thus altered niche biology in mechanisms of mammalian aging.
Collapse
|
511
|
Burger JA, Stewart DJ. CXCR4 chemokine receptor antagonists: perspectives in SCLC. Expert Opin Investig Drugs 2009; 18:481-90. [PMID: 19335276 DOI: 10.1517/13543780902804249] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Small-cell lung cancer (SCLC) is a particularly aggressive form of lung cancer characterized by early and widespread metastases and the ability to rapidly develop resistance against chemotherapeutic agents. Tumor cell migration and metastasis share many similarities with leukocyte trafficking, which is critically regulated by chemokine receptors and adhesion molecules. SCLC cells express high levels of CXCR4 (CD184), a seven-transmembrane G-protein-coupled chemokine receptor. Stromal cells within the bone marrow microenvironment and at extramedullary sites constitutively secrete stromal cell-derived factor-1 (CXCL12), the ligand for CXCR4. Activation of CXCR4 induces SCLC cell migration and adhesion to stromal cells that secrete CXCL12, which in turn provides growth- and drug resistance-signals to the tumor cells. CXCR4 antagonists, such as Plerixafor (AMD3100) and T140 analogues (TN14003/ BKT140), disrupt CXCR4-mediated SCLC cell-adhesion to stromal cells. In stromal cell co-cultures, CXCR4 antagonists also sensitize SCLC cells to cytotoxic drugs, such as etoposide, and thereby antagonize cell adhesion-mediated drug resistance. Therefore, targeting the CXCR4-CXCL12 axis is a novel, attractive therapeutic approach in SCLC. Here, we summarize preclinical data about CXCR4 in SCLC, and the current status of the preclinical and clinical development of CXCR4 antagonists.
Collapse
Affiliation(s)
- Jan A Burger
- University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX 77230-1402, USA.
| | | |
Collapse
|
512
|
Horner BM, Ferguson KK, Randolph MA, Spencer JA, Carlson AL, Hirsh EL, Lin CP, Butler PEM. In vivo observations of cell trafficking in allotransplanted vascularized skin flaps and conventional skin grafts. J Plast Reconstr Aesthet Surg 2009; 63:711-9. [PMID: 19324598 DOI: 10.1016/j.bjps.2009.01.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 01/08/2009] [Accepted: 01/12/2009] [Indexed: 11/28/2022]
Abstract
The problem of allogeneic skin rejection is a major limitation to more widespread application of clinical composite tissue allotransplantation (CTA). Previous research examining skin rejection has mainly studied rejection of conventional skin grafts (CSG) using standard histological techniques. The aim of this study was to objectively assess if there were differences in the immune response to CSG and primarily vascularized skin in composite tissue allotransplants (SCTT) using in vivo techniques in order to gain new insights in to the immune response to skin allotransplants. CSG and SCTT were transplanted from standard Lewis (LEW) ad Wistar Furth (WF) to recipient transgenic green fluorescent Lewis rats (LEW-GFP). In vivo confocal microscopy was used to observe cell trafficking within skin of the transplants. In addition, immunohistochemical staining was performed on skin biopsies to reveal possible expression of class II major histocompatibility antigens. A difference was observed in the immune response to SCTT compared to CSG. SCTT had a greater density cellular infiltrate than CSG (p<0.03) that was focused more at the center of the transplant (p<0.05) than at the edges, likely due to the immediate vascularization of the skin. Recipient dendritic cells were only observed in rejecting SCTT, not CSG. Furthermore, dermal endothelial class II MHC expression was only observed in allogeneic SCTT. The immune response in both SCTT and CSG was focused on targets in the dermis, with infiltrating cells clustering around hair follicles (CSG and SCTT; p<0.01) and blood vessels (SCTT; p<0.01) in allogeneic transplants. This study suggests that there are significant differences between rejection of SCTT and CSG that may limit the relevance of much of the historical data on skin graft rejection when applied to composite tissue allotransplantation. Furthermore, the use of novel in vivo techniques identified characteristics of the immune response to allograft skin not previously described, which may be useful in directing future approaches to overcoming allograft skin rejection.
Collapse
Affiliation(s)
- Benjamin M Horner
- Plastic Surgery Research Laboratory, Department of Surgery, WAC 435, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
513
|
Haematopoietic stem cells depend on Galpha(s)-mediated signalling to engraft bone marrow. Nature 2009; 459:103-7. [PMID: 19322176 PMCID: PMC2761017 DOI: 10.1038/nature07859] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 01/19/2009] [Indexed: 11/30/2022]
Abstract
Hematopoietic stem/progenitor cells (HSPC) transition in location during development1 and circulate in mammals throughout life2, moving into and out of the bloodstream to engage bone marrow (BM) niches in sequential steps of homing, engraftment and retention3–5. We show here that HSPC engraftment of BM in fetal development is dependent upon the guanine nucleotide binding protein stimulatory alpha subunit (Gsα). Adult Gsα−/− HSPCs differentiate and undergo chemotaxis, but also do not home to or engraft in the BM in adult mice and demonstrate marked inability to engage the marrow microvasculature. If deleted after engraftment, Gsα did not lead to lack of retention in the marrow, rather cytokine-induced mobilization into the blood was impaired. Testing whether activation of Gsα affects HSPC, pharmacologic activators enhanced homing and engraftment in vivo. Gsα governs specific aspects of HSPC localization under physiologic conditions in vivo and may be pharmacologically targeted to improve transplantation efficiency.
