scholarly journals Using the Microwell-mesh to culture microtissues in vitro and as a carrier to implant microtissues in vivo into mice

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Melissa E. Monterosso ◽  
Kathryn Futrega ◽  
William B. Lott ◽  
Ian Vela ◽  
Elizabeth D. Williams ◽  
...  

AbstractProstate cancer (PCa) patient-derived xenografts (PDXs) are commonly propagated by serial transplantation of “pieces” of tumour in mice, but the cellular composition of pieces is not standardised. Herein, we optimised a microwell platform, the Microwell-mesh, to aggregate precise numbers of cells into arrays of microtissues, and then implanted the Microwell-mesh into NOD-scid IL2γ−/− (NSG) mice to study microtissue growth. First, mesh pore size was optimised using microtissues assembled from bone marrow-derived stromal cells, with mesh opening dimensions of 100×100 μm achieving superior microtissue vascularisation relative to mesh with 36×36 μm mesh openings. The optimised Microwell-mesh was used to assemble and implant PCa cell microtissue arrays (hereafter microtissues formed from cancer cells are referred to as microtumours) into mice. PCa cells were enriched from three different PDX lines, LuCaP35, LuCaP141, and BM18. 3D microtumours showed greater in vitro viability than 2D cultures, but neither proliferated. Microtumours were successfully established in mice 81% (57 of 70), 67% (4 of 6), 76% (19 of 25) for LuCaP35, LuCaP141, and BM18 PCa cells, respectively. Microtumour growth was tracked using live animal imaging for size or bioluminescence signal. If augmented with further imaging advances and cell bar coding, this microtumour model could enable greater resolution of PCa PDX drug response, and lead to the more efficient use of animals. The concept of microtissue assembly in the Microwell-mesh, and implantation in vivo may also have utility in implantation of islets, hair follicles or other organ-specific cells that self-assemble into 3D structures, providing an important bridge between in vitro assembly of mini-organs and in vivo implantation.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2580-2580
Author(s):  
Xin Li ◽  
Wen Ling ◽  
Sharmin Khan ◽  
Yuping Wang ◽  
Angela Pennisi ◽  
...  

Abstract Abstract 2580 Intravenously administered mesenchymal stem cells (MSCs) are trapped in pulmonary vascular bed and only few MSCs home to bone or other tissues in physiological or pathological conditions. Following intracardiac injection MSCs pass the lung barrier but their homing to bone and tissue localization is uncertain. The aim of the study was to investigate trafficking and exact localization of human MSCs following intracardiac injection into unchallenged mice and a xenograft bone tumor model. MSCs were isolated from human fetal bones (ABR Inc, Alameda CA) and expanded in DMEM-LG medium supplemented with 10% FBS. Global gene expression profiling revealed that the cultured MSCs were devoid of hematopoietic cells and expressed typical mesenchymal markers such as CD166, CD146 and CD90. We have previously shown that these MSCs are capable of differentiation into osteoblasts and adipocytes and retain their differentiation potential after multiple passages (Haematologica 2006). The MSCs were transduced with a luciferase/GFP reporter in a lentiviral vector and were maximally passaged 8 times before used in vivo. Detection of MSCs in mice was determined by live-animal imaging and ex vivo bioluminescence activity using the IVIS system, by microscopic examination of GFP-expressing cells and by immunohistochemistry for GFP. MSCs (1×106 cells/mouse) were intracardiacly injected into unconditioned SCID mice (n=8) using Dovetail Slide Micromanipulator that ensures accurate injection. Following 2 or 7 days after MSC injection to SCID mice, live-animal imaging revealed bioluminescence activity mainly in the mice abdomen but not bone, while ex vivo examination detected MSCs in various abdominal organs, primarily in reproductive organs, intestine and pancreas. Careful microscopic examination revealed localization of MSCs in draining lymph nodes attached to these organs by connective tissue. Immunohistochemistry showed GFP-expressing MSCs in the adjacent mesenteric lymph nodes but not within the organs. To confirm our findings, MSCs were intracardially injected into C57BL6 mice (n=6) that harbor functional lymph nodes. Evans blue dye which is known to accumulate in and identify lymph nodes, was injected into the rear footpad or lateral tail base of the mice, 3 hours after MSC injection and 30 minutes prior to bioluminescence and florescence analyses. The Evans blue dye and GFP positivity were co-localized, indicating specific trafficking of MSCs to lymph nodes. Culturing of the dissected lymph nodes resulted in release of GFP-expressing MSCs which regained their in vitro morphology. For testing MSCs trafficking in a xenograft model, we used our SCID-rab system constructed by implanting a 4-weeks old rabbit bone into which human myeloma cells were directly injected (Leukemia 2004; Blood 2007). In this model myeloma cells grow restrictively in the implanted bone. MSCs injected intracardiacly into SCID-rab mice were mostly found in mesenteric lymph nodes but were also detected in the myelomatous bone 72 hours after MSCs injection, validating the ability of tumor cells to attract MSCs and that these MSCs are capable of transmigration. We conclude that MSCs primarily traffic to draining lymph nodes, partially explaining their in vivo immunomodulatory activity, and that understanding the mechanism by which MSCs traffic to lymph nodes may help develop approaches to shift their homing to desired organs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 646-646
Author(s):  
Xin Li ◽  
Wen Ling ◽  
Angela Pennisi ◽  
Jianmei Chen ◽  
Sharmin Khan ◽  
...  

