microRNA-690 regulates induced pluripotent stem cells (iPSCs …

Stem Cell Research & Therapy201910:59

https://doi.org/10.1186/s13287-019-1154-8

The Author(s).2019

The regulatory mechanism of insulin-producing cells (IPCs) differentiation from induced pluripotent stem cells (iPSCs) in vitro is very important in the phylogenetics of pancreatic islets, the molecular pathogenesis of diabetes, and the acquisition of high-quality pancreatic -cells derived from stem cells for cell therapy.

miPSCs were induced for IPCs differentiation. miRNA microarray assays were performed by using total RNA from our iPCs-derived IPCs containing undifferentiated iPSCs and iPSCs-derived IPCSs at day 4, day 14, and day 21 during step 3 to screen the differentially expressed miRNAs (DEmiRNAs) related to IPCs differentiation, and putative target genes of DEmiRNAs were predicted by bioinformatics analysis. miR-690 was selected for further research, and MPCs were transfected by miR-690-agomir to confirm whether it was involved in the regulation of IPCs differentiation in iPSCs. Quantitative Real-Time PCR (qRT-PCR), Western blotting, and immunostaining assays were performed to examine the pancreatic function of IPCs at mRNA and protein level respectively. Flow cytometry and ELISA were performed to detect differentiation efficiency and insulin content and secretion from iPSCs-derived IPCs in response to stimulation at different concentration of glucose. The targeting of the 3-untranslated region of Sox9 by miR-690 was examined by luciferase assay.

We found that miR-690 was expressed dynamically during IPCs differentiation according to the miRNA array results and that overexpression of miR-690 significantly impaired the maturation and insulinogenesis of IPCs derived from iPSCs both in vitro and in vivo. Bioinformatic prediction and mechanistic analysis revealed that miR-690 plays a pivotal role during the differentiation of IPCs by directly targeting the transcription factor sex-determining region Y (SRY)-box9. Furthermore, downstream experiments indicated that miR-690 is likely to act as an inactivated regulator of the Wnt signaling pathway in this process.

We discovered a previously unknown interaction between miR-690 and sox9 but also revealed a new regulatory signaling pathway of the miR-690/Sox9 axis during iPSCs-induced IPCs differentiation.

Type 1 diabetes (T1D) is defined as dysregulation of homeostatic control of blood glucose due to an absolute insulin deficiency caused by autoimmune destruction of insulin-secreting pancreatic -cells [1]. The transplantation of -cells from a pancreatic donor or augmentation of endogenous -cells regeneration may lead to a cure for T1D. Unfortunately, these methods are restricted by donor tissue availability and tissue rejection and are thus far from being widely applied [2]. Insulin-producing cells (IPCs) derived from pluripotent stem cells in vitro may provide an alternative source of -cells [3]; however, the rate of development of functional and mature IPCs is very low according to the present protocols [4], which will be improved by a thorough understanding of pancreatic organogenesis, including proliferation, differentiation, migration, and maturation of pancreatic progenitor cells.

Considerable evidence has verified that microRNAs (miRNAs) in pancreatic cells regulate gene expression through post-transcriptional modulation [5, 6]. Recently, the global influence of miRNAs on pancreatic development has been assessed by Dicer-knockout mouse embryos. Dicer deficiency resulted in alterations of islet architecture and differentiation markers, accompanied by enhanced apoptosis and defects in all types of endocrine cell formation, particularly that of -cells [7]. Similarly, miR-375 is expressed specifically in pancreatic islets and regulates the proliferation and insulin secretion of -cells by targeting myotrophin (MTPN) and phosphoinositide-dependent protein kinase-1 [8]. Knockdown of miR-375 in ob/ob mice led to a disproportionate ratio of -cells to -cells, high plasma glucagon levels, or even diabetes [9]. In addition, other miRNAs, such as miR-7 and miR-199b-5p, have been studied functionally and reported to selectively affect the development of pancreatic islets, promoting the proliferation of -cells and miR-124a and regulating Foxa2 expression and intracellular signaling in -cells [1012]. These findings, as highlighted above, encouraged us to identify different layers of miRNA regulatory networks, which will provide greater insights into the roles of noncoding RNAs and help further elucidate -cell biology, pancreas formation, and the molecular mechanisms of diabetes etiopathogenesis.

During pancreatic development, the sex-determining region Y (SRY)-box9 (Sox9) factor, which is known to function in campomelic dysplasia, XY sex reversal, and skeletal malformations, has been linked to the proliferation and differentiation of endocrine progenitors [13, 14]. Analysis of cases with Sox9 loss in pancreatic progenitor cells demonstrated a proportional reduction in FoxA2 and Onecut1 expression, along with upregulation of Hnf1b (TCF2), which resulted in a dramatic decrease in endocrine cells without changes in exocrine compartments [15]. Despite a fair understanding of the molecular mechanism by which Sox9 controls pancreatic development, only a few pathways regulated by Sox9 are known. Wnt/-catenin signaling (WNT) has been demonstrated to participate broadly in the differentiation of stem cells, showing a negative regulatory relationship with Sox9 in various contexts [16, 17]. Furthermore, both CTNNB1 (-catenin) and pGSK3 act as downstream target genes, increasing transcriptional activity and decreasing degradation by overexpression of Sox9 [14].

In this study, we identified miR-690 as a differentially expressed transcript during induced pluripotent stem cell (iPSCs)-induced IPCs differentiation in vitro. Surprisingly, predicted mRNA targets, such as Sox9, CTNNB1 (-Catenin), and Stat3, were found to be crucial during the specification of pancreatic progenitor cells and terminal maturation of endocrine cells. Furthermore, the augmentation of miR-690 destabilized IPCs differentiation through direct binding to Sox9 and was likely to have a repressive effect on the Wnt pathway, suggesting an unreported role of miR-690 in modulating key transcription factors and signaling pathways.

C57BL/6J mice were from the animal center of Nantong University. All animal experiments were performed according to the Institutional Animal Care guidelines and were approved by the Animal Ethics Committee of the Medical School of Nantong University.

Mouse GFP-iPSCs were obtained from the Innovative Cellular Therapeutics, Ltd. (Shanghai, China), maintained on feeders in mESC culture conditions, and induced to differentiate into pancreatic IPCs via a three-step protocol as previously described.

Total RNA was isolated using RNAiso Plus (TaKaRa). The first-strand cDNA synthesis for miRNA was performed by using the RevertAid First Strand cDNA Synthesis Kit (Thermo Scientific) and following the manufacturers instructions. The relative expression levels of each miRNA and mRNA were calculated by the 2Ct method as previously described, and GAPDH and U6 were used as the internal normalization controls. Each experiment was performed independently and repeated three times. The qRT-PCR primer sequences were designed and synthesized by GenScript Biotech Corp. (Nanjing, China).

miRNA profiling of iPSC-derived IPCs was carried out by the Professional Oebiotech Corporation (Shanghai, China). In brief, total extracted RNA was labeled with the Agilent miRNA Complete Labeling and Hyb kit (Agilent, Santa Clara, CA, USA) and hybridized to an Agilent Mouse microRNA microarray V21.0. Then, a Gene Expression Wash Buffer kit (Agilent) was used to wash the microarray. Differentially expressed miRNAs (DEmiRNAs) were identified using GeneSpring software (version 13.1, Agilent Technologies, fold change 1.5, P value 0.05). TargetScan and microRNA.org were used to select target genes of DEmiRNAs (P0.05 for both gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis). The feasible regulatory relationships between miRNAs and target genes were analyzed using Cytoscape software (http://www.cytoscape.org/).

Cells were washed with PBS and lysed on ice for 30min with RIPA buffer (high) (Solarbio). Protein concentrations were detected using the BCA Protein Assay (Thermo Fisher Scientific). Total proteins were separated by SDS-PAGE, blotted on PVDF membranes (Millipore, Bedford, MA, USA), and probed with primary antibody in Antibody Dilution Buffer (Solarbio) at 4C overnight. After three washes in TBST, the membranes were incubated with HRP-conjugated secondary antibodies for visualization. Primary antibodies and HRP-conjugated secondary antibodies are listed: anti-Sox9 antibody (Abcam), anti-beta catenin antibody (Abcam), anti-beta actin antibody as a loading control (Abcam), anti-phospho-GSK-3 (Ser9) rabbit mAb (Cell Signaling Technology), anti-phospho-CyclinD1 (Ser90) antibody (affinity), and goat anti-rabbit HRP antibody (affinity).

iPSCs-derived IPCs were transferred into new 24-well plates for 12h. After preincubation in Krebs-Ringer bicarbonate buffer (KRB) without glucose for 120min, the cells were stimulated with KRB containing 0, 5, 15, 30, and 45mM glucose for 120min. The supernatant was collected. Insulin content and secretion from iPSC-derived IPCs were assessed by ELISAs, which were carried out using an ultrasensitive mouse insulin assay kit (Mercodia) following the manufacturers instructions.

iPSCs-derived IPCs grown on glass coverslips were washed with PBS and fixed with 4% paraformaldehyde for 15min at room temperature. Then, these cells were washed thrice (10min every time) and permeabilized with 0.5% (v/v) Triton X-100 for 15min at room temperature. Next, 5% donkey serum was added for 60min, and the cells were stained with different primary antibodies at 4C overnight. Then, the cells were stained with fluorescence secondary antibodies for 1h and DAPI (Solarbio) for 15min. Images were acquired using a Zeiss LSM 510 META confocal microscope (Carl Zeiss, Ltd.). Primary antibodies are listed as follows: anti-insulin antibody (Abcam), anti-C-peptide antibody (Abcam), anti-PDX1 antibody (Abcam), anti-SOX9 antibody (Abcam), antibody-beta catenin antibody (Abcam), anti-NKX6.1 (D804R) rabbit mAb (Cell Signaling Technology). Secondary antibodies included donkey anti-rabbit (Alexa Fluor 647, Abcam), donkey anti-rabbit (Alexa Fluor 555, Abcam), goat anti-guinea pig (Alexa Fluor 647, Abcam), donkey F(ab,)2 anti-goat (Alexa Fluor 594, Abcam), and donkey anti-goat (Alexa Fluor 647, Abcam) antibodies.

