scholarly journals Cytogenetic Testing by Fluorescence in Situ Hybridization Is Improved by Plasma Cell Sorting in Multiple Myeloma

Author(s):  
Jihye Ha ◽  
Hyunsoo Cho ◽  
Taek Gyu Lee ◽  
Saeam Shin ◽  
Haerim Chung ◽  
...  

Abstract Accurate detection of cytogenetic abnormalities has become more important for improving risk-adapted treatment strategies in multiple myeloma (MM). However, precise cytogenetic testing by fluorescence in situ hybridization (FISH) is challenged by the dilution effect of bone marrow specimens and poor growth of plasma cells ex vivo. To address these issues, we compared the performances of three different enrichment modalities for FISH: direct FISH, fluorescence immunophenotyping and interphase cytogenetics as a tool for the investigation of neoplasms (FICTION) technique, and a plasma cell sorting FISH with fluorescence-activated cell sorter (FACS). We examined cytogenetic abnormalities in bone marrow cells of 493 patients with newly diagnosed MM and compared the efficacy of each modality. FISH disclosed cytogenetic abnormalities in 38.0% of samples by direct FISH, 56.3% by FICTION, and 95.5% by FACS-FISH, and the percentage of cells with abnormal signals detected by FISH was higher by FACS-FISH than direct FISH or FICTION. Our results suggest that the efficacy of FISH is dependent on the plasma cell enrichment modalities and reveal that plasma cell sorting FISH with FACS enables better detection of cytogenetic abnormalities in diagnostic MM samples with low plasma cell frequency.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 719-719
Author(s):  
Xuefeng Wang ◽  
Simon C. Shin ◽  
Chiang Andy ◽  
Dao Pan ◽  
David Rawlings ◽  
...  

Abstract Introduction Platelets may comprise an ideal vehicle for delivering FVIII in hemophilia A (HemA) as FVIII stored in platelet α-granules is protected from neutralization by inhibitory antibodies and, during bleeding, activated platelets locally excrete their contents to promote clot formation. In order to avoid specific challenges posed by ex vivo gene delivery including, in particular, the requirement to pre-condition the subject, we evaluated intraosseous (IO) infusion of self-inactivating lentiviral vectors (LV) for in situ gene transfer into bone marrow cells. In previous studies, we confirmed that hematopoietic stem cells (HSCs) can be efficiently transduced to express GFP after IO administration of LV driven by a MND promoter (M-GFP-LV). Methods In the current study, we aimed at limiting transgene expression to the megakaryocyte lineages using IO delivery of 20 µL LV containing either GFP (G-GFP-LV) or a B-domain variant human FVIII (G-F8-LV) gene under the control of megakaryocyte strictly specific promoter glycoprotein 1bα (Gp1bα). Results In M-GFP-LV treated control mice, GFP was detected in 6.4% of HSCs, 3.4% of B220+, and 9.0% of CD11c+ bone marrow cells on day 29. In contrast, in G-GFP-LV (6.0E+08 TU/mL) treated mice, GFP was undetectable in bone marrow HSCs, B220+, CD11c+ or CD11b+ cells. GFP expression level in platelets of G-GFP-LV treated mice was ten folds of that in M-GFP-LV treated mice (0.1% vs 0.01%). It indicated that in platelets, the activity of Gp1bα was stronger than that of MND. More importantly, GFP expression levels were stable over 100 days, suggesting that platelets containing the transgene products did not elicit transgene-specific immune responses. Next, we treated HemA mice with G-F8-LV (6.0 E+07 TU/mL). There was no detectable hFVIII expression in bone marrow HSCs on day 8 or in blood cells (CD3ε+, B220+, CD11c+ or CD11b+) on day 35. However, up to 3% platelets express hFVIII on day 91. These results suggested that HSCs in HemA mice were successfully transduced by G-F8-LV after IO infusion, and in the long term, FVIII was synthesized in megakaryocytes and stored in platelet α-granules. In treated mice, the average percentage of platelets expressing hFVIII was stable at 1-2% from day 27 to day 160. The average FVIII antigen level in platelets on day 112 was 1 mU per 1 × 108 platelets, which was comparable with platelet FVIII in transgenic and ex vivo gene therapy treated mice. We also evaluated LV-treated HemA animals for phenotypic correction of bleeding diathesis by tail clip assay. The blood loss was 41% (n=7), 48% (n=5) and 33% (n=5) compared with control HemA (normalized to 100%), mock treated HemA (∼100%), and wild-type (2.5%) mice on days 35, 118 and 160, respectively. Additionally, there was neither detectable FVIII activity nor anti-FVIII antibodies in blood on day 160, which indicated that there was insignificant leaky expression of FVIII in other cells. Finally, we also infused G-F8-LV into HemA inhibitor mice. Inhibitors were induced by repeated injection of 3U recombinant hFVIII. The average antibody level was 80 Bethesda Unit before IO infusion of the vectors. In G-F8-LV treated mice, the average hFVIII antigen level on day 27 was 0.74 mU per 1 x 108 platelets (n=5). Bleeding assay was performed on day 160. The blood loss of treated mice was significantly reduced compared with untreated HemA mice, indicating that IO infusion of G-F8-LV can overcome anti-FVIII antibodies and correct hemophilia phenotype. Conclusion We have successfully transduced HSCs in situ by a single infusion of LVs into bone marrow to correct hemophilia A. Gp1bα promoter in lentiviral vectors can specifically direct the transgene expression in mouse platelets. Following IO infusion of G-F8-LV, FVIII stored in platelets can persistently and partially correct the HemA phenotype for at least five months (experimental duration) in mice with and without pre-existing inhibitors. Overall, direct transduction of bone marrow cells targeting platelet-specific FVIII expression may provide an effective therapy to treat severe hemophilia A patients with high-titer inhibitors. Disclosures: No relevant conflicts of interest to declare.