Collapse
|
514
|
Isoform-selective phosphoinositide 3'-kinase inhibitors inhibit CXCR4 signaling and overcome stromal cell-mediated drug resistance in chronic lymphocytic leukemia: a novel therapeutic approach. Blood 2009; 113:5549-57. [PMID: 19318683 DOI: 10.1182/blood-2008-06-165068] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks) are among the most frequently activated signaling pathways in cancer. In chronic lymphocytic leukemia (CLL), signals from the microenvironment are critical for expansion of the malignant B cells, and cause constitutive activation of PI3Ks. CXCR4 is a key receptor for CLL cell migration and adhesion to marrow stromal cells (MSCs). Because of the importance of CXCR4 and PI3Ks for CLL-microenvironment cross-talk, we investigated the activity of novel, isoform-selective PI3K inhibitors that target different isoforms of the p110-kDa subunit. Inhibition with p110alpha inhibitors (PIK-90 and PI-103) resulted in a significant reduction of chemotaxis and actin polymerization to CXCL12 and reduced migration beneath MSC (pseudoemperipolesis). Western blot and reverse phase protein array analyses consistently demonstrated that PIK-90 and PI-103 inhibited phosphorylation of Akt and S6, whereas p110delta or p110beta/p110delta inhibitors were less effective. In suspension and MSC cocultures, PI-103 and PIK-90 were potent inducers of CLL cell apoptosis. Moreover, these p110alpha inhibitors enhanced the cytotoxicity of fludarabine and reversed the protective effect of MSC on fludarabine-induced apoptosis. Collectively, our data demonstrate that p110alpha inhibitors antagonize stromal cell-derived migration, survival, and drug-resistance signals and therefore provide a rational to explore the therapeutic activity of these promising agents in CLL.
Collapse
|
515
|
Cannabinoid receptor 2 mediates the retention of immature B cells in bone marrow sinusoids. Nat Immunol 2009; 10:403-11. [PMID: 19252491 PMCID: PMC2768754 DOI: 10.1038/ni.1710] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Accepted: 01/26/2009] [Indexed: 01/23/2023]
Abstract
Immature B cells developing in the bone marrow are found in the parenchyma and sinusoids. The mechanisms that control the positioning of B cells in the sinusoids are not understood. Here we show that the integrin alpha(4)beta(1) (VLA-4) and its ligand VCAM-1 were required, whereas the chemokine receptor CXCR4 was dispensable, for sinusoidal retention of B cells. Instead, cannabinoid receptor 2 (CB2), a Galpha(i) protein-coupled receptor upregulated in immature B cells, was required for sinusoidal retention. Using two-photon microscopy, we found immature B cells entering and crawling in sinusoids; these immature B cells were displaced by CB2 antagonism. Moreover, CB2-deficient mice had a lower frequency of immunoglobulin lambda-chain-positive B cells in the peripheral blood and spleen. Our findings identify unique requirements for the retention of B cells in the bone marrow sinusoidal niche and suggest involvement of CB2 in the generation of the B cell repertoire.
Collapse
|
516
|
Mantle cell lymphoma cells express high levels of CXCR4, CXCR5, and VLA-4 (CD49d): importance for interactions with the stromal microenvironment and specific targeting. Blood 2009; 113:4604-13. [PMID: 19228923 DOI: 10.1182/blood-2008-10-185827] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mantle cell lymphoma (MCL) is characterized by an early, widespread dissemination and residual disease after conventional treatment, but the mechanisms responsible for lymphoma cell motility and drug resistance are largely unknown. There is growing evidence suggesting that chemokine receptors and adhesion molecules are critical for malignant B-cell trafficking and homing to supportive tissue microenvironments, where they receive survival and drug resistance signals. Therefore, we examined chemokine receptor and adhesion molecule expression and function in MCL cells and their importance for migration and adhesion to marrow stromal cells (MSCs). We found that MCL cells display high levels of functional CXCR4 and CXCR5 chemokine receptors and VLA-4 adhesion molecules. We also report that MCL cells adhere and spontaneously migrate beneath MSCs in a CXCR4- and VLA-4-dependent fashion (pseudoemperipolesis). Moreover, we demonstrate that MSCs confer drug resistance to MCL cells, particularly to MCL cells that migrate beneath MSC. To target MCL-MSC interactions, we tested Plerixafor, a CXCR4 antagonist, and natalizumab, a VLA-4 antibody. Both agents blocked functional responses to the respective ligands and inhibited adhesive interactions between MCL cells and MSCs. These findings provide a rationale to further investigate the therapeutic potential of these drugs in MCL.
Collapse
|
517
|
Stem cell regulation via dynamic interactions of the nervous and immune systems with the microenvironment. Cell Stem Cell 2009; 3:484-92. [PMID: 18983964 DOI: 10.1016/j.stem.2008.10.006] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Physiological interactions between the nervous and immune systems with components of the local microenvironment are needed to maintain homeostasis throughout the body. Dynamic regulation of bone remodeling, hematopoietic stem cells, and their evolving niches via neurotransmitter signaling are part of the host defense and repair mechanisms. This crosstalk links activated leukocytes, neuronal, and stromal cells, which combine to directly and indirectly regulate hematopoietic stem cells. Together, interactions between diverse systems create a regulatory "brain-bone-blood triad," contributing an additional dimension to the concept of the hematopoietic stem cell niche.
Collapse
|
518
|
CXCR4 inhibitor AMD3100 disrupts the interaction of multiple myeloma cells with the bone marrow microenvironment and enhances their sensitivity to therapy. Blood 2009; 113:4341-51. [PMID: 19139079 DOI: 10.1182/blood-2008-10-186668] [Citation(s) in RCA: 327] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The interaction of multiple myeloma (MM) cells with their microenvironment in the bone marrow (BM) provides a protective environment and resistance to therapeutic agents. We hypothesized that disruption of the interaction of MM cells with their BM milieu would lead to their sensitization to therapeutic agents such as bortezomib, melphalan, doxorubicin, and dexamethasone. We report that the CXCR4 inhibitor AMD3100 induces disruption of the interaction of MM cells with the BM reflected by mobilization of MM cells into the circulation in vivo, with kinetics that differed from that of hematopoietic stem cells. AMD3100 enhanced sensitivity of MM cell to multiple therapeutic agents in vitro by disrupting adhesion of MM cells to bone marrow stromal cells (BMSCs). Moreover, AMD3100 increased mobilization of MM cells to the circulation in vivo, increased the ratio of apoptotic circulating MM cells, and enhanced the tumor reduction induced by bortezomib. Mechanistically, AMD3100 significantly inhibited Akt phosphorylation and enhanced poly(ADP-ribose) polymerase (PARP) cleavage as a result of bortezomib, in the presence of BMSCs in coculture. These experiments provide a proof of concept for the use of agents that disrupt interaction with the microenvironment for enhancement of efficacy of cytotoxic agents in cancer therapy.