For identification of the insulin-positive population, 1106 iPSCs-derived IPCs were digested with trypsin, washed with PBS, and resuspended as single cells by incubation in Reagent 1: Fixation (Beckman Coulter) for 15min. Then, the cells were washed once in PBS, incubated in Reagent 2: Permeabilization (Beckman Coulter) for 20min, and washed once in PBS. Next, the cells were resuspended in PBS with primary antibody and incubated for 30min. The cells were then washed with PBS twice and analyzed with the BD FACSCalibur system (BD Biosciences). The results were analyzed using FlowJo software. All procedures were carried out at room temperature. The primary antibody was anti-h/b/m insulin APC-conjugated rat IgG2A (R&D Systems). The isotype antibody was rat IgG2A control APC-conjugated.

A luciferase reporter assay was performed to observe interactions between miR-690 and Sox9. Wild-type Sox9 and the mutant Sox9 were cloned into the Pezx-FR02 reporter vector for miR-690 targeting. Pezx-FR02 or Pezx-FR02-Sox9-MUT was co-transfected with miR-690 mimic or miRNA mimic control. Firefly and Renilla luciferase activities were assayed with a Dual-Luciferase Assay (Promega, Madison, USA) at 48h post-transfection according to the manufacturers instructions.

Data are presented as the meanstandard deviation (SD) from at least three independent experiments. Significant differences in the relative miRNA or mRNA levels between the experimental groups and their negative controls were determined via Students t test using GraphPad Prism 7.0 (GraphPad Software, Inc.). A P value <0.05 was considered significant.

The differentiation protocol has been described by Huang et al. (Fig.

a, b) [

,

]. The iPSCs obtaining from the Innovative Cellular Therapeutics, Ltd., were identified (Additionalfile

: Figure S1). Importantly, pancreatic -cells are the only IPCs in humans and animals. C-peptide is the active form of insulin. We detected these two markers of mature -cells in iPSC-derived IPCs on day 21 of step 3 to evaluate the efficiency of these insulin-secreting cells. Immunofluorescence assays showed that the majorities of the cells were positive for insulin and C-peptide (Fig.

c). The flow cytometry results also showed that 41.3%0.35% of iPSCs-derived IPCs at the final stage were insulin

(Fig.

d). To determine whether the differentiated cells respond to glucose stimulation, we assessed insulin secretion by exposing IPCs to glucose at different concentrations (0, 5, 15, and 30mM). Treatment with glucose increased insulin secretion in these IPCs, with a peak at the 15mM glucose concentration. No more insulin was induced when the glucose concentration increased to 30mM, suggesting that these IPCs reached the upper limit of their insulin secretion capacity in response to glucose (Fig.

e).

Overview of the differentiation protocol. a Summary of the three-step differentiation protocol. EBs embryoid bodies, MPs multilineage progenitor. b Morphologies of differentiating iPSCs into IPCs at different time points during differentiation. Scale bar: 20m. c Immunofluorescence assay of iPSCs at step 3 on day 21. Co-immunostaining of insulin (red) with C-peptide (green); nuclear DAPI staining is shown in blue. Scale bar: 75m. d Flow cytometry plots illustrating the protein expression of insulin in populations of iPSC-derived IPCs. Black text indicates the percentage of insulin. e Glucose-stimulated insulin secretion in vitro. iPSC-derived IPCs on day 21 of the three-step protocol were exposed to different glucose concentrations (0, 5, and 15mM). The insulin concentration levels were determined

To screen the differentially expressed miRNAs (DEmiRNAs) related to IPCs differentiation, we performed miRNA microarray assays by using total RNA from our iPSCs-derived IPCs containing undifferentiated iPSCs and iPSCs-derived IPCs at day 4 (early stage), day 14 (middle stage), and day 21 (late stage) during step 3. A Venn diagram was used to compare several miRNAs differentially expressed during the three-step induction. The results showed that there were 13 common miRNAs during the three-step induction (Fig.

a). The miRNA expression levels at different time points were clustered and are shown graphically (Fig.

b).

Differentially expressed miRNA profiling and bioinformatic analysis. Differentially expressed miRNAs (P<0.05) were analyzed by hierarchical clustering of log2 values. a Venn diagram showing separate and overlapping differential expression of miRNAs during iPSCs-derived IPCs at the early, middle, and late stages of step 3 compared to that of iPSCs. b Heatmap shows selected differentially expressed miRNAs (fold change 1.5 and P value <0.05). c The regulatory network of miRNA-target genes. Green circles represent target genes, and purple circles represent differentially expressed miRNAs. d Differentially expressed pathways were analyzed by gene ontology (GO) analysis. e KEGG pathway enrichment analysis for target genes. The size of the bubbles represents the number of target genes associated with each pathway

To further understand the role of 13 common DEmiRNAs in iPSCs-derived IPCs, we performed the bioinformatics prediction analysis using two databases (TargetScan and miRanda) respectively to search for putative target genes. There were 332 common target genes after combining data predicted by two databases (miRanda threshold value: binding energy 16.0, align score 158, TargetScan threshold value: context score percentile 30, data not shown). We explored the connections between the DEmiRNAs and putative target genes by building a regulatory network for miRNA-target genes using Cytoscape software (Fig.2c). Then, we investigated the target genes in the KEGG pathways to further study the biological function of the DEmiRNAs (Fig.2d). Interestingly, the WNT signaling pathway was located at the top of the 20 most enriched pathways. Our pathway analysis partly revealed the function of the signature miRNAs, and signal-related function was highlighted among all the subsystems, which was consistent with GO function analyses of the target genes (Fig.2e). To verify the bioinformatic results, we performed qPCR, showing that miR-296, miR-331, miR-345, and miR-690 levels were consistent with the previous trends (Additionalfile2: Figure S2). Of the transcripts that we identified, miR-690, which was persistently highly expressed in the full step 3, drew our attention, as it was reported to regulate Runx2-induced osteogenic differentiation of myogenic progenitor cells; these findings suggest that it may mediate organism differentiation and development. Then, we concentrated on the miR-690 functions during IPCs differentiation.

To explore the specific function of miR-690 in the progression of the three-step induced differentiation, we constructed an agomir vector targeting miR-690 (miR-690-agomir), and miR-690 was overexpressed in MPCs on day 4. The overexpression efficiency of agomir-miR-690 was confirmed by qPCR analysis (Fig.

a). Upregulated miR-690 in MPCs reduced the mRNA expression of several key transcription factors critical for early pancreatic development such as Pdx1, Ngn3, Nkx6.1, Gata4, and Pax4, although the deletion of these nonspecific factors alone was enough to abrogate pancreatic lineage induction (Fig.

b). Immunostaining assays partially verified the results of quantitative RT-PCR analysis (Fig.

c). As expected, IPCs overexpressing miR-690 showed a weak response to glucose stimulation, and high expression of these markers was correlated with the maturation of -cells. Moreover, flow cytometry showed that the population of insulin

cells significantly decreased from 42.4%0.25% to 22.8%0.007% from cells with NC-agomir compared to cells with miR-690-agomir (Fig.

a). The ELISA results of mature IPCs (late stage/day 21) showed that insulin secretion decreased after glucose stimulation (Fig.

b), indicating that IPCs were unable to reduce their glucose concentrations compared with NC cells. Also, we found that IPCs generated after overexpression of miR-690 showed significantly lower mRNA levels of mature -cell and mature -cell markers, such as insulin 1, insulin 2,

,

, and

, than NC-agomir-transfected cells on day 21 of the late stage through quantitative RT-PCR analysis. Interestingly,

expression of -cells was opposite to that of mature -cells and mature -cells, and Mafa expression showed no significant difference between the two groups of cells (Fig.

c). In addition, immunostaining assays confirmed that the co-expression of insulin/C-peptide, insulin/Nkx6.1, and insulin/Pdx1 was consistent with the results from previous quantitative RT-PCR assays (Fig.

d). All these findings showed that miR-690 suppressed the maturation and endocrine functions of IPCs derived from iPSCs, indicating that miR-690 might be a critical regulator of -cells differentiation.