1996 ◽  
Vol 44 (9) ◽  
pp. 1069-1074 ◽  
Author(s):  
S K Nilsson ◽  
R Hulspas ◽  
H U Weier ◽  
P J Quesenberry

Studies of transplantation biology rely on the detection of donor hemopoietic cells in transplant recipients. Traditionally this has been achieved through ex vivo techniques, including flow cytometric analysis of cell surface markers to detect cells expressing specific epitopes, histochemical detection of cytoplasmic proteins, and the detection of Y chromosome-specific sequences by DNA hybridization. Studies using congenic models, such as the Ly5.1/5.2 mouse, or the utilization of fluorescent dyes, such as PKH-26, have allowed more in-depth analysis of transplantation, beginning to address key issues such as cell homing through cell tracking and elucidation of the "stem cell niche." However, these methods are limited by labeling sensitivity, specificity, crossreactivity and, in the case of PKH-26 labeling, the number of cell divisions the transplanted cells can make before the signal disappears. We have developed a fluorescent in situ hybridization (FISH) technique that utilizes a murine Y chromosome-specific "painting" probe to identify in situ individual transplanted male cells in paraffin-embedded sections of female whole bone marrow while maintaining good morphological integrity. This method is highly sensitive and specific, labeling more than 99% of male cells and no female cells, allowing each transplant to be assessed at the individual cell level. The technique provides unique opportunities to follow the path taken by transplanted cells, both during homing into the marrow and through their maturation and differentiation into mature, functional hemopoietic cells.


1993 ◽  
Vol 178 (3) ◽  
pp. 1091-1096 ◽  
Author(s):  
P Corradini ◽  
M Boccadoro ◽  
C Voena ◽  
A Pileri

Multiple myeloma is a B cell malignancy characterized by the expansion of plasma cells producing monoclonal immunoglobulins (Ig). It has been regarded as a tumor arising at the B, pre-B lymphocyte, or even stem cell level. Precursor cells are presumed to proliferate and differentiate giving rise to the plasma cell clonal expansion. Antigenic features and specific Ig gene rearrangement shared by B lymphocytes and myeloma cells have supported this hypothesis. However, the existence of such a precursor is based upon indirect evidence and is still an open question. During differentiation, B cells rearrange variable (V) regions of Ig heavy chain genes, providing a specific marker of clonality. Using an anchor polymerase chain reaction assay, these rearranged regions from five patients with multiple myeloma were cloned and sequenced. The switch of the Ig constant (C) region was used to define the B cell differentiation stage: V regions are linked to C mu genes in pre-B and B lymphocytes (pre-switch B cells), but to C gamma or C alpha in post-switch B lymphocytes and plasma cells (post-switch B cells). Analysis of bone marrow cells at diagnosis revealed the presence of pre-switch B cells bearing plasma cell V regions still joined to the C mu gene. These cells were not identified in peripheral blood, where tumor post-switch B cells were detected. These pre-switch B cells may be regarded as potential myeloma cell precursors.


