scholarly journals Tumor-associated fibroblasts predominantly come from local and not circulating precursors

2016 ◽  
Vol 113 (27) ◽  
pp. 7551-7556 ◽  
Author(s):  
Ainhoa Arina ◽  
Christian Idel ◽  
Elizabeth M. Hyjek ◽  
Maria-Luisa Alegre ◽  
Ying Wang ◽  
...  

Fibroblasts are common cell types in cancer stroma and lay down collagen required for survival and growth of cancer cells. Although some cancer therapy strategies target tumor fibroblasts, their origin remains controversial. Multiple publications suggest circulating mesenchymal precursors as a source of tumor-associated fibroblasts. However, we show by three independent approaches that tumor fibroblasts derive primarily from local, sessile precursors. First, transplantable tumors developing in a mouse expressing green fluorescent reporter protein (EGFP) under control of the type I collagen (Col-I) promoter (COL-EGFP) had green stroma, whereas we could not find COL-EGFP+ cells in tumors developing in the parabiotic partner lacking the fluorescent reporter. Lack of incorporation of COL-EGFP+ cells from the circulation into tumors was confirmed in parabiotic pairs of COL-EGFP mice and transgenic mice developing autochthonous intestinal adenomas. Second, transplantable tumors developing in chimeric mice reconstituted with bone marrow cells from COL-EGFP mice very rarely showed stromal fibroblasts expressing EGFP. Finally, cancer cells injected under full-thickness COL-EGFP skin grafts transplanted in nonreporter mice developed into tumors containing green stromal cells. Using multicolor in vivo confocal microscopy, we found that Col-I–expressing fibroblasts constituted approximately one-third of the stromal mass and formed a continuous sheet wrapping the tumor vessels. In summary, tumors form their fibroblastic stroma predominantly from precursors present in the local tumor microenvironment, whereas the contribution of bone marrow-derived circulating precursors is rare.

2009 ◽  
Vol 131 (10) ◽  
Author(s):  
Jinjin Ma ◽  
Kristen Goble ◽  
Michael Smietana ◽  
Tatiana Kostrominova ◽  
Lisa Larkin ◽  
...  

The incidence of ligament injury has recently been estimated at 400,000/year. The preferred treatment is reconstruction using an allograft, but outcomes are limited by donor availability, biomechanical incompatibility, and immune rejection. The creation of an engineered ligament in vitro solely from patient bone marrow stromal cells (has the potential to greatly enhance outcomes in knee reconstructions. Our laboratory has developed a scaffoldless method to engineer three-dimensional (3D) ligament and bone constructs from rat bone marrow stem cells in vitro. Coculture of these two engineered constructs results in a 3D bone-ligament-bone (BLB) construct with viable entheses, which was successfully used for medial collateral ligament (MCL) replacement in a rat model. 1 month and 2 month implantations were applied to the engineered BLBs. Implantation of 3D BLBs in a MCL replacement application demonstrated that our in vitro engineered tissues grew and remodeled quickly in vivo to an advanced phenotype and partially restored function of the knee. The explanted 3D BLB ligament region stained positively for type I collagen and elastin and was well vascularized after 1 and 2 months in vivo. Tangent moduli of the ligament portion of the 3D BLB 1 month explants increased by a factor of 2.4 over in vitro controls, to a value equivalent to those observed in 14-day-old neonatal rat MCLs. The 3D BLB 1 month explants also exhibited a functionally graded response that closely matched native MCL inhomogeneity, indicating the constructs functionally adapted in vivo.


Blood ◽  
2002 ◽  
Vol 99 (9) ◽  
pp. 3263-3271 ◽  
Author(s):  
Maria Montoya ◽  
Giovanna Schiavoni ◽  
Fabrizio Mattei ◽  
Ion Gresser ◽  
Filippo Belardelli ◽  
...  

Abstract Resting dendritic cells (DCs) are resident in most tissues and can be activated by environmental stimuli to mature into potent antigen-presenting cells. One important stimulus for DC activation is infection; DCs can be triggered through receptors that recognize microbial components directly or by contact with infection-induced cytokines. We show here that murine DCs undergo phenotypic maturation upon exposure to type I interferons (type I IFNs) in vivo or in vitro. Moreover, DCs either derived from bone marrow cells in vitro or isolated from the spleens of normal animals express IFN-α and IFN-β, suggesting that type I IFNs can act in an autocrine manner to activate DCs. Consistent with this idea, the ability to respond to type I IFN was required for the generation of fully activated DCs from bone marrow precursors, as DCs derived from the bone marrow of mice lacking a functional receptor for type I IFN had reduced expression of costimulatory and adhesion molecules and a diminished ability to stimulate naive T-cell proliferation compared with DCs derived from control bone marrow. Furthermore, the addition of neutralizing anti–IFN-α/β antibody to purified splenic DCs in vitro partially blocked the “spontaneous” activation of these cells, inhibiting the up-regulation of costimulatory molecules, secretion of IFN-γ, and T-cell stimulatory activity. These results show that DCs both secrete and respond to type I IFN, identifying type I interferons as autocrine DC activators.