Overexpression of miR-690 inhibits pancreatic differentiation potential. This group of experiments tested the functions of iPSCs-derived IPCs on day 4 of the second step. Quantitative RT-PCR analysis of the expression levels of a miR-690 and b several key transcription factors during the development of pancreatic -cells (Pdx1, NGN3, Nkx2.2, Nkx6.1, Gata4, Gata6, Pax4, Pax6). GAPDH was used as the internal control. Error bars show meanstandard deviation (SD) (n=3). *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001. c Immunofluorescence assay (Nkx6.1 and Pdx1, green; nuclear, blue; scale bar 75m) for protein expression level of Nkx6.1 and Pdx1

Overexpression of miR-690 impaired the functions of terminal iPSCs-derived IPCs. This group of experiments tested the functions of iPSCs-derived IPCs on day 21 of the second step. a Flow cytometry plots illustrating the protein expression of insulin in populations of iPSCs-derived IPCs. Black text indicates the percentage of insulin. b Glucose-stimulated insulin secretion in vitro. iPSCs-derived IPCs on day 21 of the third step were exposed to different glucose concentrations (0, 5, 15, and 45mM). The insulin concentration levels were determined. c Quantitative RT-PCR analysis of the mRNA expression levels of key endocrine markers (insulin1, insulin2, GCG, SST, GCK, Mafa, ISL, Glut2). GAPDH was used as the internal control. Error bars show meanstandard deviation (SD) (n=3). *P<0.05, **P<0.01, ***P<0.001. d Immunofluorescence assays of protein expression levels of some key markers. Co-immunostaining of insulin (red) with C-peptide (green), insulin (red) with Pdx1 (green), insulin with Nkx6.1 (green); nuclear DAPI staining is shown in blue. Scale bar 75m

To further dissect the molecular mechanism of the inhibitory effect of miR-690 on IPCs differentiation, we performed the bioinformatics prediction analysis by using TargetScan and miRanda and combined with the results from RNA-seq (Huang, et al.) to predict the target genes of DEmiRNAs. miR-690 has 15 putative target genes (Prkca, Nedd4l, Ulk2, Prkcz, Csnk1g1, Mllt3, Enah, Pcgf3, Impa1, Stat3, Grm5, Cnot6, Sox9, Wasl, and Ctnnb1). Then, we built a regulatory network to show the connections between DEmiRNAs and target genes (Fig.

c). Among these predicted genes,

, a marker of pancreatic progenitor cells, and the genes encoding key transcription factors for the development of -cells were notable. Next, we performed a dual-luciferase reporter assay to experimentally determine whether miR-690 targeted Sox9 directly. We transfected HEK293T cells with a luciferase plasmid containing the wild-type 3 UTRs of Sox9 or its mutant version downstream of the firefly luciferase cDNA in the pEZX-FR02 vector (Fig.

a). The results showed that the co-transfection of miR-690 mimics into 293T cells led to a decrease of up to 83% in luciferase activity by miR-690 but had nearly no effect on the mutant reporter activity (Fig.

b). Furthermore, knockdown of miR-690 reversed the repressive effects of siRNA-Sox9 on the mRNA and protein levels of Sox9 (Fig.

ce). These findings indicated that Sox9 was the authentic target of miR-690 in our induced IPCs.

miR-690 directly targeted Sox9 in iPSCs-derived IPCs. a Predicted miR-690 targeting sequence in the 3UTR of Sox9 (Sox9 WT-3UTR) and the mutant form of the Sox9 3UTR (Sox9 MUT-3UTR). b Dual-luciferase reporter assays to determine the influence of miR-690 on Sox9 3UTR activity in iPSCs-derived IPCs. Data are the meanSD of three independent assays. ce Quantitative RT-PCR and Western blotting analyses of the effects of miR-690 knockdown (miR-690 inhibitor) on the expression level of Sox9 and the effects of miR-690 knockdown (miR-690) on the repressive effects of Sox9 knockdown (Sox9 siRNA). -Actin was used as the loading control. GAPDH was used as the internal control for mRNA. Error bars show the SD (n=3)

Sox9 has been reported to play a role in regulating Wnt signaling, which influences pancreatic development and modulates mature -cell functions, such as insulin secretion, survival, and proliferation. Sox9 was chosen for further analysis in our study and validated by both qPCR analysis at the mRNA level and Western blot and immunostaining assays at the protein level (Fig.

ac and h). Because the phosphorylation and inactivation of GSK3- may lead to activation and nuclear translocation of -catenin, we detected the level of GSK3- phosphorylation when miR-690 was overexpressed. As expected, a more than 1.5-fold decrease in phosphorylated GSK3- and a more than 2-fold decrease in -catenin activity were observed (Fig.

dh). We speculated that in our induced models, miR-690 may inactivate the WNT signaling pathway through Sox9 which will be the focus of our future research (Fig.

i).

miR-690 may affect the differentiation of IPCs by inactivating the expression of the Wnt signaling pathway. a Quantitative RT-PCR analysis of the expression levels Sox9 and -catenin. The scale bar represents 100m. Western blotting analysis of the effects of miR-690 overexpression on Sox9 (b, c), p-GSK3 (phosphorylated-GSK3) (d, e), and -catenin (f, g) (690-OE means 690 overexpression/miR-690 agomir). -Actin was used as the loading control. h Immunofluorescence assay (Sox9 and -catenin, red; nuclei, blue; scale bar 75m) of the protein level of Sox9 and -catenin. i Schematic diagram of the supposed role of miR-690 in iPSCs-derived IPCs differentiation.

We next sought to explore whether miR-690 could modulate glucose homeostasis by transplanting miR-690-overexpressing IPCs and negative control cells into anemic capsule kidneys of mice treated with streptozotocin (STZ), which specifically destroys mouse -cells (Fig.

a). After transplantation, populations from the NC group needed nearly 28days to reverse the hyperglycemia. Although the blood glucose level was decreased, mice transplanted with the miR-690 agomir still showed glycemia (Fig.

b). Not surprisingly, the body weight of transplanted mice in the miR-690 overexpression group was significantly lower than that of the control group and healthy mice (data has not shown). At 40days post-transplant, excised iPSCs-derived IPCs grafts were highly compact and homogenous and did not have regions of expanded ducts (Fig.

c). Immunofluorescence staining revealed insulin-positive clusters of cells in the graft surrounded by connective tissue producing endocrine hormones (Fig.

d).

iPSCs-derived IPCs reverse diabetes in vivo. a Image of the entire kidney with iPSCs-derived IPCs engrafted under the kidney capsule and harvested at 25days post-transplant. (~1106 cells/mouse, n=6 /group). b Blood glucose levels were measured pre- and post-transplantation for over 30days. c Hematoxylin and eosin (H&E) staining image of iPSCs-derived IPCs grafts in the kidney capsule 25days after transplantation. Scale bar 200m. d Immunofluorescence staining of whole grafts for insulin (red); nuclear DAPI staining is shown in blue. Scale bar 75m

iPSCs, which are derived from somatic cells, allow for the patient-specific functional -cells in vitro that will free diabetic populations from daily insulin injections and prevent life-threatening complications, generate sufficient -cells for transplantation, and also avert immune suppression to repress auto- and allo-immunity [1, 19]. Although many attempts have been made to acquire mature, glucose-responsive IPCs entirely in vitro, the results of these studies lacked convincing evidence [19]. Multiple core transcription factors, signaling pathways, and noncoding RNAs have been confirmed to be required for pancreatic -cells differentiation potential in potent stem cells [10, 2024]. Increasing evidence shows that miRNAs, as important epigenetic factors that regulate gene expression and determine cell fate in pancreatic -cells, mediate -cells biological activities, including differentiation, proliferation, apoptosis, and insulin secretion [6, 25]. However, the mechanisms of miRNAs in -cells differentiation of iPSCs remain unknown.

This study adopted a three-step protocol mimicking normal pancreatic formation to screen for differentiation-associated miRNAs during iPSCs-induced IPCs differentiation in culture. According to the miRNA array analysis data, 13 miRNAs with markedly different expression levels were identified (Additionalfile1: Figure S1), and we found that miR-690 was significantly upregulated in step 3 compared to that in iPSCs. To explore the specific function of miR-690 in IPCs differentiation, we overexpressed miR-690 in progenitor cells on day 4 of step 3 and found that pancreatic progenitor markers, such as Pdx1 and Sox9, and the early endocrine progenitors NGN3, Nkx6.1, and Pax4 were downregulated after 48h. At the final stage of our protocol, miR-690 overexpression significantly impaired the maturation and endocrine function of IPCs (Fig.3). However, the mRNA level of SST increased unexpectedly after miR-690 overexpression, suggesting that this miRNA may promote the differentiation of -cells.