Blood ◽  
1992 ◽  
Vol 79 (7) ◽  
pp. 1775-1780 ◽  
Author(s):  
RJ Kreitman ◽  
CB Siegall ◽  
DJ FitzGerald ◽  
J Epstein ◽  
B Barlogie ◽  
...  

Abstract IL-6-PE4E is a recombinant protein consisting of interleukin-6 (IL-6) fused to a mutant form of Pseudomonas exotoxin in which four basic amino acids are changed to glutamate (PE4E). The chimeric toxin has been previously shown to specifically kill malignant hepatic, prostatic, epidermoid, and myeloma cell lines in vitro. To explore the possible clinical utility of IL-6-PE4E, particularly as an agent for ex vivo purging of marrow for autologous bone marrow transplantation (ABMT), we tested malignant cells from patients with multiple myeloma for sensitivity to this chimeric toxin. Ficoll-purified bone marrow cells were incubated with and without IL-6-toxin for 2 to 3 days. Eight of the 15 myeloma patients had cells that were sensitive to IL-6-toxin as measured by a decrease in the level of protein synthesis. Cells from five patients were very sensitive to IL-6-PE4E, with 50% inhibition of protein synthesis (ID50) achieved at or below 6 ng/mL (7 x 10(-11) mol/L). Cells from three additional patients showed moderate sensitivity, with ID50s between 30 and 140 ng/mL. The remaining seven samples showed little or no sensitivity, with ID50s greater than or equal to 400 ng/mL. Normal bone marrow cells or normal BFU-E and CFU-GM were resistant to the IL-6-toxin even at 1,000 ng/mL. Neither IL-6, IL- 2-PE4E, nor an enzymatically deficient mutant of IL-6-PE4E was cytotoxic toward the myeloma cells, indicating that the cytotoxic effect of IL-6-PE4E required the adenosine diphosphate-ribosylation function as well as the specific ligand. Our data suggest that IL-6- toxin could be effective in ex vivo marrow purging in selected multiple myeloma patients who are candidates for ABMT, and that this toxin should also be investigated further for in vivo therapy.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 12-13
Author(s):  
Beau M Idler ◽  
Olivia Perez De Acha ◽  
Owen Lockerbie ◽  
Ken Flanagan ◽  
Fredrik Lehmann ◽  
...  