Collapse
|
519
|
Colmone A, Amorim M, Pontier AL, Wang S, Jablonski E, Sipkins DA. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science 2009; 322:1861-5. [PMID: 19095944 DOI: 10.1126/science.1164390] [Citation(s) in RCA: 454] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The host tissue microenvironment influences malignant cell proliferation and metastasis, but little is known about how tumor-induced changes in the microenvironment affect benign cellular ecosystems. Applying dynamic in vivo imaging to a mouse model, we show that leukemic cell growth disrupts normal hematopoietic progenitor cell (HPC) bone marrow niches and creates abnormal microenvironments that sequester transplanted human CD34+ (HPC-enriched) cells. CD34+ cells in leukemic mice declined in number over time and failed to mobilize into the peripheral circulation in response to cytokine stimulation. Neutralization of stem cell factor (SCF) secreted by leukemic cells inhibited CD34+ cell migration into malignant niches, normalized CD34+ cell numbers, and restored CD34+ cell mobilization in leukemic mice. These data suggest that the tumor microenvironment causes HPC dysfunction by usurping normal HPC niches and that therapeutic inhibition of HPC interaction with tumor niches may help maintain normal progenitor cell function in the setting of malignancy.
Collapse
Affiliation(s)
- Angela Colmone
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, 5841 South Maryland Avenue MC 2115, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
520
|
Konopleva M, Tabe Y, Zeng Z, Andreeff M. Therapeutic targeting of microenvironmental interactions in leukemia: mechanisms and approaches. Drug Resist Updat 2009; 12:103-13. [PMID: 19632887 PMCID: PMC3640296 DOI: 10.1016/j.drup.2009.06.001] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 06/29/2009] [Accepted: 06/29/2009] [Indexed: 02/03/2023]
Abstract
In hematological malignancies, there are dynamic interactions between leukemic cells and cells of the bone marrow microenvironment. Specific niches within the bone marrow microenvironment provide a sanctuary for subpopulations of leukemic cells to evade chemotherapy-induced death and allow acquisition of a drug-resistant phenotype. This review focuses on molecular and cellular biology of the normal hematopoietic stem cell and the leukemia stem cell niche, and of the molecular pathways critical for microenvironment/leukemia interactions. The key emerging therapeutic targets include chemokine receptors (CXCR4), adhesion molecules (VLA4 and CD44), and hypoxia-related proteins HIF-1alpha and VEGF. Finally, the genetic and epigenetic abnormalities of leukemia-associated stroma will be discussed. This complex interplay provides a rationale for appropriately tailored molecular therapies targeting not only leukemic cells but also their microenvironment to ensure improved outcomes in leukemia.
Collapse
Affiliation(s)
- Marina Konopleva
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030,Section of Molecular Hematology and Therapy, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030
| | - Yoko Tabe
- Department of Clinical Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Zhihong Zeng
- Section of Molecular Hematology and Therapy, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030
| | - Michael Andreeff
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030,Section of Molecular Hematology and Therapy, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030
| |
Collapse
|
521
|
Physiopathologie des métastases osseuses. ONCOLOGIE 2009. [DOI: 10.1007/s10269-008-0977-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
522
|
Abstract
Hematopoietic and epithelial cancer cells express CXCR4, a seven-transmembrane G-protein-coupled chemokine receptor. Stromal cells within the bone marrow microenvironment constitutively secrete stromal cell-derived factor-1 (SDF-1/CXCL12), the ligand for CXCR4. Activation of CXCR4 induces leukemia cell trafficking and homing to the marrow microenvironment, where CXCL12 retains leukemia cells in close contact with marrow stromal cells that provide growth and drug resistance signals. CXCR4 antagonists, such as Plerixafor (AMD3100) and T140 analogs, can disrupt adhesive tumor-stroma interactions and mobilize leukemia cells from their protective stromal microenvironment, making them more accessible to conventional drugs. Therefore, targeting the CXCR4-CXCL12 axis is a novel, attractive therapeutic approach that is explored in ongoing clinical trials in leukemia patients. Initially, CXCR4 antagonists were developed for the treatment of HIV, where CXCR4 functions as a co-receptor for virus entry into T cells. Subsequently, CXCR4 antagonists were noticed to induce leukocytosis, and are currently used clinically for mobilization of hematopoietic stem cells. However, because CXCR4 plays a key role in cross-talk between leukemia cells (and a variety of other tumor cells) and their microenvironment, cancer treatment may become the ultimate application of CXCR4 antagonists. Here, we summarize the development of CXCR4 antagonists and their preclinical and clinical activities, focusing on leukemia and other cancers.
Collapse
|
523
|
Abstract
In normal adult tissues, paracrine signals that derive from the stem cell niche, or microenvironment, play an important role in regulating the critical balance between activity and quiescence of stem cells. Similarly, evidence has emerged to support the hypothesis that signals derived from the microenvironment regulate cancer cells in an analogous manner. We recently reported that in basal cell carcinoma of the skin and in diverse other solid tumors, fibroblasts that comprise the tumor cell niche are, indeed, molecularly distinct from those that comprise the normal stroma. In particular, we found evidence suggesting that expression of secreted BMP antagonists by tumor-associated stromal cells may promote self-renewal of tumor stem cells in vivo. This chapter describes methods for identifying and evaluating the molecular signals that derive from fibroblasts in human tumors.