To elaborate on the mechanism by which miR-690 regulates IPCs formation, we used bioinformatic analysis. Combined with the RNA-seq data detected previously, these results identified Sox9 as an underlying target gene of miR-690. Sox9 is widely known as a pancreatic progenitor marker that influences endocrine pancreatic development and modulation of mature -cells functions [14]. The prevailing theory is that miRNAs regulate gene expression post-transcriptionally by binding to the 3 untranslated sequence of the targeted mRNA to silence its corresponding target genes [26, 27]. Then, we demonstrated that Sox9 was a direct target of miR-690 using a luciferase reporter assay (Fig.5). Furthermore, overexpression of miR-690 decreased the protein levels of Sox9 and -catenin (Fig.6), indicating that this noncoding RNA may regulate the Wnt signaling pathway, which has been thoroughly investigated and is necessary for controlling the development of -cells and their function [16, 28, 29]. These findings suggested that the important function of miR-690 during IPCs differentiation was predominantly regulated by the miR-690/Sox9 and -catenin axes, confirming that the interactions of miRNAs and transcription factors were involved in the differentiation of mouse iPSCs to IPCs. -catenin is an important effector of the Wnt pathway [30]. To date, the role of Wnt signaling in pancreatic development has been disputed. The majority of studies have noted the primary role of Wnt signaling in the development of the exocrine compartments of the pancreas and confirmed that abolishment of the signaling pathway resulted in an almost complete lack of exocrine cells; however, the influence of Wnt signaling on endocrine cells, especially on pancreatic -cell development, is still undefined [31]. Previous studies have reported that knockdown of the Sox9 gene in human islet epithelial cells significantly decreases the expression of phosphorylated GSK3- at the protein level, leading to a prominent decline in the expression of cyclin D1 and other target genes of the Wnt signaling pathway [14]. Therefore, we examined the Wnt signaling activity by detecting the expression of p-GSK3-. Interestingly, the results showed that miR-690 overexpression simultaneously decreased Sox9 and phosphorylated GSK3- at the protein level. We speculated that miR-690 may mediate the Wnt signaling pathway via binding to Sox9 and lead to a decline in phosphorylation of GSK3- and a decrease in -catenin, which are the effectors of this pathway. Furthermore, other researchers have shown that pancreatic -cells differentiation is complex and a result of the interaction of multiple signaling pathways, such as Notch, Fgf, Wnt, and others. Thus, the specific regulatory mechanism between miR-690 and the Wnt signaling pathway and whether other signaling pathways are regulated by miR-690 require further exploration.

Recently, miR-690 was reported to mediate osteogenic differentiation of human myogenic progenitor cells through its target NF-kappaB p65, indicating that miR-690 may play different roles in the development and differentiation of different organs and tissues [32]. Many studies have shown that Sox9 downregulation is important for early lineage bifurcation of endocrine progenitors and pancreatic -cells development [15, 3336]. In our study, the expression of miR-690 at an appropriate level is vital to the maturation and differentiation of IPCs. However, prematurely upregulated Sox9 resulted in deficient IPC differentiation in vitro, indicating that miR-690 activity may need to be within a narrow range to avoid detrimental consequences. Therefore, further exploration of the function of the miR-690/Sox9/Wnt signaling pathway in pancreatic -cells differentiation, development, and maturation may be required to systematically uncover the critical function and mechanism of miR-690 in vitro and in vivo.

We found that miR-690, a rarely studied noncoding RNA, played an important role in the differentiation of iPSCs-derived IPCs. MiR-690 regulates the expression of transcription factor Sox9 and may have an influence on Wnt signaling pathway in the differentiation process. These findings not only indicate that miR-690 mediates differentiation of iPSCs-derived IPCs through Sox9 and affects Wnt signaling pathway, but also provide novel evidence for the regulatory potential mechanisms of miRNAs in development associated with insulin-producing cells derived from induced pluripotent cells.

Yang Xu, Yan Huang and Yibing Guo contributed equally to this work.

Differentially expressed miRNAs

Embryoid Bodies

GATA binding protein 4

GATA binding protein 6

Glucagon

Glucokinase

Facilitated glucose transporter, member 2

Gene ontology

Hematoxylin and eosin

Insulin-producing cell

Induced pluripotent stem cell

ISL LIM homeobox

Kyoto Encyclopedia of Genes and Genomes

v-maf musculoaponeurotic fibrosarcoma oncogene family, protein A

MicroRNA

Multilineage precursor stem cell

Negative control

Neurogenin 3

NK2 homeobox 2

NK6 homeobox 1

Paired box 4

Paired box 6

Pancreatic and duodenal homeobox 1

Phosphorylated glycogen synthase kinase-3

Real-time quantitative polymerase chain reaction

Original post:
microRNA-690 regulates induced pluripotent stem cells (iPSCs ...

Induced pluripotent stem cells have been generated for the …

Induced pluripotent stem cells have been generated for the first time from tumor cells in order to study therapies for tumors developed in patients with hereditary diseases with predisposition to cancer

The Hereditary Cancer Research Group at the Germans Trias i Pujol Research Institute (IGTP) on the Can Ruti Campus, Badalona has for the first time generated induced pluripotent stem cells (iPSCs) from tumors from people with the hereditary disease Neurofibromatosis type 1 (NF1). The work has been carried out in conjunction with Angel Raya of the Centre for Regenerative Medicine of Barcelona (CMRB) and published in Stem Cell Reports, the official journal for the International Society for Stem Cell Research (ISSCR).

iPSCs are stem cells capable of giving rise to most other types of cell in the body. It is quite normal to generate stem cells by reprogramming skin cells extracted from patients with hereditary diseases such as NF1 to study them as a model for the disease, but the IGTP researchers have now generated iPSCs for the first time as a valid model for NF1 from cells from tumors. Instead of using skin cells, we have reprogrammed cells from tumors from patients with NF1 in order to have a model of cells genetically identical to the tumor cells, explains Eduard Serra, leader of the work at the IGTP.

One of the difficulties of studying these pathologies with cells obtained directly from benign tumors is that they are finite, but now we have achieved a cellular model which will not run out because, due to their characteristics, we can culture these iPSCs indefinitely and then convert them into the same cells that make up a tumor, Serra adds. The work has invested most of the efforts in converting these iPSCs into Schwann cells, which are cells which make up plexiform neurofibromas, typical of NF1. The resulting cells have the same capacity to proliferate as the original tumor cells. As iPSCs are an endless source of cells, we have been able to generate the resource we needed. Now we can test drugs that inhibit proliferation, study the mechanisms by which these tumors develop and try to stop them developing into malignant tumors, explains Meritxell Carri, first author of the article.

New ways to study NF1

The Hereditary Cancer Research Group at the IGTP has been studying Neurofibromatoses for many years, this includes NF1, a minority disease that affects 1 in every 3,000 people in the world. It is a hereditary disease, which brings a high predisposition to develop tumors in the peripheral nervous system. One of these types of tumor is the plexiform neurofibromas. It is a tumor that appears at birth, or in the first years of life, and forms on major nerves in the body, disorganizing and thickening the tissue surrounding the nerve and forming a tumor mass that can reach great dimensions. These tumors can impede functionality and disfigure the part of the body where they appear, which can be on the extremities or even on the face. Additionally, there is a risk that these tumors progress to become malignant; a sarcoma of the peripheral nerve sheath.

Although they start as benign tumors, we do not have drugs to put the patient into remission. To develop effective drugs for this type of tumor we need cellular models that are faithful copies of the tumor and that do not run out, this is the model that now has been achieved.

Available to the scientific community

The cell lines generated have been deposited in the Carlos III Stem Cell Bank at the node kept at the CMRB in Barcelona. They are available to other researchers in the world who want to study this disease and these tumors.

This work was led by Eduard Serra at the IGTP and carried out in collaboration with Angel Raya at the CMRB alongside other participating institutions such as the Catalan Institute of Oncology (ICO), Sage Bionetworks, and the Germans Trias Hospital. This project has been financed by the Neurofibromatosis Therapuetic Acceleration Program (NTAP, http://www.n-tap.org), a program based at the Johns Hopkins University School of Medicine in Baltimore, Maryland in the United States, whose mission is to accelerate the development of therapies for Neurofibromatosis Type 1. The team has also had the support of the Spanish and Catalan Associations of Neurofibromatosis Patients.

The Johns Hopkins University, and the Germans Trias i Pujol Research Institute (IGTP) (http://www.germanstrias.org/) are academic institutions based respectively in Baltimore, Maryland, and Badalona (Barcelona), Spain.