Introduction: Despite the progress that has been made for standard risk multiple myeloma (MM), subsets of patients with the most advanced and aggressive plasma cell dyscrasias still suffer comparatively poor outcomes. One example is plasma cell leukemia (PCL), which carries a median overall survival of under two years. For patients with PCL, response to front line therapy occurs but is often short-lived, ultimately giving way to aggressive multi-drug resistant disease and patient mortality. Thus, there is a need for the development of new strategies that improve the prognoses for these patients. Melflufen (melphalan flufenamide) is a first-in-class peptide-drug conjugate that is currently in late-phase clinical trials for multiple myeloma. This highly lipophilic agent is preferentially retained in malignant plasma cells (MPCs), where overexpressed aminopeptidases lead to trapping of the alkylator melphalan. We evaluated the anti-myeloma effects of melflufen on patient samples treated ex vivo, and found pronounced sensitivity to melflufen in most samples, with particularly potent efficacy in PCL samples. Methods: Bone marrow aspirate or peripheral blood samples were obtained from patients with plasma cell disorders after IRB approval and informed consent. Ex vivo efficacy of melflufen and melphalan were compared using our Myeloma Drug Sensitivity Testing (My-DST) platform that optimizes viability and tests the malignant cells in the context of the normal cells from their microenvironment (Walker et al, Blood Advances, 2020). In brief, mononuclear cells from patients with plasma cell dyscrasia, including MM and PCL, were isolated and cultured in triplicate wells with titrations of melphalan, melflufen or untreated controls for 48 hours. Post-treatment survival was measured by high-throughput flow cytometry with antibodies for CD138, CD38, CD45 and CD19, and a live/dead dye to discriminate viable MPCs from normal bone marrow cells. EC50 values were determined from these titrations using nonlinear regression curve fits. When the EC50 for melflufen was established in My-DST, a single dose concentration of 10 nM was used to screen patient samples and distinguish relative sensitivity or resistance. Results: Using the My-DST approach with 48 hour drug treatments, melflufen significantly decreased the viable MPC populations, whereas melphalan had little effect (Fig 1A). Concurrent titrations revealed significantly higher MPC sensitivity to melfufen (mean melphalan EC50 = not reached, mean melflufen EC50 = 22.9 nM) (Fig 1B). By comparison to another alkylator, cyclophosphamide's active metabolite has an EC50 of 3.75 µM in this assay. Response to melflufen was accentuated in 2/3 PCL samples tested (HTB-1802.1, HTB-1389.1), with the EC50 < 1nM (Fig 1B). Melflufen demonstrated toxicity in CD45 positive white blood cells, which is consistent with neutropenia observed in clinical trials (data not shown). In single dose screening studies in additional MM patient samples, 4/8 (50%) showed >20% decrease in viable MPCs after incubation with melflufen at 10 nM (Fig 1C). Overall, using those parameters for ex vivo "response" to meflufen, 3/3 patients with PCL responded, 5/6 patients with del(17p) responded, and 3/3 patients with c-MYC translocations responded (Fig 1C, italics). In addition, 3/5 samples from patients that were clinically daratumumab-refractory displayed sensitivity to melflufen. Of five samples from patients with prior exposure to alkylators, four were sensitive to melflufen. Conclusion: Overall, these data support that the peptide-drug-conjugate melflufen shows a broad efficacy across samples from patients with plasma cell disorders. Patients facing poor prognoses, including those with PCL, high-risk cytogenetics and daratumumab-refractory disease, have a great need for new treatments. Thus, the encouraging ex vivo results with melflufen in samples from these aggressive subsets support further clinical exploration. In particular, our preliminary data suggest that plasma cell leukemia patients may be exquisitely sensitive to melflufen. To follow-up these findings, we will expand the number of samples tested from PCL and other forms of high-risk MM samples. Ultimately, if the trend for accentuated sensitivity in plasma cell leukemia holds, a clinical approach for melflufen in these patients may improve outcomes for this group. Figure 1 Disclosures Lockerbie: Oncopeptides AB: Current Employment. Flanagan:Oncopeptides AB: Current Employment. Lehmann:Oncopeptides AB: Current Employment. Forsberg:Celgene: Speakers Bureau; Genentech, Inc., Sanofi, Karyopharm, Abbvie: Research Funding. Mark:Takeda: Consultancy; Kayopharm: Consultancy; Bristol-Myers Squibb: Research Funding; Janssen: Research Funding; Celgene: Consultancy; Amgen: Consultancy; Sanofi: Consultancy; Janssen: Consultancy. Sherbenou:Oncopeptides Inc.: Research Funding.


Blood ◽  
1992 ◽  
Vol 79 (7) ◽  
pp. 1775-1780 ◽  
Author(s):  
RJ Kreitman ◽  
CB Siegall ◽  
DJ FitzGerald ◽  
J Epstein ◽  
B Barlogie ◽  
...  

IL-6-PE4E is a recombinant protein consisting of interleukin-6 (IL-6) fused to a mutant form of Pseudomonas exotoxin in which four basic amino acids are changed to glutamate (PE4E). The chimeric toxin has been previously shown to specifically kill malignant hepatic, prostatic, epidermoid, and myeloma cell lines in vitro. To explore the possible clinical utility of IL-6-PE4E, particularly as an agent for ex vivo purging of marrow for autologous bone marrow transplantation (ABMT), we tested malignant cells from patients with multiple myeloma for sensitivity to this chimeric toxin. Ficoll-purified bone marrow cells were incubated with and without IL-6-toxin for 2 to 3 days. Eight of the 15 myeloma patients had cells that were sensitive to IL-6-toxin as measured by a decrease in the level of protein synthesis. Cells from five patients were very sensitive to IL-6-PE4E, with 50% inhibition of protein synthesis (ID50) achieved at or below 6 ng/mL (7 x 10(-11) mol/L). Cells from three additional patients showed moderate sensitivity, with ID50s between 30 and 140 ng/mL. The remaining seven samples showed little or no sensitivity, with ID50s greater than or equal to 400 ng/mL. Normal bone marrow cells or normal BFU-E and CFU-GM were resistant to the IL-6-toxin even at 1,000 ng/mL. Neither IL-6, IL- 2-PE4E, nor an enzymatically deficient mutant of IL-6-PE4E was cytotoxic toward the myeloma cells, indicating that the cytotoxic effect of IL-6-PE4E required the adenosine diphosphate-ribosylation function as well as the specific ligand. Our data suggest that IL-6- toxin could be effective in ex vivo marrow purging in selected multiple myeloma patients who are candidates for ABMT, and that this toxin should also be investigated further for in vivo therapy.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4898-4898
Author(s):  
Eun Hae Cho ◽  
Sang-Mi Lee ◽  
Hyeon-Seok Eom ◽  
In-Suk Kim ◽  
Gyeong-Won Lee ◽  
...  