Collapse
|
524
|
Lewandowski D, Romeo PH. [Follow-up using optical imaging of hematopoietic reconstitution or the development of leukemia in vivo]. Ann Pathol 2008; 28 Spec No 1:S18-9. [PMID: 18984288 DOI: 10.1016/j.annpat.2008.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Daniel Lewandowski
- Laboratoire de recherche sur la réparation et la transcription dans les cellules souches (LRTS), institut de radiobiologie cellulaire et moléculaire (IRCM), CEA, DSV, 18, route du Panorama, 92265 Fontenay-aux-Roses cedex, France
| | | |
Collapse
|
525
|
Live-animal tracking of individual haematopoietic stem/progenitor cells in their niche. Nature 2008; 457:92-6. [PMID: 19052546 DOI: 10.1038/nature07434] [Citation(s) in RCA: 672] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Accepted: 09/17/2008] [Indexed: 12/15/2022]
Abstract
Stem cells reside in a specialized, regulatory environment termed the niche that dictates how they generate, maintain and repair tissues. We have previously documented that transplanted haematopoietic stem and progenitor cell populations localize to subdomains of bone-marrow microvessels where the chemokine CXCL12 is particularly abundant. Using a combination of high-resolution confocal microscopy and two-photon video imaging of individual haematopoietic cells in the calvarium bone marrow of living mice over time, we examine the relationship of haematopoietic stem and progenitor cells to blood vessels, osteoblasts and endosteal surface as they home and engraft in irradiated and c-Kit-receptor-deficient recipient mice. Osteoblasts were enmeshed in microvessels and relative positioning of stem/progenitor cells within this complex tissue was nonrandom and dynamic. Both cell autonomous and non-autonomous factors influenced primitive cell localization. Different haematopoietic cell subsets localized to distinct locations according to the stage of differentiation. When physiological challenges drove either engraftment or expansion, bone-marrow stem/progenitor cells assumed positions in close proximity to bone and osteoblasts. Our analysis permits observing in real time, at a single cell level, processes that previously have been studied only by their long-term outcome at the organismal level.
Collapse
|
526
|
|
527
|
Sweeney E, Campbell M, Watkins K, Hunter CA, Jacenko O. Altered endochondral ossification in collagen X mouse models leads to impaired immune responses. Dev Dyn 2008; 237:2693-704. [PMID: 18629872 DOI: 10.1002/dvdy.21594] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Disruption of collagen X function in hypertrophic cartilage undergoing endochondral ossification was previously linked to altered hematopoiesis in collagen X transgenic (Tg) and null (KO) mice (Jacenko et al., [2002] Am J Pathol 160:2019-2034). Mice displayed altered growth plates, diminished trabecular bone, and marrow hypoplasia with an aberrant lymphocyte profile throughout life. This study identifies altered B220+, CD4+, and CD8+ lymphocyte numbers, as well as CD4+/fox3P+ T regulatory cells in the collagen X mice. Additionally, diminished in vitro splenocyte responses to mitogens and an inability of mice to survive a challenge with Toxoplasma gondii, confirm impaired immune responses. In concert, ELISA and protein arrays identify aberrant levels of inflammatory, chemo-attractant, and matrix binding cytokines in collagen X mouse sera. These data link the disruption of collagen X function in the chondro-osseous junction to an altered hematopoietic stem cell niche in the marrow, resulting in impaired immune function.
Collapse
Affiliation(s)
- E Sweeney
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania 19104-6046, USA
| | | | | | | | | |
Collapse
|
528
|
Di Rosa F. T-lymphocyte interaction with stromal, bone and hematopoietic cells in the bone marrow. Immunol Cell Biol 2008; 87:20-9. [PMID: 19030018 DOI: 10.1038/icb.2008.84] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mature T cells in the bone marrow (BM) are in constant exchange with the blood pool. Within the BM, T-cell recognition of antigen presented by dendritic cell (DC) can occur, nevertheless it is thought that BM T cells mostly receive non-antigenic signals by either stimulatory, for example, interleukin (IL)-7, IL-15, tumor necrosis factor family members, or inhibitory molecules, for example, transforming growth factor-beta. The net balance is in favor of T-cell proliferation. Indeed, the percentage of proliferating T cells is higher in the BM than in spleen and lymph nodes, both within CD4 and CD8 T cells. High numbers of memory T cells proliferate in the BM, as they preferentially home to the BM and have an increased turnover as compared with naive T cells. I propose here that the BM plays an essential role in maintaining normal peripheral T-lymphocyte numbers and antigen-specific memory for both CD4 and CD8 T cells. I also discuss BM T-cell contribution to the homeostasis of bone metabolism as well as of hematopoiesis. It emerges that BM T cells play unexpected roles in several diseases, for example AIDS and osteoporosis. A better knowledge on BM T cells has implications for currently used clinical interventions, for example, vaccination, BM transplantation, mesenchymal stem cell-based therapies.
Collapse
Affiliation(s)
- Francesca Di Rosa
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy.
| |
Collapse
|
529
|
Favaro E, Amadori A, Indraccolo S. Cellular interactions in the vascular niche: implications in the regulation of tumor dormancy. APMIS 2008; 116:648-59. [PMID: 18834409 DOI: 10.1111/j.1600-0463.2008.01025.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Angiogenesis plays an established role in the promotion of growth of dormant micrometastasis, because blood vessels deliver oxygen and nutrients to the tumor microenvironment. In addition to this feeding function, however, there is accumulating evidence suggesting that endothelial cells-and perhaps other cellular components of the microenvironment--could communicate both positive and negative signals to tumor cells. This cross-talk between heterogeneous cell types could turn out to be important in the regulation of cancer cell behavior. Normal cells recruited during the angiogenic process, or attracted to future sites of metastasis by soluble products released by cancer cells, have been shown to create a niche favorable to tumor cell proliferation and survival. In addition, following an exogenous angiogenic spike, as may occur during inflammation, the same mechanisms could lead to re-activation of poorly angiogenic tumor cells seeded into tissues. In this review, we discuss the possible implications of this hypothesis for our understanding of the phenomenon of tumor dormancy.