The Center of Regenerative Medicine in Barcelona (CMR[B]) (www.cmrb.eu) is a public research centre (non-profit foundation) based in Barcelona, Spain. The overall mission of the CMR[B] is to conduct fundamental research of excellence forthe advancement inthe clinical translation of regenerative medicine strategies basedin pluripotent stem cells, mainly in the context of heart failure, neurodegenerative diseases, non-malignant hematological diseases and age-related macular degeneration.

Sage Bionetworks is a nonprofitbiomedical research and technology organization. We develop and apply open practices to data-driven research for the advancement of human health. Our interdisciplinary team of scientists and engineers work together to provide researchers access to technology tools and scientific approaches to share data, benchmark methods, and explore collective insights, all backed by Sages gold-standard governance protocols and commitment to user-centered design.Sage, founded in Seattle in 2009, is supported through a portfolio of competitive research grants, commercial partnerships, and philanthropic contributions. Learn more atwww.sagebionetworks.org.

Read more from the original source:
Induced pluripotent stem cells have been generated for the ...

Induction of Human Embryonic and Induced Pluripotent Stem …

Public Summary:

The authors evaluated different methods to induce human embryonic stem cells and induced pluripotent stem cells into urothelial cells, which are specialized cells that line the inside of the bladder, ureters and kidneys. They also evaluated regulatory events that occur during the differentiation of these cells. This technology shows the feasibility of producing urologic tissue from stem cells for bioengineering purposes.

Scientific Abstract:

In vitro generation of human urothelium from stem cells would be a major advancement in the regenerative medicine field, providing alternate nonurologic and/or nonautologous tissue sources for bladder grafts. Such a model would also help decipher the mechanisms of urothelial differentiation and would facilitate investigation of deviated differentiation of normal progenitors into urothelial cancer stem cells, perhaps elucidating areas of intervention for improved treatments. Thus far, in vitro derivation of urothelium from human embryonic stem cells (hESCs) or human induced pluripotent stem (hiPS) cells has not been reported. The goal of this work was to develop an efficient in vitro protocol for the induction of hESCs into urothelium through an intermediary definitive endoderm step and free of matrices and cell contact. During directed differentiation in a urothelial-specific medium ("Uromedium"), hESCs produced up to 60% urothelium, as determined by uroplakin expression; subsequent propagation selected for 90% urothelium. Alteration of the epithelial and mesenchymal cell signaling contribution through noncell contact coculture or conditioned media did not enhance the production of urothelium. Temporospatial evaluation of transcription factors known to be involved in urothelial specification showed association of IRF1, GET1, and GATA4 with uroplakin expression. Additional hESC and hiPS cell lines could also be induced into urothelium using this in vitro system. These results demonstrate that derivation and propagation of urothelium from hESCs and hiPS cells can be efficiently accomplished in vitro in the absence of matrices, cell contact, or adult cell signaling and that the induction process appears to mimic normal differentiation.

See the original post:
Induction of Human Embryonic and Induced Pluripotent Stem ...

Stem Cell Knee Therapy Cost | Stem Cell Knee Injection …

Knee pain affects millions of Americans and can become so severe that the pain limits sporting activities, work productivity and daily activities. When a patients ability to live happily is hindered by severe, chronic knee pain, painful knee surgery or knee replacement surgery may be recommended by a physician. A stem cell knee injection treatment is now available to patients who want to postpone knee surgery. In certain patients, stem cell knee therapy cost is more beneficial than a surgery and rehabilitation program. OPTIMAL Pain & Regenerative Medicines board-certified physicians are leading providers in the Dallas, Arlington and Fort Worth, Texas area in treating knee pain with autologous stem cell therapy techniques.

An autologous stem cell knee injection is a non-invasive knee treatment used to treat chronic pain in patients suffering from ligament injuries, tendon injuries, fractures and degenerative joint disorders, such as osteoarthritis. Autologous stem cells are harvested from a patients own body and have the natural ability to help a damaged area heal. Since the stem cells used for stem cell knee injections are extracted directly from a patients hip area, patients do not experience risk factors like the ones present with the use of highly controversial embryonic stem cells.

A stem cell knee injection requires a sample of bone marrow to be removed from the patients own body. After the bone marrow is harvested, the sample is spun in a special machine called a centrifuge to separate a combination of pluripotent stem cells, platelets and white blood cells. The combination is then injected into the damaged knee area to regenerate and promote healing.

When traditional knee repair options, such as weight loss, pain medication and physical therapy, do not alleviate chronic knee pain, some physicians may recommend a knee surgery or knee replacement surgery. Surgery is not always a viable option for all patients. In these cases, a stem cell knee injection may prove to be a beneficial treatment option.

Some patients may be concerned about stem cell knee therapy cost and effectiveness. The physicians at OPTIMAL Pain & Regenerative Medicine are highly trained and experienced at stem cell knee injections. Patients do not need to worry about getting a treatment that is not needed or beneficial to his/hers healing process.

Our physicians follow a strict protocol to ensure a patient is a good candidate and will benefit from the autologous stem cell injection therapy. These protocols include:

When these protocols are followed, stem cell knee injections are highly effective in many patients. The postponement of a painful knee surgery and long rehabilitation process is often a key benefit of autologous stem cell therapy and offsets stem cell knee therapy cost.

For more resources on stem cell knee injections, or stem cell knee therapy cost, please contact OPTIMAL Pain & Regenerative Medicine, located in the Dallas, Arlington and Fort Worth, Texas area.

Read this article:
Stem Cell Knee Therapy Cost | Stem Cell Knee Injection ...

Stem Cell Transplantation | Leukemia and Lymphoma Society

Stem cell transplantation, sometimes referred to as bone marrow transplant, is a procedure that replaces unhealthy blood-forming cells with healthy cells. Stem cell transplantation allows doctors to give large doses of chemotherapy or radiation therapy to increase the chance of eliminating blood cancer in the marrow and then restoring normal blood cell production. Researchers continue to improve stem cell transplantation procedures, making them an option for more patients.

The basis for stem cell transplantation is that blood cells (red cells, white cells and platelets) and immune cells (lymphocytes) arise from the stem cells, which are present in marrow, peripheral blood and cord blood. Intense chemotherapy or radiation therapy kills the patient's stem cells. This stops the stem cells from making enough blood and immune cells.

The patient receives high-dose chemotherapy and/or radiation therapy, followed by the stem cell transplant. A donor's stem cells are then transfused into the patient's blood. The transplanted stem cells go from the patient's blood to his or her marrow.

The donor is usually a brother or a sister if one is available and if he or she is a match for the patient. Otherwise, an unrelated person with stem cells that match the patient's tissue type can be used. These matched unrelated donors (MUDs) can be found through stem cell donor banks or registries.

The new cells grow and provide a supply of red cells, white cells (including immune cells) and platelets. The donated stem cells make immune cells that are not totally matched with the patient's cells. (Patients and donors are matched to major tissue types but not minor tissue types.) For this reason, the donor immune cells may recognize the patient's cancer cells' minor tissue types as foreign and kill the cancer cells. This is called "graft versus cancer effect."

If you're a candidate for a stem cell transplant, your doctor will usually recommend one of three types:

A fourth type of stem cell transplantation, syngeneic transplantation, is much less common than the other three. Syngeneic transplantation is rare for the simple reason that it's only used on identical twins. In addition, the donor twin and the recipient twin must have identical genetic makeup and tissue type.

Your doctor considers several factors when deciding whether you're a candidate for stem cell transplantation. For allogeneic stem cell transplantation, your doctor takes into account:

When considering whether you're a candidate for an autologous stem cell transplantation, your doctor takes into account:

Allogeneic stem cell transplant is more successful in younger patients than older patients. About three-quarters of the people who develop a blood cancer are older than 50. In general, older individuals are more likely to:

However, the above factors are generalizations, and there's no specific age cutoff for stem cell transplantation.

Other factors and the response of the underlying disease to initial cancer therapy determine when your doctor considers transplant options. Some patients undergo stem cell transplantation in first remission. For other patients, it's recommended later in the course of treatment for relapsed or refractory disease.

Before you undergo stem cell transplantation, you'll need pretreatment, also called conditioning treatment. You'll be given high-dose chemotherapy or radiation therapy to:

Pretreatment for a reduced-intensity allogeneic stem cell transplant involves lower dosages of chemotherapy drugs or radiation than for a standard allogeneic stem cell transplant.

Donor stem cells are transferred to patients by infusion, a procedure similar to a blood transfusion. Blood is delivered through a catheter a thin flexible tube into a large blood vessel, usually in your chest.

Infusing the stem cells usually takes several hours. You'll be checked frequently for signs of fever, chills, hives, a drop in blood pressure or shortness of breath. You may experience side effects such as headache, nausea, flushing and shortness of breath from the cryopreservative used to freeze the stem cells. If so, you'll be treated and then continue infusion.