Abstract Abstract 4898 Introduction The technique of fluorescence immunophenotyping and interphase cytogenetics as a tool for the investigation of neoplasms has recently been introduced to detect molecular cytogenetic abnormalities in plasma cell myeloma of bone marrow (BM) aspirate. However, in case of sub-optimal BM aspirate or the focal distribution of myeloma in the BM, the plasma cells are significantly lower in the BM aspirate than those of biopsy section. Therefore, we have developed a sensitive fluorescence in situ hybridization (FISH) technique which is combined with immunochemistry and is applicable to BM biopsy section for molecular cytogenetic study of plasma cell neoplasms. Patients and Methods Conventional cytogenetic analysis and FISH results of BM samples of 35 multiple myeloma (MM) patients at the time of diagnosis have been evaluated. The probe for IgH rearrangement has been used for hybridization with myeloma cells coupled with CD138 immunostain at BM biopsy section. Results Nineteen patients (54.3%) had abnormal FISH IgH results in biopsy section, whereas seven (20%) cases had abnormal findings in BM aspirate. FISH IgH analysis at biopsy section revealed various signal patterns and proportions (range 6-87%) of cells with atypical signals out of CD138 positive cells. Among five cases with <10% of plasma cells at BM aspirate, four (80%) had abnormal FISH results at biopsy section, whereas one (20%) had abnormal signals at aspirate. There is no correlation between the proportions of cells with atypical signal corrected by the plasma cell count at BM aspirate and the proportions of cells with atypical signal at biopsy section. Conclusions FISH analysis combined with immunostain which is applied at biopsy section is a highly sensitive and convenient technique to detect and quantify monoclonal plasma cells. It could be used for molecular cytogenetic study in plasma cell neoplasms even though there are less than 10% of plasma cells at BM aspirate and the monitoring of residual disease. Disclosures Kong: National cancer center, Korea: Research Funding.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2930-2930
Author(s):  
Eric Crawford ◽  
Victoria Golembiewski-Ruiz ◽  
Mingya Liu ◽  
Robert D MacPhee

Abstract Abstract 2930 Background: Multiple myeloma (MM) is characterized by the clonal proliferation of terminally differentiated plasma cells. These dyscrasias range from the phenotypically benign monoclonal gammopathy of unknown significance (MGUS), to the clinically significant form, multiple myeloma (MM). Molecular cytogenetic abnormalities are one of the measures that has been used to stratify patients. As the clinical cut-off values for individual fluorescence in situ hybridization (FISH) probes can exceed the total percentage of abnormal plasma cells within a specimen, the International Myeloma Workshop Consensus Panel recommends analysis for genetic abnormalities by FISH, preferably after plasma cell sorting, as one means to stratify patients into standard risk or high risk categories. Purpose: We present a novel method for plasma cell isolation in which antibody coated para-magnetic immunobeads are used in a two-step separation process that deposits an enriched population of CD138+ plasma cells directly onto a predefined area of a microscope slide in a manner suitable for subsequent staining and FISH analyses. Materials and Methods: The clinical materials utilized were appropriately anonymized extraneous volumes of bone marrow aspirates obtained after all routine clinical testing requests had been fulfilled. Briefly, 100ml of bone marrow is incubated with a blocking buffer and paramagnetic immunobeads covalently conjugated to anti-CD138 antibody (WaveSense, Irvine, CA) for 45 minutes at room temperature with gentle agitation. Primary enrichment is achieved when the tube is placed in a magnetic dock for 15 minutes allowing the paramagnetic immunobead-plasma cell-complexes to sediment on the side of the reaction tube. Unbound cells remain in the supernatant solution which is gently removed. The cells from the supernatant are still suitable for additional analysis for hematologic disorders not involving a CD138+ lineage. The residual CD138+ cell population is resuspended in blocking buffer. This enriched cell suspension is pipetted into an EpiSep® (WaveSense, Irvine, CA) chambered slide. As the solution diffuses through the unit, the suspended plasma cell-paramagnetic bead complexes are immobilized at the slide's surface as they pass close to a fixed magnet in the slide dock while the residual supernatant solution is eluted into adjacent chambers containing compact absorbent pads. A small volume of Carnoy's fixative is introduced through the EpiSep port to affect an initial fixation of the captured plasma cells deposited on the microscope slide's surface. The unit is then manually separated from the microscope slide. Samples were analyzed with a panel of FISH probes to assess copy number changes for chromosomes 5, 7, 13, 1p/1q, 17p and t(4;14), t(11;14) and t(14;16). Results: Bone marrow from 32 patients with indications of possible or known plasma cell disorders were enriched for CD138+ plasma cells. Conventional non-enriched FISH analysis detected reportable abnormalities in 11 (34%) of patients. With enrichment, abnormalities were detected in 21 (66%) of patients. When compared to abnormalities detected by FISH on non-enriched marrow, new abnormalities were detected in 14 (44%) of cases. Of the14 cases, 5 (36%) cases had abnormalities that shifted the prognosis from standard risk to high. An additional 3 cases (21%) had patterns consistent with an uncharacterized IGH rearrangement which may also hold a higher risk. The increased rate of abnormal cells ranged from 1.2x to 44.0x with an average increase of 9.7x. Cut-off values for the FISH analysis with this enrichment method were comparable to other methods of plasma cell enrichment. Discussion and Conclusions: We evaluated the use of a novel method to isolate and present only the target cell lineage. Deposition of a sorted plasma cell suspension via a slide chamber limits loss of plasma cells and confines these cells to a defined area for analysis. This method preserves cell morphology and avoids loss of cells that can occur with manual slide dropping methods. We found this strategy significantly increased both the sensitivity of detection levels and accuracy for determinations of these low-level, clinically prognostic genetic alterations while reducing the amount of bone marrow required for complete FISH analysis. Disclosures: MacPhee: WaveSense: Consultancy.