Collapse
Affiliation(s)
- Elena Favaro
- Department of Oncology and Surgical Sciences, Oncology Section, University of Padova, Padova, Italy
| | | | | |
Collapse
|
530
|
Abstract
PURPOSE OF REVIEW Hematopoietic stem cells (HSCs) normally reside in the bone marrow but can be forced into the blood, a process termed mobilization used clinically to harvest large numbers of HSCs for transplantation. Currently the mobilizing agent of choice is granulocyte colony-stimulating factor; however, not all patients mobilize well. This article reviews recent advances in understanding the molecular mechanisms responsible for the retention of HSCs in the bone marrow, which are perturbed during HSC mobilization, and the clinical application of these findings. RECENT FINDINGS The interaction between the chemokine SDF-1/CXCL12 and its receptor CXCR4 is critical to retain HSCs within the bone marrow, leading to the discovery that small synthetic CXCR4 antagonists are potent mobilizing agents that synergize with granulocyte colony-stimulating factor. Separate research has shown that HSC numbers in the bone marrow can be boosted by increasing the number of osteoblasts that support HSCs. SUMMARY HSC mobilization induced by granulocyte colony-stimulating factor may be enhanced by directly targeting the chemotactic interaction between HSCs and bone marrow stroma with CXCR4 antagonists. When the primary problem is reduced, however, HSC numbers in the bone marrow, due to repeated chemotherapy/radiotherapy treatments, an alternative is to enhance HSC content by enhancing bone formation prior to mobilization.
Collapse
|
531
|
Agliano A, Martin-Padura I, Mancuso P, Marighetti P, Rabascio C, Pruneri G, Shultz LD, Bertolini F. Human acute leukemia cells injected in NOD/LtSz-scid/IL-2Rγnull mice generate a faster and more efficient disease compared to other NOD/scid-related strains. Int J Cancer 2008; 123:2222-7. [DOI: 10.1002/ijc.23772] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
532
|
Abstract
PURPOSE OF REVIEW To provide an overview of the hematopoietic stem cell (HSC) niche in the bone marrow. In addition to highlighting recent advances in the field, we will also discuss components of the niche that may contribute to the development of cancer, or cancer metastases to the bone. RECENT FINDINGS Much progress has been very recently made in the understanding of the cellular and molecular interactions in the HSC microenvironment. These recent findings point out the extraordinary complexity of the HSC microenvironment. Emerging data also suggest convergence of signals important for HSC and for leukemia or metastatic disease support. SUMMARY The HSC niche comprises complex interactions between multiple cell types and molecules requiring cell-cell signaling as well as local secretion. These components can be thought of as therapeutic targets not only for HSC expansion, but also to modify behavior of hematopoietic malignancies and cancer metastases to the bone.
Collapse
|
533
|
Lo Celso C, Klein RJ, Scadden DT. Analysis of the hematopoietic stem cell niche. ACTA ACUST UNITED AC 2008; Chapter 2:Unit 2A.5. [PMID: 18785177 DOI: 10.1002/9780470151808.sc02a05s3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Hematopoietic stem cells (HSCs) continuously replenish all blood cell lineages not only to maintain the normal rapid turnover of differentiated cells but also to respond to injury and stress. Cell-extrinsic mechanisms are critical determinants of the fine balance between HSC self-renewal and differentiation. The bone marrow microenvironment has emerged as a new area of intense study to identify which of its many components constitute the HSC niche and regulate HSC fate. While HSCs have been isolated, characterized and used in clinical practice for many years thanks to the development of very specific assays and technology (i.e., bone marrow transplants and fluorescence activated cell sorting), study of the HSC niche has evolved by combining experimental designs developed in different fields. In this unit we describe a collection of protocols spanning a wide range of techniques that can help every researcher tackling questions regarding the nature of the HSC niche.
Collapse
|
534
|
White RM, Sessa A, Burke C, Bowman T, LeBlanc J, Ceol C, Bourque C, Dovey M, Goessling W, Burns CE, Zon LI. Transparent adult zebrafish as a tool for in vivo transplantation analysis. Cell Stem Cell 2008; 2:183-9. [PMID: 18371439 DOI: 10.1016/j.stem.2007.11.002] [Citation(s) in RCA: 943] [Impact Index Per Article: 58.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Revised: 10/18/2007] [Accepted: 11/15/2007] [Indexed: 12/19/2022]
Abstract
The zebrafish is a useful model for understanding normal and cancer stem cells, but analysis has been limited to embryogenesis due to the opacity of the adult fish. To address this, we have created a transparent adult zebrafish in which we transplanted either hematopoietic stem/progenitor cells or tumor cells. In a hematopoiesis radiation recovery assay, transplantation of GFP-labeled marrow cells allowed for striking in vivo visual assessment of engraftment from 2 hr-5 weeks posttransplant. Using FACS analysis, both transparent and wild-type fish had equal engraftment, but this could only be visualized in the transparent recipient. In a tumor engraftment model, transplantation of RAS-melanoma cells allowed for visualization of tumor engraftment, proliferation, and distant metastases in as little as 5 days, which is not seen in wild-type recipients until 3 to 4 weeks. This transparent adult zebrafish serves as the ideal combination of both sensitivity and resolution for in vivo stem cell analyses.
Collapse
Affiliation(s)
- Richard Mark White
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
535
|
Getts DR, Terry RL, Getts MT, Müller M, Rana S, Shrestha B, Radford J, Van Rooijen N, Campbell IL, King NJC. Ly6c+ "inflammatory monocytes" are microglial precursors recruited in a pathogenic manner in West Nile virus encephalitis. ACTA ACUST UNITED AC 2008; 205:2319-37. [PMID: 18779347 PMCID: PMC2556789 DOI: 10.1084/jem.20080421] [Citation(s) in RCA: 263] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In a lethal West Nile virus (WNV) model, central nervous system infection triggered a threefold increase in CD45int/CD11b+/CD11c− microglia at days 6–7 postinfection (p.i.). Few microglia were proliferating, suggesting that the increased numbers were derived from a migratory precursor cell. Depletion of “circulating” (Gr1−(Ly6Clo)CX3CR1+) and “inflammatory” (Gr1hi/Ly6Chi/CCR2+) classical monocytes during infection abrogated the increase in microglia. C57BL/6 chimeras reconstituted with cFMS–enhanced green fluorescent protein (EGFP) bone marrow (BM) showed large numbers of peripherally derived (GFP+) microglia expressing GR1+(Ly6C+) at day 7 p.i., suggesting that the inflammatory monocyte is a microglial precursor. This was confirmed by adoptive transfer of labeled BM (Ly6Chi/CD115+) or circulating inflammatory monocytes that trafficked to the WNV-infected brain and expressed a microglial phenotype. CCL2 is a chemokine that is highly expressed during WNV infection and important in inflammatory monocyte trafficking. Neutralization of CCL2 not only reduced the number of GFP+ microglia in the brain during WNV infection but prolonged the life of infected animals. Therefore, CCL2-dependent inflammatory monocyte migration is critical for increases in microglia during WNV infection and may also play a pathogenic role during WNV encephalitis.