Read more:
Stem Cell Transplantation | Leukemia and Lymphoma Society

Oregon Regenerative Medicine – Advanced Joint and Injury …

Oregon Regenerative Medicine is dedicated to empowering our clients to create healthy, active longevity through the use of PRP and Stem Cell joint regeneration, Prolotherapy, Platelet Rich Plasma PRP, condition-specific nutritional programs, hormone optimization, education, and inspiration. We live and practice what we preach. We are the experts in Regenerative Medicinein the Portland and Lake Oswego region. Since 1978, we have pioneered the use of safe, effective natural medicine in Oregon.

Regenerative Medicine includes the use of non-surgical injection procedures for the permanent repair of damaged tendons, ligaments, joints and skin. Our regenerative and biological treatments include Prolotherapy, Platelet Rich Plasma PRP, and Adult Stem Cell Therapies. These treatments enhance the natural cycles of repair in aging and chronically injured joints, ligaments, tendons, and skin. Regenerative orthopedic injections are an effective treatment for all manner of acute and chronic pain in any joint. We specialize in back and neck injuries,as well as osteoarthritis and injuries of thehip, knee, shoulder, elbow, wrist, hand, TMJ, foot and ankle. For our patients who have been told that their only solution is surgery or a lifetime on pain medications, the vast majority have been able to achieve drug-free, pain-free function without surgery or joint replacement.

At Oregon Regenerative Medicine, we useAdipose-Derived Stem Cell Therapy to treat a wide range of orthopedic and degenerative diseases, including inflammatory and rheumatoid arthritis. We use adult stem cells that are harvested from your own adipose tissue. Unlike embryonic stem cells, adult stem cells are approved by the FDA for research and treatment of a wide variety of conditions. Adult adipose tissue is the most abundant source of stem cells in the human body and has shown great promise in the treatment of a host of conditions.

Your skin is a living, breathing organ that reflects your overall health. Our holistic dermatology doctors provide individualized skin care for all ages. We specialize in the special needs of aging skin. We seek the underlying cause of skin disorders and consider the metabolic, hormonal, nutritional and emotional factors that are essential to creating healthy skin. We tailor your treatment to your specific needs, using restorative naturopathic medical therapies along with a full complement of aesthetic and regenerative skin treatments. We are experts in Collagen Induction Therapy, Derma Pen, Live Stem Cell Fat Transfer and PRP Facelifts.

We go beyond the treatment of damaged joints. We have been practicing holistic and functional medicine in Lake Oswego since 1978. Our goal is to address the underlying causes of illness and restore normal function. We know that nutrition is the foundation for healing and that condition-specific diets enhance regeneration of tissue. We assess each individual beginning with a detailed history, a thorough physical exam and appropriate lab testing to gain a clear understanding of current health issues. We then create treatment plans and options that are tailored to each individual. Our treatments are evidence-based and proven effective, both by modern science and traditional healing wisdom. Our holistic approach to health respects the vis medicatrix naturae, the healing power of nature. Therapies may include botanical medicine, specific vitamin and nutrient therapy, acupuncture, I.V. Therapy, bio-identical hormone replacement therapy, homeopathy, prolotherapy, PRP therapies, Adult Stem Cell injections, myofascial release and more.

View post:
Oregon Regenerative Medicine - Advanced Joint and Injury ...

Bone marrow transplant – Mayo Clinic

Overview

A bone marrow transplant is a procedure that infuses healthy blood stem cells into your body to replace your damaged or diseased bone marrow. A bone marrow transplant is also called a stem cell transplant.

A bone marrow transplant may be necessary if your bone marrow stops working and doesn't produce enough healthy blood cells.

Bone marrow transplants may use cells from your own body (autologous transplant) or from a donor (allogeneic transplant).

At Mayo Clinic, doctors who specialize in blood diseases (hematologists) form a multidisciplinary team with other experts to provide personalized, whole-person care to adults and children undergoing bone marrow transplants.

Your transplant team may include hematologists, cancer specialists (oncologists), mental health specialists (psychologists and psychiatrists), a bone marrow transplant scheduling coordinator, transfusion medicine nurses, trained and specialized nurses, physician assistants, social workers, a nurse coordinator, a clinical nurse specialist, a dietitian, pharmacists, a chaplain and a child life specialist for children undergoing bone marrow transplant.

Children and adolescents undergoing bone marrow transplants receive care at the Children's Center at Mayo Clinic's campus in Minnesota. At Mayo Clinic's campus in Arizona, pediatric experts collaborate with the Phoenix Children's Hospital to provide care to young patients. Together the two oversee a single bone marrow transplant program for children. Pediatric patients receive care from Mayo Clinic specialists in Florida through a partnership with Nemours Children's Specialty Care and Wolfson Children's Hospital.

Mayo Clinic's approach

A bone marrow transplant may be used to:

Bone marrow transplants can benefit people with a variety of both cancerous (malignant) and noncancerous (benign) diseases, including:

Bone marrow is the spongy tissue inside some bones. Its job is to produce blood cells. If your bone marrow isn't functioning properly because of cancer or another disease, you may receive a stem cell transplant.

To prepare for a stem cell transplant, you receive chemotherapy to kill the diseased cells and malfunctioning bone marrow. Then, transplanted blood stem cells are put into your bloodstream. The transplanted stem cells find their way to your marrow, where ideally they begin producing new, healthy blood cells.

Mayo Clinic doctors have extensive experience performing bone marrow transplants for adults and children with a variety of cancerous and noncancerous diseases. Each year, more than 700 people undergo bone marrow transplants at Mayo Clinic.

The first bone marrow transplant at Mayo Clinic occurred in 1963. Bone marrow transplant procedures are performed by doctors at Mayo Clinic in Rochester, Minn., Mayo Clinic in Jacksonville, Fla., and Mayo Clinic in Scottsdale, Ariz., which are ranked among the Best Hospitals for cancer by U.S. News & World Report.

The long history of bone marrow transplants performed at Mayo Clinic means that doctors are prepared with the knowledge and resources to provide you with expert, personalized care.

A bone marrow transplant poses many risks of complications, some potentially fatal.

The risk can depend on many factors, including the type of disease or condition, the type of transplant, and the age and health of the person receiving the transplant.

Although some people experience minimal problems with a bone marrow transplant, others may develop complications that may require treatment or hospitalization. Some complications could even be life-threatening.

Complications that can arise with a bone marrow transplant include:

Your doctor can explain your risk of complications from a bone marrow transplant. Together you can weigh the risks and benefits to decide whether a bone marrow transplant is right for you.

If you receive a transplant that uses stem cells from a donor (allogeneic transplant), you may be at risk of developing graft-versus-host disease (GVHD). This condition occurs when the donor stem cells that make up your new immune system see your body's tissues and organs as something foreign and attack them.

Many people who have an allogeneic transplant get GVHD at some point. The risk of GVHD is a bit greater if the stem cells come from an unrelated donor, but it can happen to anyone who gets a bone marrow transplant from a donor.

GVHD may happen at any time after your transplant. However, it's more common after your bone marrow has started to make healthy cells.

There are two kinds of GVHD: acute and chronic. Acute GVHD usually happens earlier, during the first months after your transplant. It typically affects your skin, digestive tract or liver. Chronic GVHD typically develops later and can affect many organs.

Chronic GVHD signs and symptoms include:

You'll undergo a series of tests and procedures to assess your general health and the status of your condition, and to ensure that you're physically prepared for the transplant. The evaluation may take several days or more.

In addition, a surgeon or radiologist will implant a long thin tube (intravenous catheter) into a large vein in your chest or neck. The catheter, often called a central line, usually remains in place for the duration of your treatment. Your transplant team will use the central line to infuse the transplanted stem cells and other medications and blood products into your body.

If a transplant using your own stem cells (autologous transplant) is planned, you'll undergo a procedure called apheresis (af-uh-REE-sis) to collect blood stem cells.

Before apheresis, you'll receive daily injections of growth factor to increase stem cell production and move stem cells into your circulating blood so that they can be collected.

During apheresis, blood is drawn from a vein and circulated through a machine. The machine separates your blood into different parts, including stem cells. These stem cells are collected and frozen for future use in the transplant. The remaining blood is returned to your body.

If a transplant using stem cells from a donor (allogeneic transplant) is planned, you will need a donor. When you have a donor, stem cells are gathered from that person for the transplant. This process is often called a stem cell harvest or bone marrow harvest. Stem cells can come from your donor's blood or bone marrow. Your transplant team decides which is better for you based on your situation.

Another type of allogeneic transplant uses stem cells from the blood of umbilical cords (cord blood transplant). Mothers can choose to donate umbilical cords after their babies' births. The blood from these cords is frozen and stored in a cord blood bank until needed for a bone marrow transplant.

After you complete your pretransplant tests and procedures, you begin a process known as conditioning. During conditioning, you'll undergo chemotherapy and possibly radiation to:

The type of conditioning process you receive depends on a number of factors, including your disease, overall health and the type of transplant planned. You may have both chemotherapy and radiation or just one of these treatments as part of your conditioning treatment.