Abstract Induction of osteolytic bone disease in multiple myeloma (MM) is caused by activation of osteoclastogenesis and suppression of osteoblastogenesis. Bone formation is reduced mainly through production of inhibitors of osteoblast differentiation by MM cells and by impaired osteogenic differentiation of endogenous mesenchymal stem cells (MSCs). Recently, human placenta has emerged as a potentially valuable source of progenitor cells for multiple therapeutic purposes, including bone repair and cancer (Parolini et al., Stem Cells26:300–311, 2008). The aim of the study was to investigate the effects of human placenta-derived adherent cells (PDAC™) on MM bone disease and tumor growth in the SCID-rab mouse model for MM. PDAC™ are mesenchymal like adherent cells isolated from postpartum human placenta and capable of supporting bone formation in vivo. Bone disease was evaluated by measurements of bone mineral density (BMD) and visualized by X-rays. MM growth was determined by human immunoglobulin (hIg) ELISA and live animal imaging. For in vivo tracking PDAC™ or our stroma-dependent BN MM cell line was transduced with a luciferase/GFP reporter in a lentiviral vector. In the first in vivo experiment, 10 SCID-rab mice were engrafted with a patient’s MM cells. Following establishment of MM and detection of bone disease, luciferase-expressing PDAC™ (1×106 cells/bone) or phosphate-buffered saline (PBS) were injected directly into implanted myelomatous bones in SCID-rab mice (5 mice/group). At experiment’s end (5 wk after cytotherapy) PDAC™ could be detected in mice by live animal imaging. Whereas in control mice, BMD of the implanted bone was reduced from pretreatment levels by 8±4%, administration of PDAC™ resulted in increased BMD of the implanted bone in all mice by 132±20% from pretreatment levels (p<0.0006). Levels of hIg in mice sera (tumor burden) at experiment’s end were 202±56 μg/ml and 12±4 μg/ml in PBS- and PDAC™ -treated hosts, respectively (p<0.007). In the second in vivo experiment hosts engrafted with our luciferase-expressing BN MM line (Li et al., BJH 2007) were similarly injected with PDAC™ or PBS (8 mice/group). Six wk following treatment the BMD of the implanted bone in the control PBS group was reduced by 31±33% (p<0.0009) from pretreatment levels while in PDAC™ –treated group it was slightly reduced by 2±6% from pretreatment levels. Treatment with PDAC™ had no effect on in vivo growth of the BN MM cell line, indicating that prevention of bone disease by PDAC™ was not a consequence of reduced MM. In contrast to fetal MSCs, PDAC™ expressed high levels of OPG (>30 fold) and low levels of RANKL (<5 fold) as determined by qRT-PCR. Differentiation of osteoclast precursors in media supplemented with RANKL and M-CSF was reduced in the presence of PDAC™ or their conditioned media by 60±6% (p<0.004), an effect that was partially blocked by OPG neutralizing antibody (p<0.04). PDAC™ also induced apoptosis of osteoclast precursors as determined by annexin V/PI staining. We conclude that PDAC™ prevent bone loss and promote bone formation in myelomatous bone through simultaneous inhibition of osteoclastogenesis and stimulation of osteoblastogenesis, and that engraftment of PDAC™ inhibits growth of primary MM in vivo.


2018 ◽  
Vol 6 (20) ◽  
pp. 3366-3371 ◽  
Author(s):  
Vikram Singh ◽  
Kundan S. Rawat ◽  
Shachi Mishra ◽  
Tanvi Baghel ◽  
Soobiya Fatima ◽  
...  

Luminescent carbon quantum dots (CQDs) prepared from aqueous beetroot extract were developed as unique fluorescent nanomaterials for in vivo live animal imaging applications.