Collapse
Affiliation(s)
- Daniel R Getts
- The Discipline of Pathology, School of Medical Sciences, The University of Sydney, Sydney NSW 2006, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
536
|
Fernandez L, Rodriguez S, Huang H, Chora A, Fernandes J, Mumaw C, Cruz E, Pollok K, Cristina F, Price JE, Ferkowicz MJ, Scadden DT, Clauss M, Cardoso AA, Carlesso N. Tumor necrosis factor-alpha and endothelial cells modulate Notch signaling in the bone marrow microenvironment during inflammation. Exp Hematol 2008; 36:545-558. [PMID: 18439488 DOI: 10.1016/j.exphem.2007.12.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 11/28/2007] [Accepted: 12/24/2007] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Homeostasis of the hematopoietic compartment is challenged and maintained during conditions of stress by mechanisms that are poorly defined. To understand how the bone marrow (BM) microenvironment influences hematopoiesis, we explored the role of Notch signaling and BM endothelial cells in providing microenvironmental cues to hematopoietic cells in the presence of inflammatory stimuli. MATERIALS AND METHODS The human BM endothelial cell line (BMEC) and primary human BM endothelial cells were analyzed for expression of Notch ligands and the ability to expand hematopoietic progenitors in an in vitro coculture system. In vivo experiments were carried out to identify modulation of Notch signaling in BM endothelial and hematopoietic cells in mice challenged with tumor necrosis factor-alpha (TNF-alpha) or lipopolysaccharide (LPS), or in Tie2-tmTNF-alpha transgenic mice characterized by constitutive TNF-alpha activation. RESULTS BM endothelial cells were found to express Jagged ligands and to greatly support progenitor's colony-forming ability. This effect was markedly decreased by Notch antagonists and augmented by increasing levels of Jagged2. Physiologic upregulation of Jagged2 expression on BMEC was observed upon TNF-alpha activation. Injection of TNF-alpha or LPS upregulated three- to fourfold Jagged2 expression on murine BM endothelial cells in vivo and resulted in increased Notch activation on murine hematopoietic stem/progenitor cells. Similarly, constitutive activation of endothelial cells in Tie2-tmTNF-alpha mice was characterized by increased expression of Jagged2 and by augmented Notch activation on hematopoietic stem/progenitor cells. CONCLUSIONS Our results provide the first evidence that BM endothelial cells promote expansion of hematopoietic progenitor cells by a Notch-dependent mechanism and that TNF-alpha and LPS can modulate the levels of Notch ligand expression and Notch activation in the BM microenvironment in vivo.
Collapse
Affiliation(s)
- Luis Fernandez
- Center of Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass., USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
537
|
Abstract
The existence of a stem cell niche, or physiological microenvironment, consisting of specialized cells that directly and indirectly participate in stem cell regulation has been verified for mammalian adult stem cells in the intestinal, neural, epidermal, and hematopoietic systems. In light of these findings, it has been proposed that a "cancer stem cell niche" also exists and that interactions with this tumor niche may specify a self-renewing population of tumor cells. We discuss emerging data that support the idea of a veritable cancer stem cell niche and propose several models for the relationship between cancer cells and their niches.
Collapse
Affiliation(s)
- Julie B Sneddon
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA.
| | | |
Collapse
|
538
|
Henschler R, Deak E, Seifried E. Homing of Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2008; 35:306-312. [PMID: 21512647 DOI: 10.1159/000143110] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2008] [Accepted: 07/02/2008] [Indexed: 12/20/2022]
Abstract
SUMMARY: Mesenchymal stem cells (MSCs) are primarily fibroblast-like cells. Yet, once studied under conditions of shear stress when flowing along endothelial cells in vitro or in blood vessels, as well as in classic migration assays such as chemotaxis assays, MSCs have recently been found to function similarly to leukocytes in many ways. Firstly, MSCs express several homing receptors which are typically activated during extravasation of leukocytes. Secondly, some of these receptors are definitely functional, and required for their tissue localization in certain physiological or pathological contexts. Clinical protocols have in the last few years provided the first data on whether and how human MSCs may work in patients once delivered locally e.g. by injection, or systemically via the intra-arterial or intravenous route. Still, analysis of the ability of MSCs to activate specific homing receptors has up to now received relatively little attention. Moreover, maintenance or alterations of homing receptor expression or functions during good manufacturing practice (GMP) preparation steps, and documentation of presence and function of individual pathways on MSC preparations for clinical use are often missed. Hence, we review here mechanisms predicted to be relevant for adhesion, migration, and homing competence of MSCs. We also discuss some early data on homing of MSCs, deduced from preclinical experiments and from the few clinical studies with MSCs. Finally, we introduce some assays which could be applied to monitor preservation of the homing capacity of MSCs during GMP preparation.
Collapse
Affiliation(s)
- Reinhard Henschler
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Center, Johann Wolfgang Goethe-University Frankfurt, Germany
| | | | | |
Collapse
|
539
|
Buijs JT, van der Pluijm G. Osteotropic cancers: from primary tumor to bone. Cancer Lett 2008; 273:177-93. [PMID: 18632203 DOI: 10.1016/j.canlet.2008.05.044] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Revised: 03/31/2008] [Accepted: 05/29/2008] [Indexed: 12/16/2022]
Abstract
It has long been recognized that primary cancers spread to distant organs with characteristic preference. Bone metastases occur in approximately 70% of patients with advanced breast and prostate cancer, causing severe morbidity and hospitalization. In the last decade, we have gained a better understanding of the mechanisms by which certain tumor types tend to metastasize specifically to bone. It appears that the interaction between the organ microenvironment and cancer cells is fundamental for establishing metastatic growth. Accordingly, Stephen Paget's 'seed and soil' hypothesis - stating that circulating cancer cells (the 'seeds') disperse in all directions, but can accomplish metastases only in organs where the microenvironment (the 'soil') is permissive for their growth - still holds forth today. For this reason, this review uses the 'seed and soil' hypothesis as a template to discuss novel insight and developments in the bone metastasis field.