Side effects of the conditioning process can include:

You may be able to take medications or other measures to reduce such side effects.

Based on your age and health history, your doctor may recommend lower doses or different types of chemotherapy or radiation for your conditioning treatment. This is called reduced-intensity conditioning.

Reduced-intensity conditioning kills some cancer cells and somewhat suppresses your immune system. Then, the donor's cells are infused into your body. Donor cells replace cells in your bone marrow over time. Immune factors in the donor cells may then fight your cancer cells.

Your bone marrow transplant occurs after you complete the conditioning process. On the day of your transplant, called day zero, stem cells are infused into your body through your central line.

The transplant infusion is painless. You are awake during the procedure.

The transplanted stem cells make their way to your bone marrow, where they begin creating new blood cells. It can take a few weeks for new blood cells to be produced and for your blood counts to begin recovering.

Bone marrow or blood stem cells that have been frozen and thawed contain a preservative that protects the cells. Just before the transplant, you may receive medications to reduce the side effects the preservative may cause. You'll also likely be given IV fluids (hydration) before and after your transplant to help rid your body of the preservative.

Side effects of the preservative may include:

Not everyone experiences side effects from the preservative, and for some people those side effects are minimal.

When the new stem cells enter your body, they begin to travel through your body and to your bone marrow. In time, they multiply and begin to make new, healthy blood cells. This is called engraftment. It usually takes several weeks before the number of blood cells in your body starts to return to normal. In some people, it may take longer.

In the days and weeks after your bone marrow transplant, you'll have blood tests and other tests to monitor your condition. You may need medicine to manage complications, such as nausea and diarrhea.

After your bone marrow transplant, you'll remain under close medical care. If you're experiencing infections or other complications, you may need to stay in the hospital for several days or sometimes longer. Depending on the type of transplant and the risk of complications, you'll need to remain near the hospital for several weeks to months to allow close monitoring.

You may also need periodic transfusions of red blood cells and platelets until your bone marrow begins producing enough of those cells on its own.

You may be at greater risk of infections or other complications for months to years after your transplant.

A bone marrow transplant can cure some diseases and put others into remission. Goals of a bone marrow transplant depend on your individual situation, but usually include controlling or curing your disease, extending your life, and improving your quality of life.

Some people complete bone marrow transplantation with few side effects and complications. Others experience numerous challenging problems, both short and long term. The severity of side effects and the success of the transplant vary from person to person and sometimes can be difficult to predict before the transplant.

It can be discouraging if significant challenges arise during the transplant process. However, it is sometimes helpful to remember that there are many survivors who also experienced some very difficult days during the transplant process but ultimately had successful transplants and have returned to normal activities with a good quality of life.

Explore Mayo Clinic studies testing new treatments, interventions and tests as a means to prevent, detect, treat or manage this disease.

Mayo Clinic doctors and scientists are involved in cutting-edge research that allows them to apply the latest advances to patient care.

Innovations include:

At Mayo Clinic, some bone marrow transplants are performed as hospital-based outpatient procedures, which reduces the amount of time you'll need to spend in the hospital.

Living with a bone marrow transplant or waiting for a bone marrow transplant can be difficult, and it's normal to have fears and concerns.

Having support from your friends and family can be helpful. Also, you and your family may benefit from joining a support group of people who understand what you're going through and who can provide support. Support groups offer a place for you and your family to share fears, concerns, difficulties and successes with people who have had similar experiences. You may meet people who have already had a transplant or who are waiting for a transplant.

To learn about transplant support groups in your community, ask your transplant team or social worker for information. Also, several support groups are offered at Mayo Clinic in Arizona, Florida and Minnesota.

Mayo Clinic researchers study medications and treatments for people who have had bone marrow transplants, including new medications to help you stay healthy after your bone marrow transplant.

If your bone marrow transplant is using stem cells from a donor (allogeneic transplant), you may be at risk of graft-versus-host disease. This condition occurs when a donor's transplanted stem cells attack the recipient's body. Doctors may prescribe medications to help prevent graft-versus-host disease and reduce your immune system's reaction (immunosuppressive medications).

After your transplant, it will take time for your immune system to recover. You may be given antibiotics to prevent infections. You may also be prescribed antifungal, antibacterial or antiviral medications. Doctors continue to study and develop several new medications, including new antifungal medications, antibacterial medications, antiviral medications and immunosuppressive medications.

After your bone marrow transplant, you may need to adjust your diet to stay healthy and to prevent excessive weight gain. Maintaining a healthy weight can help prevent high blood pressure, high cholesterol and other negative health effects.

Your nutrition specialist (dietitian) and other members of your transplant team will work with you to create a healthy-eating plan that meets your needs and complements your lifestyle. Your dietitian may also give you food suggestions to control side effects of chemotherapy and radiation, such as nausea.

Your dietitian will also provide you with healthy food options and ideas to use in your eating plan. Your dietitian's recommendations may include:

After your bone marrow transplant, you may make exercise and physical activity a regular part of your life to continue to improve your health and fitness. Exercising regularly helps you control your weight, strengthen your bones, increase your endurance, strengthen your muscles and keep your heart healthy.

Your treatment team may work with you to set up a routine exercise program to meet your needs. You may perform exercises daily, such as walking and other activities. As you recover, you can slowly increase your physical activity.

Jan. 24, 2019

Read more:
Bone marrow transplant - Mayo Clinic

Stem Cell Therapy Sarasota | Advanced Rejuventation

Welcome to Advanced Rejuvenation

At our state of the art clinic we are using cutting edge regenerative techniques to improve the health and well being of our patients. At Advanced Rejuvenation, we provide care for people suffering from diseases that may have limited treatment options and may respond to alternative and cell based regenerative treatments. Advanced Rejuvenation emphasizes quality and our well trained physicians are highly committed to the advancement of regenerative medicine.

Dr. Jeff Sack is a board certified cardiologist who is specializing in conservative Regenerative Cardiology using Hyperbaric Oxygen, Ozone Therapy, NAD, Vitamin infusions, Chelation Therapy, Lipid Exchange, PK Protocol, Peptide Therapy, Stem Cells, Exosomes and Lifestyle / Nutritional Programs. These modalities are designed to stimulate the cardio and vascular system to repair itself. Advanced Rejuvenation is introducing these cutting edge therapies to stimulate the body to recover from heart attack, stroke and peripheral vascular disease.

Beth Moran, Nurse Practitioner, has over 30 years experience in assisting clients in finding their path to wellness. Beth has been a guest lecturer at Stony Brook University. She has also been interviewed on CBS, The McNeil Lehrer Report, Medical News Network, The Gar Beth Moran is also a co-founder of The Guild of Holistic Practitioners, and has been a pioneer in the field of holistic health care on the East End of Null Show and numerous other TV and radio shows. For five years, Beth hosted her own TV show on Cablevision, interviewing holistic health care providers. Beth Moran is the author of Intuitive Healing A Womans Guide To Finding The Healer Within, which gives a great deal of information about the mind and body and encourages patients to listen and advocate for themselves. Using Hormone Replacement Therapy (HRT), Thyroid Balance, Hyperbaric Oxygen, Ozone, Stem Cell & Prolozone Therapy, IV Nutritional Therapies, PK Protocol, Treatment for CIRS and Chronic Lyme and Bio Identical Hormones.

Dr. Bramson received his undergraduate degree from the University of South Florida in 1974. He attended the Illinois College of Podiatry Medicine, where he graduated in 1978. He later went on to obtain a degree at Essex University in Maryland as a Physician Assistant in 1997. Dr Bramson worked as both a Podiatrist and an Orthopedic Physician Assistant for close to 15 years for a orthopedic practice in Fort Washington, Maryland. He has extensive experience in Orthopedics which includes assisting in hundreds of surgical procedures such as total knee and hip replacements. He is now playing for the other side using PRP, stem cells, Exosome, prolotherapy and prolozone to stimulate the body to repair itself.With hisextensive background as a podiatrist, he brings a great advantage to Advanced Rejuvenation with our lower extremity cases such as lower back pain, hip, knee, foot and ankle.