2021 ◽  
Vol 42 (Supplement_1) ◽  
pp. S87-S88
Author(s):  
Kuzhali Muthumalaiappan ◽  
Maria Camargo Johnson ◽  
Julia Walczak ◽  
Vimal Subramaniam ◽  
Anthony J Baldea ◽  
...  

Abstract Introduction Previous burn and traumatic injury studies have established that adrenergic signaling is increased after burn injury and may lead to an impairment of hematopoietic cell development in the bone marrow (BM). Nonetheless, mesenchymal stem cells (MSCs), which have gained momentum in regenerative medicine also play a predominant role in the BM niche. Understanding the propensity of the adrenergic receptor (AR) response by MSCs can be utilized for devising targeted therapies. However, the traditional plastic adherence procedure using ex vivo culture of BM cells for several weeks may skew the actual characteristics of MSCs. Our current study focused on isolating MSCs from freshly obtained BM in a murine scald burn model with a goal to characterize the expression pattern of native AR subgroups present on BM MSCs as compared to sham mice. Methods Eight, two-month-old adult female mice were subjected to a 15% total body 3rd degree burn or sham burn. The mice were sacrificed 7 days later. Femurs were removed and total bone marrow cells were flushed out. Multi parametric flow cytometry was used to gate for cells negative for hematopoietic cell markers (CD45, CD11B) and positive for MSC markers (CD105, CD106, SSEA, Ly6A) and AR subgroups (α1, α2, β1, β2, β3). We measured the number of BM MSCs, quantified the subtypes of ARs present on MSCs, and compared the ratio of AR antibody binding per total MSC population. Results Overall the frequency of MSCs per million total BM cells decreased by 48% post-burn injury with165,300 ± 194 in sham versus 110,000 ± 30 in burn displayed as bar graph in Panel A. Over 90% of MSCs consistently express β2 AR and only 10% express α2 AR subgroup in both scald and sham burn. Presence of other subgroups ranged from 50% to 80% of MSCs as seen in histograms to the right of dotted line in Panel B. Our AR propensity score based on AR mean fluorescence intensity adjusted to total number of MSCs present was increased by 2.8-fold for α1, 2.5-fold for β1, 1.6-fold for β3, and 1.3-fold for β2 AR subgroups (Panel C). These findings indicate burn injury not only decreases the frequency of BM MSCs but also increases the affinity of certain AR subgroups present on MSCs. Since BM MSCs are the major source of cytokines, chemokines and growth factors; detailed studies on AR mediated signaling in BM MSCs is warranted. Conclusions Polarization of AR signaling in BM MSCs by burn-induced catecholamines may have broader implications for comorbidities such as bone resorption and muscle wasting observed in human patients post burn trauma.


Sign in / Sign up

Export Citation Format

Share Document