Collapse
Affiliation(s)
- Jeroen T Buijs
- Department of Urology, Leiden University Medical Center, Leiden, The Netherlands.
| | | |
Collapse
|
540
|
Fleming HE, Janzen V, Lo Celso C, Guo J, Leahy KM, Kronenberg HM, Scadden DT. Wnt signaling in the niche enforces hematopoietic stem cell quiescence and is necessary to preserve self-renewal in vivo. Cell Stem Cell 2008; 2:274-83. [PMID: 18371452 DOI: 10.1016/j.stem.2008.01.003] [Citation(s) in RCA: 372] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Revised: 12/03/2007] [Accepted: 01/02/2008] [Indexed: 12/11/2022]
Abstract
Wingless (Wnt) is a potent morphogen demonstrated in multiple cell lineages to promote the expansion and maintenance of stem and progenitor cell populations. Wnt effects are highly context dependent, and varying effects of Wnt signaling on hematopoietic stem cells (HSCs) have been reported. We explored the impact of Wnt signaling in vivo, specifically in the context of the HSC niche by using an osteoblast-specific promoter driving expression of the paninhibitor of canonical Wnt signaling, Dickkopf1 (Dkk1). Here we report that Wnt signaling was markedly inhibited in HSCs and, unexpectedly given prior reports, reduction in HSC Wnt signaling resulted in reduced p21Cip1 expression, increased cell cycling, and a progressive decline in regenerative function after transplantation. This effect was microenvironment determined, but irreversible if the cells were transferred to a normal host. Wnt pathway activation in the niche is required to limit HSC proliferation and preserve the reconstituting function of endogenous hematopoietic stem cells.
Collapse
Affiliation(s)
- Heather E Fleming
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
541
|
Alkatout I, Kabelitz D, Kalthoff H, Tiwari S. Prowling wolves in sheep's clothing: the search for tumor stem cells. Biol Chem 2008; 389:799-811. [DOI: 10.1515/bc.2008.094] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AbstractThe importance of a subset of cells which have ‘stem like’ characteristics and are capable of tumor initiation has been reported for a range of tumors. Isolation of these tumor-initiating cells (TICs) has largely been based on differential cell surface protein expression. However, there is still much debate on the functional significance of these markers in initiating tumors, as many properties of tumor initiation are modified by cell-cell interactions. In particular, the relationship between TICs and their microenvironment is poorly understood but has therapeutic implications, as the microenvironment can maintain tumor cells in a prolonged period of quiescence. However, a major limitation in advancing our understanding of the crosstalk between TICs and their microenvironment is the lack of sensitive techniques which allow thein vivotracking and monitoring of TICs. Application of newin vivocellular and molecular imaging technologies holds much promise in uncovering the mysteries of TIC behavior at the three-dimensional level. This review will describe recent advances in our understanding of the TIC concept and how the application ofin vivoimaging techniques can advance our understanding of the biological fate of TICs. A supplementary resource guide describing TICs from different malignancies is also presented.
Collapse
|
542
|
Cummings RJ, Mitra S, Lord EM, Foster TH. Antibody-labeled fluorescence imaging of dendritic cell populations in vivo. JOURNAL OF BIOMEDICAL OPTICS 2008; 13:044041. [PMID: 19021368 PMCID: PMC2596711 DOI: 10.1117/1.2966122] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
We report an optical molecular imaging technique that exploits local administration of fluorophore-conjugated antibodies and confocal fluorescence microscopy to achieve high-contrast imaging of host cell populations in normal and tumor tissue in living mice. The method achieves micron-scale spatial resolution to depths greater than 100 mum. We illustrate the capabilities of this approach by imaging two dendritic cell populations in the skin and normal and tumor vasculature in vivo.
Collapse
Affiliation(s)
- Ryan J. Cummings
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642
| | - Soumya Mitra
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY 14642
| | - Edith M. Lord
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642
| | - Thomas H. Foster
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
543
|
Hombach-Klonisch S, Paranjothy T, Wiechec E, Pocar P, Mustafa T, Seifert A, Zahl C, Gerlach KL, Biermann K, Steger K, Hoang-Vu C, Schulze-Osthoff K, Los M. Cancer stem cells as targets for cancer therapy: selected cancers as examples. Arch Immunol Ther Exp (Warsz) 2008; 56:165-80. [PMID: 18512024 DOI: 10.1007/s00005-008-0023-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 05/17/2008] [Indexed: 12/16/2022]
Abstract
It is becoming increasingly evident that cancer constitutes a group of diseases involving altered stem-cell maturation/differentiation and the disturbance of regenerative processes. The observed malignant transformation is merely a symptom of normal differentiation processes gone astray rather than the primary event. This review focuses on the role of cancer stem cells (CSCs) in three common but also relatively under-investigated cancers: head and neck, ovarian, and testicular cancer. For didactic purpose, the physiology of stem cells is first introduced using hematopoietic and mesenchymal stem cells as examples. This is followed by a discussion of the (possible) role of CSCs in head and neck, ovarian, and testicular cancer. Aside from basic information about the pathophysiology of these cancers, current research results focused on the discovery of molecular markers specific to these cancers are also discussed. The last part of the review is largely dedicated to signaling pathways active within various normal and CSC types (e.g. Nanog, Nestin, Notch1, Notch2, Oct3 and 4, Wnt). Different elements of these pathways are also discussed in the context of therapeutic opportunities for the development of targeted therapies aimed at CSCs. Finally, alternative targeted anticancer therapies arising from recently identified molecules with cancer-(semi-)selective capabilities (e.g. apoptin, Brevinin-2R) are considered.