Blood ◽  
1993 ◽  
Vol 82 (12) ◽  
pp. 3580-3591
Author(s):  
N Falla ◽  
Vlasselaer Van ◽  
J Bierkens ◽  
B Borremans ◽  
G Schoeters ◽  
...  

In the presence of beta-glycerophosphate and vitamin C, cultures of normal mouse bone marrow cells form three-dimensional structures that stain positive with the Von Kossa technique and express alkaline phosphatase (ALP), collagen type I, and osteocalcin. Little is known about the characteristics and frequency of the cells that contribute to this phenomenon. Most likely, mature osteoblastic cells do not contribute to the nodule formation because no osteocalcin expressing cells are detected in the flushed marrow by in situ hybridization. Limiting dilution analysis shows that, in normal bone marrow, 1 of 2.2 x 10(5) cells has the potency to form a bone nodule and to express ALP, collagen, and osteocalcin in a temporal fashion. Upon in vivo treatment with 5-fluorouracil (5-FU), this frequency increases 12-fold, eg, 1 in 1.75 x 10(4) cells shows osteogenic activity. In comparison, fibroblast colony forming cells occur at a frequency of 1 of 2.5 x 10(4) or 1 of 5 x 10(3) plated cells in normal or 5-FU-treated marrow, respectively. Using density centrifugation, the majority of the osteoprogenitor cells in 5-FU marrow are found in the low-density (1.066 to 1.067 g/mL) fractions. In addition, these cells bind to nylon wool but not to plastic and aggregate in the presence of wheat germ agglutinin and soybean agglutinin. Scanning and transmission electron microscopy shows that the bone nodules in 5-FU marrow cultures are composed of fibroblastoid cells embedded in a mineralized collagen matrix. In conclusion, our results show that a quiescent cell population in the murine bone marrow with fibroblastoid characteristics contributes to the formation of bone-like nodules in vitro.


Spine ◽  
2005 ◽  
Vol 30 (10) ◽  
pp. 1134-1138 ◽  
Author(s):  
Akihito Minamide ◽  
Munehito Yoshida ◽  
Mamoru Kawakami ◽  
Satoru Yamasaki ◽  
Hirotsugu Kojima ◽  
...  

Blood ◽  
1991 ◽  
Vol 78 (11) ◽  
pp. 2841-2847 ◽  
Author(s):  
CM Dubois ◽  
FW Ruscetti ◽  
JR Keller ◽  
JJ Oppenheim ◽  
K Hestdal ◽  
...  

Abstract Interleukin-1 (IL-1) has profound stimulatory effects on hematopoiesis but the mechanism(s) of action remain unknown. The direct action of IL- 1 on hematopoietic progenitor cells requires the presence of a specific IL-1 receptor (IL-1R). In this report, we tested the effect of in vivo IL-1 treatment on the expression of IL-1R on bone marrow (BM) cells. Injection of mice with IL-1 results in a marked upregulation of IL-1R on light-density BM cells as on a subpopulation enriched for myeloid precursors. Pretreatment of mice with anti-type I IL-1R antibody (35F5), which has been shown to prevent the radioprotective effect of IL-1, also blocked IL-1-induced IL-1R expression on BM cells. This antibody did not directly bind and block IL-1 binding to the type II IL- 1R expressed on hematopoietic cells, suggesting that IL-1R upregulation by IL-1 is indirect. It is therefore possible that IL-1 acts on type I IL-1R-expressing accessory cells such as stromal cells or T cells to induce production of hematopoietic growth factors (HGFs). In support of this, granulocyte colony-stimulating factor administration can induce the increase of IL-1R on BM cells. Thus, the increased expression of IL- 1R on hematopoietic BM cells by IL-1 is indirect, probably mediated in part through endogenous HGF production. These results also suggest that the restorative hematopoietic effect of IL-1 occurs through both indirect and direct mechanisms.


Bone ◽  
1997 ◽  
Vol 20 (2) ◽  
pp. 101-107 ◽  
Author(s):  
M. Mizuno ◽  
M. Shindo ◽  
D. Kobayashi ◽  
E. Tsuruga ◽  
A. Amemiya ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 673-673
Author(s):  
Amanda C. LaRue ◽  
Masahiro Masuya ◽  
Paul A. Fleming ◽  
Richard P. Visconti ◽  
Makio Ogawa ◽  
...  