1992 ◽  
Vol 6 (3) ◽  
pp. 911-913 ◽  
Author(s):  
Joaquin J. Jimenez ◽  
Marty E. Sawaya ◽  
Adel A. Yunis
Keyword(s):  

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Marwa H. Gouda ◽  
Safaa M. Ali ◽  
Sarah Samir Othman ◽  
Samia A. Abd Al-Aziz ◽  
Marwa M. Abu-Serie ◽  
...  

AbstractWound healing is a complicated multicellular process that involves several kinds of cells including macrophages, fibroblasts, endothelial cells, keratinocytes and platelets that are leading to their differentiation towards an anti-inflammatory response for producing several chemokines, cytokine and growth factors. In this study, electrospun nanofiber scaffold named (MNS) is composed of polyvinyl alcohol (PVA)/iota carrageenan (IC) and doped with partially reduced graphene oxide (prGO) that is successfully synthesized for wound healing and skin repair. The fabricated MNS was tested in case of infection and un-infection with E. coli and Staphylococcus and in both of the presence and in the absence of yeast as a natural nutritional supplement. Numerous biochemical parameters including total protein, albumin, urea and LDH, and hematological parameters were evaluated. Results revealed that the MNS was proved to be effective on most of the measured parameters and had exhibited efficient antibacterial inhibition activity. Whereas it can be used as an effective antimicrobial agent in wound healing, however, histopathological findings confirmed that the MNS caused re-epithelialization and the presence of yeast induced hair follicles growth and subsequently it may be used to hide formed head wound scar.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 659-659
Author(s):  
Kevin A. Goncalves ◽  
Megan D. Hoban ◽  
Jennifer L. Proctor ◽  
Hillary L. Adams ◽  
Sharon L. Hyzy ◽  
...  

Abstract Background. The ability to expand human hematopoietic stem cells (HSCs) has the potential to improve outcomes in HSC transplantation and increase the dose of gene-modified HSCs. While many approaches have been reported to expand HSCs, a direct comparison of the various methods to expand transplantable HSCs has not been published and clinical outcome data for the various methods is incomplete. In the present study, we compared several small molecule approaches reported to expand human HSCs including HDAC inhibitors, the aryl hydrocarbon antagonist, SR1, and UM171, a small molecule with unknown mechanism, for the ability to expand phenotypic HSC during in vitro culture and to expand cells that engraft NSG mice. Although all strategies increased the number of phenotypic HSC (CD34+CD90+CD45RA-) in vitro, SR1 was the most effective method to increase the number of NOD-SCID engrafting cells. Importantly, we found that HDAC inhibitors and UM171 upregulated phenotypic stem cell markers on downstream progenitors, suggesting that these compounds do not expand true HSCs. Methods. Small-molecules, SR1, HDAC inhibitors (BG45, CAY10398, CAY10433, CAY10603, Entinostat, HC Toxin, LMK235, PCI-34051, Pyroxamide, Romidepsin, SAHA, Scriptaid, TMP269, Trichostatin A, or Valproic Acid) and UM171 were titrated and then evaluated at their optimal concentrations in the presence of cytokines (TPO, SCF, FLT3L, and IL6) for the ability to expand human mobilized peripheral blood (mPB)-derived CD34+ cells ex vivo . Immunophenotype and cell numbers were assessed by flow cytometry following a 7-day expansion assay in 10-point dose-response (10 µM to 0.5 nM). HSC function was evaluated by enumeration of colony forming units in methylcellulose and a subset of the compounds were evaluated by transplanting expanded cells into sub-lethally irradiated NSG mice to assess engraftment potential in vivo . All cells expanded with compounds were compared to uncultured or vehicle-cultured cells. Results. Following 7 days of expansion, SR1 (5-fold), UM171 (4-fold), or HDAC inhibitors (&gt;3-35-fold) resulted in an increase in CD34+CD90+CD45RA- number relative to cells cultured with cytokines alone; however, only SR1 (18-fold) and UM171 (8-fold) demonstrated enhanced engraftment in NSG mice. Interestingly, while HDAC inhibitors and UM171 gave the most robust increase in the number and frequency of CD34+CD90+CD45RA- cells during in vitro culture, these methods were inferior to SR1 at increasing NSG engrafting cells. The increase in CD34+CD90+CD45RA- cells observed during in vitro culture suggested that these compounds may be generating a false phenotype by upregulating CD90 and down-regulating CD45RA on progenitors that were originally CD34+CD90-CD45RA+. We tested this hypothesis by sorting CD34+CD90-CD45RA+ cells and culturing these with the various compounds. These experiments confirmed that both HDAC inhibitors (33-100 fold) and UM171 (28-fold) led to upregulation of CD90 on CD34+CD90-CD45RA+ cells after 4 days in culture. Since approximately 90% of the starting CD34+ cells were CD90-, these data suggest that most of the CD34+CD90+CD45RA- cells in cultures with HDAC inhibitors and UM171 arise from upregulation of CD90 rather than expansion of true CD34+CD90+CD45RA- cells and may explain the disconnect between in vitro HSC phenotype and NSG engraftment in vivo . This was further confirmed by evaluation of colony forming unit frequency of CD34+CD90-CD45RA+ cells after culture with compounds. Conclusions. We have showed that AHR antagonism is optimal for expanding functional human HSCs using the NSG engraftment model. We also demonstrated that UM171 and HDAC inhibitors upregulate phenotypic HSC markers on downstream progenitors. This could explain the discrepancy between impressive in vitro phenotypic expansion and insufficient functional activity in the NSG mouse model. Therefore, these data suggest caution when interpreting in vitro expansion phenotypes without confirmatory functional transplantation data, especially as these approaches move into clinical trials in patients. Disclosures Goncalves: Magenta Therapeutics: Employment, Equity Ownership. Hoban: Magenta Therapeutics: Employment, Equity Ownership. Proctor: Magenta Therapeutics: Employment, Equity Ownership. Adams: Magenta Therapeutics: Employment, Equity Ownership. Hyzy: Magenta Therapeutics: Employment, Equity Ownership. Boitano: Magenta Therapeutics: Employment, Equity Ownership, Patents & Royalties. Cooke: Magenta Therapeutics: Employment, Equity Ownership, Patents & Royalties.