Collapse
|
544
|
Wels J, Kaplan RN, Rafii S, Lyden D. Migratory neighbors and distant invaders: tumor-associated niche cells. Genes Dev 2008; 22:559-74. [PMID: 18316475 DOI: 10.1101/gad.1636908] [Citation(s) in RCA: 277] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The cancer environment is comprised of tumor cells as well as a wide network of stromal and vascular cells participating in the cellular and molecular events necessary for invasion and metastasis. Tumor secretory factors can activate the migration of host cells, both near to and far from the primary tumor site, as well as promote the exodus of cells to distant tissues. Thus, the migration of stromal cells and tumor cells among specialized microenvironments takes place throughout tumor and metastatic progression, providing evidence for the systemic nature of a malignancy. Investigations of the tumor-stromal and stromal-stromal cross-talk involved in cellular migration in cancer may lead to the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- Jared Wels
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10021, USA
| | | | | | | |
Collapse
|
545
|
|
546
|
Abstract
This review highlights major scientific developments over the past 50 years or so in concepts related to stem-cell ecology and to stem cells in motion. Many thorough and eloquent reviews have been presented in the last 5 years updating progress in these issues. Some paradigms have been challenged, others validated, or new ones brought to light. In the present review, we will confine our remarks to the historical development of progress. In doing so, we will refrain from a detailed analysis of controversial data, emphasizing instead widely accepted views and some challenging novel ones.
Collapse
|
547
|
Porter RL, Calvi LM. Communications between bone cells and hematopoietic stem cells. Arch Biochem Biophys 2008; 473:193-200. [PMID: 18410740 DOI: 10.1016/j.abb.2008.04.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 03/31/2008] [Accepted: 04/01/2008] [Indexed: 12/28/2022]
Abstract
The skeletal system, while characterized by a hard tissue component, is in fact an extraordinarily dynamic system, with disparate functions ranging from structural support, movement and locomotion and soft-organ protection, to the maintenance of calcium homeostasis. Amongst these functions, it has long been known that mammalian bones house definitive hematopoiesis. In fact, several data demonstrate that the bone microenvironment provides essential regulatory cues to the hematopoietic system. In particular, interactions between the bone-forming cells, or osteoblasts, and the most primitive Hematopoietic Stem Cells (HSC) have recently been defined. This review will focus mainly on the role of osteoblasts as HSC regulatory cells, discussing the signaling mechanisms and molecules currently thought to be involved in their modulation of HSC behavior. We will then review additional cellular components of the HSC niche, including endothelial cells and osteoclasts. Finally, we will discuss the potential clinical implications of our emerging understanding of the complex HSC microenvironment.
Collapse
Affiliation(s)
- R L Porter
- Endocrine-Metabolism Division, Department of Medicine, University of Rochester School of Medicine and Dentistry, P.O. Box 693, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | |
Collapse
|
548
|
Abstract
Optical imaging is an emerging field with a wide range of biomedical research and clinical applications, both current and future. It comprises several classes of techniques that are capable of providing information at the molecular, cellular, tissue, and whole-animal levels. These techniques match well with emerging genomic and proteomic technologies that enable development of optical "probes," as well as with nanotechnologies for multifunctional imaging and drug delivery. These advances have enormous potential to accelerate drug discovery/development by providing predictive information on mechanisms of action and biological responses.
Collapse
|
549
|
Raykov Z, Rommelaere J. Potential of tumour cells for delivering oncolytic viruses. Gene Ther 2008; 15:704-10. [DOI: 10.1038/gt.2008.34] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
550
|
Bantly AD, Gray BD, Breslin E, Weinstein EG, Muirhead KA, Ohlsson-Wilhelm BM, Moore JS. CellVue Claret, a new far-red dye, facilitates polychromatic assessment of immune cell proliferation. Immunol Invest 2008; 36:581-605. [PMID: 18161520 DOI: 10.1080/08820130701712461] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Flow cytometric analyses of immune cell proliferation, differentiation, and function are limited by the number of different fluorochromes that can be resolved simultaneously. Additional colors to expand functional analytic capability will facilitate higher dimensional analyses of heterogeneous cell populations by basic and clinical scientists. Our aim in these studies was to evaluate CellVue Claret, a fluorescent, far-red emitting, membrane intercalating dye (excitation maximum: 655 nm, emission maximum 677 nm), as an alternative and/or complementary probe to PKH26 and CFSE(1) for polychromatic studies of immune cell proliferation and function. Using a BD FACSCalibur and human peripheral blood mononuclear cells (PBMCs) from 8 different donors (2 donors studied twice), we compared CellVue Claret with the two most commonly used visible-emitting proliferation dyes, PKH26 and CFSE, in terms of: (1) compatibility with 7-Amino-actinomycin D (7-AAD) as a viability marker; (2) effect of dye labeling on lymphocyte viability; and (3) the proliferative response of CD3+ T lymphocytes from 0-96 hours as assessed by dilution of each of the 3 cell tracking dyes in cultures stimulated with anti-CD3 plus IL-2. Post-labeling recoveries and viabilities were similar for all 3 dyes, with modestly higher initial staining intensities and coefficients of variation for CellVue Claret than for CFSE or PKH26. Lymphocyte viabilities in stimulated or unstimulated cultures were also unaffected by choice of dye. Proliferative responses of viable CD3+ lymphocytes were comparable for all three dyes, whether results were reported as Proliferative Fraction (percent of cells that had divided one or more times) or as Precursor Frequency (percent of parent population that had gone on to proliferate in response to anti-CD3 plus IL-2). In summary, T cell proliferation analysis using CellVue Claret gives results equivalent to those obtained with PKH26 or CFSE, expanding the choice of proliferation dyes suitable for use in high dimensional polychromatic studies on flow cytometers with far red (633 nm-658 nm) excitation capabilities.
Collapse
Affiliation(s)
- Andrew D Bantly
- University of Pennsylvania, Pathology and Laboratory Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|