Abstract The role of the tumor stroma in solid tumor progression and invasion has become an important area of current research and has potential as a therapeutic target. The tumor stroma is composed of a variety of cell types including immune cells, inflammatory cells, fibroblasts and vascular cells (i.e., endothelial cells and pericytes). It was recently reported that crude bone marrow cells have the ability to transdifferentiate into fibroblast-like cells within the tumor capsule and fibroblast-like pericytes associated with the tumor vasculature. However, it is unclear whether these cells are of hematopoietic stem cell (HSC) or stromal/mesenchymal stem cell (SSC) origin. We tested the hypothesis that tumor fibroblasts are derived from HSCs by combining a single HSC transplantation strategy with murine tumor models. For these studies, single cells were sorted from transgenic enhanced green fluorescent protein (EGFP) murine bone marrow based on surface antigen expression (Lin−c-kit+Sca-1+CD34−) or Hoechst 33342 dye exclusion (side population). Individual cells were deposited into wells of a 96-well plate and cultured for one week. Each viable clone (derived from a single stem cell) was then transplanted into a lethally irradiated mouse. Two months later, mice showing high levels (60–90%) of multilineage hematopoietic reconstitution were injected subcutaneously with either murine melanoma or Lewis lung carcinoma cells. Murine melanoma cells (K1735-M2) were a generous gift from Mary Hendrix (University of Iowa, IA) and murine Lewis lung carcinoma (LLC1) were obtained from ATCC (Manassas, VA). After 7–10 days, tumors were extracted and processed for paraffin sectioning. Analysis of thin 3-micrometer sections using differential interference contrast (DIC) and epifluorescence microscopy showed two major populations of EGFP+ cells within the tumor. The first was found within the tumor capsule and had morphological characteristics of fibroblasts. To determine if these cells were indeed fibroblasts, in situ hybridization studies were conducted using a riboprobe for the alpha 1 subunit of type I collagen. Type I collagen mRNA expression was detected in a subpopulation of EGFP+ cells in the tumor capsule, indicating that these HSC-derived EGFP+ cells are fibroblasts. The second cell type was found associated with the blood vessels of the tumor. Analysis of the morphology of these cells using high magnification DIC and epifluorescence microscopy suggested that these cells were not endothelial cells, but support cells (pericytes) in close contact with the endothelium. Immunohistochemical examination of these cells using antibodies to CD31, a protein expressed by endothelial cells, and laser scanning confocal microscopy confirmed that these cells were not endothelial cells (EGFP+/CD31−). Transplantation of 100 Lin−c-kit+ Sca-1+CD34− bone marrow cells without culture produced the same results. These findings establish a hematopoietic origin of fibroblasts comprising the tumor capsule and pericytes of the tumor vasculature.


Blood ◽  
1993 ◽  
Vol 82 (12) ◽  
pp. 3580-3591 ◽  
Author(s):  
N Falla ◽  
Vlasselaer Van ◽  
J Bierkens ◽  
B Borremans ◽  
G Schoeters ◽  
...  

Abstract In the presence of beta-glycerophosphate and vitamin C, cultures of normal mouse bone marrow cells form three-dimensional structures that stain positive with the Von Kossa technique and express alkaline phosphatase (ALP), collagen type I, and osteocalcin. Little is known about the characteristics and frequency of the cells that contribute to this phenomenon. Most likely, mature osteoblastic cells do not contribute to the nodule formation because no osteocalcin expressing cells are detected in the flushed marrow by in situ hybridization. Limiting dilution analysis shows that, in normal bone marrow, 1 of 2.2 x 10(5) cells has the potency to form a bone nodule and to express ALP, collagen, and osteocalcin in a temporal fashion. Upon in vivo treatment with 5-fluorouracil (5-FU), this frequency increases 12-fold, eg, 1 in 1.75 x 10(4) cells shows osteogenic activity. In comparison, fibroblast colony forming cells occur at a frequency of 1 of 2.5 x 10(4) or 1 of 5 x 10(3) plated cells in normal or 5-FU-treated marrow, respectively. Using density centrifugation, the majority of the osteoprogenitor cells in 5-FU marrow are found in the low-density (1.066 to 1.067 g/mL) fractions. In addition, these cells bind to nylon wool but not to plastic and aggregate in the presence of wheat germ agglutinin and soybean agglutinin. Scanning and transmission electron microscopy shows that the bone nodules in 5-FU marrow cultures are composed of fibroblastoid cells embedded in a mineralized collagen matrix. In conclusion, our results show that a quiescent cell population in the murine bone marrow with fibroblastoid characteristics contributes to the formation of bone-like nodules in vitro.


Sign in / Sign up

Export Citation Format

Share Document