2009 ◽  
pp. 757-761
Author(s):  
CF Sharpley ◽  
KG Kauter ◽  
JR McFarlane

Cortisol is secreted by the central hypothalamo-pituitary-adrenal axis and affects many target organs and tissues, particularly in response to stressor demands and infection. Recent data reporting cortisol synthesis in hair follicles have shown the existence of a parallel “peripheral” HPA-axis. However, although there is evidence from in vitro studies and single-observation comparisons between groups that cortisol from hair follicles reflects endocrine changes associated with stressor demands, there are no reports to date of repeated measurements of in vivo cortisol responsivity in hair to transitory stressors. This issue was investigated with three males who underwent 1 min cold pressor test (CP). Cortisol response in hair to stressor demand appears to be (a) swift but transitory, (b) localized to the site of the demand and (c) independent of central HPA-axis activity.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3099-3099
Author(s):  
Mara N Zeissig ◽  
Duncan R Hewett ◽  
Krzysztof M Mrozik ◽  
Vasilios Panagopoulos ◽  
Monika Engelhardt ◽  
...  

Introduction:Multiple myeloma (MM) disease progression is dependent on the ability of the MM plasma cells (PC) to leave the bone marrow (BM), re-enter the peripheral blood (PB) and disseminate to other BM sites. Previous studies show that expression of CXCL12 by BM stromal cells is crucial for MM PC retention within the BM. However, the mechanisms which overcome this retention signal enabling MM PC egress and dissemination via the PB are poorly understood. Previous studies in haematopoietic progenitor cells have demonstrated that CCL3 overcomes the CXCL12 retention signal to drive mobilisation to the PB (Lord et al. Blood 1995). Here, we examined the role of the CCL3 chemokine receptor CCR1 in driving MM PC dissemination. Methods and results: Initially, we assessed the expression of CCR1 protein on CD138+CD38++CD45loCD19- PC from 28 MM, 8 MGUS and 2 SMM patients by flow cytometry. Results show CCR1 expression is significantly increased in newly diagnosed MM compared with premalignant MGUS and SMM patients (p=0.03; CCR1 MFI mean±SEM, MGUS: 53.0±33.6; SMM: 37.6±8.9 MM: 250.9±71.6). Furthermore, CCR1 expression on PB MM PC positively correlated with PB MM PC numbers (p=0.03; n=11 patients). To identify mechanistically how CCR1 may promote dissemination, the effect of CCL3 on the response to CXCL12 in human myeloma cell lines (HMCL) was assessed in vitro. The migration of RPMI-8226 and OPM2 cells was induced by CCL3 or CXCL12 chemoattractant in a transwell assay. Notably, pre-treatment of RPMI-8226 or OPM2 with CCL3 abrogated migration towards CXCL12 and blocked F-actin remodelling in response to CXCL12 in vitro. These findings suggest that CCL3 can desensitise cells to exogenous CXCL12, providing a potential mechanism facilitating loss of the CXCL12 retention signal. To confirm whether CCR1 is required for driving MM PC dissemination, homozygous CCR1 knockout (KO) cells were generated using a lentiviral CRISPR/Cas9 system in OPM2 cells. CCR1-KO OPM2 cells were confirmed to have no detectable CCR1 expression by flow cytometry and could no longer migrate towards CCL3 in vitro. Empty vector (EV) or CCR1-KO OPM2 MM PC were injected into the tibia of immune-compromised NOD-scidgamma (NSG) mice. After 4 weeks, primary tumour within the injected tibia and disseminated tumour in the PB and the contralateral tibia and femur was assessed by flow cytometry. We found that mice bearing CCR1-KO cells have a 45.5% decrease in primary tumour growth (p=0.008; % GFP+ of total mononuclear cells, EV: 77.2±17.2; CCR1-KO: 42.1±24.4), a 97.8% reduction in PB MM PC (p<0.0001; EV: 1.39±0.7; CCR1-KO: 0.03±0.046) anda 99.9% reduction in BM tumour dissemination (p<0.0001; EV: 49.5±17; CCR1-KO: 0.019±0.013), compared with controls. In a supportive study, CCR1 was expressed in the murine MM cell line 5TGM1 using lentiviral transduction. 5TGM1-CCR1 cells were confirmed to express CCR1 by qPCR and were able to migrate towards CCL3 in vitro. 5TGM1-CCR1 or EV cells were injected into the tibiae of C57BL/KaLwRij mice and allowed to initiate systemic MM disease for 3.5 weeks. Importantly, while 55% of control mice exhibited disseminated tumours, this increased to 92% with CCR1 expression (p<0.0001; n=12/group). These data suggest that CCR1 expression on MM PC may play an important role in MM PC dissemination. To determine whether therapeutic inhibition of CCR1 prevents dissemination, the effect of a small molecule CCR1 inhibitor, CCR1i, was assessed in vivo. OPM2 EV or RPMI-8226 cells were injected into the tibia of NSG mice and, after 3 days, mice were treated with CCR1i (15mg/kg) or vehicle twice daily by oral gavage for 25 days. OPM2-inoculated CCR1i-treated mice had 66.1% lower PB MM PC (p<0.0001; % GFP+ of total mononuclear cells, vehicle: 23.9±7.2; CCR1i: 8.1±3.8) and a 22.1% reduction in BM dissemination (p=0.0002; vehicle: 78.1±4.8;CCR1i: 60.8±7.1) compared with controls. Similarly, CCR1i treatment reduced BM dissemination by 59.6% in RPMI-8226 bearing mice (p<0.0001; % GFP+ of total mononuclear cells, vehicle: 0.86±0.15; CCR1i: 0.26±0.05). This suggests that CCR1 inhibition can slow tumour dissemination in vivo. Conclusion:This study identified CCR1 as a novel driver of MM PC dissemination in vivo, at least in part by overcoming the CXCL12 retention signal. Importantly, this study demonstrated for the first time that targeting CCR1 can be a viable therapeutic strategy to limit dissemination and potentially slow disease progression. Disclosures Croucher: Trovagene: Employment.


1999 ◽  
Vol 67 (12) ◽  
pp. 6637-6642 ◽  
Author(s):  
Marianne Kretschmar ◽  
Bernhard Hube ◽  
Thomas Bertsch ◽  
Dominique Sanglard ◽  
Renate Merker ◽  
...  

ABSTRACT Peritonitis with Candida albicans is an important complication of bowel perforation and continuous ambulatory peritoneal dialysis. To define potential virulence factors, we investigated 50 strains of C. albicans in a murine peritonitis model. There was considerable variation in their virulence in this model when virulence was measured as release of organ-specific enzymes into the plasma of infected mice. Alanine aminotransferase (ALT) and α-amylase (AM) were used as parameters for damage of the liver and pancreas, respectively. The activities of ALT and AM in the plasma correlated with invasion into the organs measured in histologic sections and the median germ tube length induced with serum in vitro. When the activity of proteinases was inhibited in vivo with pepstatin A, there was a significant reduction of ALT and AM activities. This indicates that proteinases contributed to virulence in this model. Using strains ofC. albicans with disruption of secreted aspartyl proteinase gene SAP1, SAP2, SAP3, orSAP4 through SAP6 (collectively referred to asSAP4-6), we showed that only a Δsap4-6 triple mutant induced a significantly reduced activity of ALT in comparison to the reference strain. In contrast to the Δsap1, Δsap2, and Δsap3 mutants, the ALT induced by the Δsap4-6 mutant could not be further reduced by pepstatin A treatment, which indicates that Sap4-6 may contribute to virulence in this model.


Sign in / Sign up

Export Citation Format

Share Document