Dr. John A. Lieurance is a Doctor of Chiropractic, Registered Medical Assistant and Performs Musculoskeletal Ultrasound.He has practiced in Sarasota for 24 years. Dr. Lieurance has a gift for difficult cases where other practitioners have failed.With the successful integration ofFunctional Neurology, Chiropractic, Naturopathy, Nutrition using the Asyra, Detoxification Programs and LumoMed inner ear therapy. His Musculoskeletal Ultrasound training includes over 100 hours through the Gulf Coast Ultrasound Institute, 60 hours through AAOM, 90 hours through AOAPRM, and 60 through TOBI. He also has extensive training for sterile lab procedures and the processing of blood platelets, bone marrow aspirate, and adipose tissue through Oregen Biologics, Emcyte Corporation, Ron Gardener, M.D. (orthopedic surgeon), Regenestem and the Ageless Regenerative Institute. He has been an assistant instructor for hands on practicum for diagnosis using musculoskeletal ultrasound for the 3rd Annual Platelet Rich Plasma & Regenerative Medicine Symposium in Los Angeles, California in 2015, and was a speaker at the Florida Chiropractic Physicians Association (FCPA) in Orlando, Florida in 2016 onClinical Applications of Musculoskeletal Ultrasound. He has completed training with Dr Richie Shoemaker in 2019 for diagnosis & treatment of CIRS and with the Shoemaker Protocol. His techniques include Continuing Chiropractic Education:Chiropractic Neurology, Applied Kinesiology AK, Cold Laser Therapy, Extremity Adjusting, Sports Injury Taping, Rehabilitation of Sports Injuries, Lumbar and Cervical Disc Decompression, Pettibon Scoliosis through the Carrick Institute of Functional Neurology as well as Vestibular Rehabilitation & Movement Disorders through the Carrick Institute in 2012,16, 17, 18. Dr. Lieurance is the developer of Functional Cranial Release and teaches, as well as certifies, these methods to doctors around the world.

Alex received his graduate degree from the East West College of Natural Medicine where he excelled in his understanding and practical application of both Eastern and Western Medicine. In addition to his Oriental Medical knowledge, he has an equal proficiency in Western Medical diagnosis, which resulted under the tutelage of the great Dr. Banerjee M.D. FACE (Listed in Best Doctors of America). Dr. Smithers is a board certified Doctor of Oriental Medicine and Acupuncture Physician as well as a Registered Medical Assistant. A native of Sarasota, Dr. Smithers family has a long and well-known history in the community. Dr. Smithers work with Prolozone, Prolotherapy and other regenerative therapies as well as High Definition Ultrasound.His knowledge of both Eastern and Western medicine gives him a special gift in this field of medicine. He is currently scheduled to sit the medical boards at the end of the year to receive his M.D.

30 Years experience with IV nutrition to include administration of IV vitamins, PK Protocol, 10 Pass Hyperbaric Ozone, Glutathione, NAD Infusion and Silver IV. She is certified ICU nurse and if she cant get your IV started than no-one can!

The rest is here:
Stem Cell Therapy Sarasota | Advanced Rejuventation

Stem Cell for Lower Back Pain Specialists Tombal TX | The Best Stem Cell Doctors in Tombal TX

Stem Cell for Lower Back Pain Specialists Tombal TX | The Best Stem Cell Doctors in Tombal TX CALL US: (281) 367-6900 http://painreliefandhealing.com

http://bit.ly/TheBestStemCellTherapyT... CLICK HERE (WEBSITE) http://bit.ly/StemCellsforBackPainRel... CLICK HERE (FACEBOOK) http://bit.ly/StemCellKneePainDoctors... CLICK HERE (YOUTUBE)

Stem Cell Therapy Treatment

Stem Cell Therapy Tombal TX, Come in today feel better tonight! Pain is one of the many reasons a patient comes to the doctor. At the Institute Stem Cell Therapy Tombal TX,we are very serious about treating the cause of your pain. We know it is vital to relieve the pain as soon as possible. For this reason we see patients same day most of the time. Stem Cell Therapy Tombal TX, Physical medicine procedures, acupuncture or pain management services are rapidly engaged with the goal of relieving the pain while correcting the cause.

stem cell pain Tombal TX, stem cell spine Tombal TX, stem cell doctors in Tombal TX, Stem Cell doctors Tombal TX, stem cell knee Tombal TX, stem cell knee injections Tombal TX, stem cell pain Tombal TX, stem Cell doctor Tombal TX, Stem Cell doctor in Tombal TX, stem cell joint Tombal TX, stem cell clinic in Tombal TX, stem cell treatment Tombal TX, stem cell back Tombal TX, stem cell injections Tombal TX, stem cell knee therapy for Tombal TX, stem cell clinic in Tombal TX, stem cell knee Tombal TX, stem cell pain Tombal TX, stem cell joint Tombal TX, stem cell treatment Tombal TX, stem cells injection Tombal TX, stem cell knee Tombal TX, stem cell injections Tombal TX, stem cell clinic in Tombal TX, knee pain diagnosis Tombal TX, knee pain symptom checker Tombal TX, knee pain home remedies Tombal TX, knee pain when walking Tombal TX, knee pain Tombal TX, knee pain treatment Tombal TX, Knee pain Treatments in Tombal TX, Knee pain treatment in Tombal TX, knee pain doctors Tombal TX, knee pain doctors in Tombal TX, The Best knee pain doctor Tombal TX, The best knee pain doctors in Tombal TX, knee pain specialist Tombal TX, knee pain specialist in Tombal TX, knee pain relief doctor Tombal TX, knee pain relief doctors Tombal TX, knee pain treatment Tombal TX, knee pain treatment in Tombal TX, knee pain treatments Tombal TX, Knee pain Treatments in Tombal TX, Chronic knee pain Tombal TX, Chronic knee pain Doctor Tombal TX, Chronic knee pain doctors in Tombal TX, chronic knee pain specialist Tombal TX, chronic knee pain specialists Tombal TX, stem cell treatment Tombal TX, stem cell clinic Tombal TX, stem cell joint Tombal TX, stem cell center Tombal TX, Stem Cell Doctors for lower back pain in Tombal TX, The Best Stem Cell Doctors for lower back pain in Tombal TX, Stem cell for lower back pain Specialist in Tombal TX, Stem Cell for lower back pain Specialists in Tombal TX, stem cells for herniated discs Tombal TX, Stem Cells for herniated Discs in Tombal TX, Stem Cells for herniated Discs Doctors in Tombal TX, Arthritis joint pain Tombal TX, Arthritis joint pain specialist Tombal TX, osteoarthritis doctors Tombal TX, Osteo arthritis doctor in Tombal TX, Osteoarthritis Doctors in Tombal TX, Osteoarthritis specialist Tombal TX, frozen shoulder doctor in Tombal TX, frozen shoulder doctors Tombal TX, frozen shoulder specialist Tombal TX, frozen shoulder specialist in Tombal TX, frozen shoulder specialists in Tombal TX, The best doctor for frozen shoulder Tombal TX, frozen shoulder treatment Tombal TX, _____________________________________

Read more here:
Stem Cell for Lower Back Pain Specialists Tombal TX | The Best Stem Cell Doctors in Tombal TX

Regenerative Medicine Uses And Future Potential …

What Is Regenerative Medicine? If youre suffering from ongoing, nagging chronic pain that hasnt benefited from other treatments, you could benefit from an exciting new area of pain management known as regenerative medicine. These minimally-invasive treatments offer patients pain relief, while reducing the likelihood of infection and avoiding the need for surgery. For many patients, regenerative medicines can help them get their lives back by jump-starting their bodys own natural healing processes.

Regenerative medicine includes treatments like PRP therapy and stem cell therapy. The most common conditions that have been successfully treated using regenerative medical procedures include arthritis andinjuries to cartilage, tendons, muscle, bone, spinal discs, and other tissue types. For many patients, it can help them:

In this post, we talk about how these treatments work and if they could relieve your pain.

For these patients, regenerative medicine may be able to help. Regenerative medicines are cutting-edge therapies that use chemistry, medicine, robotics, biology, computer science, genetics, and engineering to construct a biologically compatible structure for many different tissues found in the body.Although relatively new in the field of acute and chronic pain management, regenerative medical procedures do date back as early as 1962.

Regenerative medicine offers a number of different benefits, including:

The following TED Talk gives a greater overview of the possible future benefits of this therapy.

The vast majority of early regenerative procedures were tissue-based, being developed for skin grafting. The first successful tissue that was engineered for grafting procedures was finalized in the 1970s, a mere eight years after the first synthetic tissue was developed. Howard Green and colleagues from Harvard Medical School began by harvesting a skin biopsy, later perfecting the practice of growing skin epidermis.

Following these advancements, researchers developed regenerative medical improvements in stem cell research, enabling successful bone marrow transplantation for individuals suffering from leukemia.

Having discovered the ability of the body to organize and regenerate tissue after cell death, researchers aimed their future studies at the goal of regenerative medicine and tissue engineering to replace tissue that had been damaged, lost through injury, or deteriorating with advanced age. Many diseases and injuries that result from failing tissue could potentially be successfully treated using regenerative medicine therapies.

Symptoms will vary patient to patient, but may include:

Pain is thought to be an indicator of tissue damage or an underlying injury. A number of existing treatments can help a patient cope with chronic pain, but dont fully address the underlying damage or injury. New regenerative medicine therapies can target the underlying problem and promote the bodys natural healing processes. Patients who experience the following list of conditions may be viable candidates for regenerative medicine.

Osteoarthritis is a chronic joint condition that causes degenerative cartilage changes in the joints. It is believed that the damage can be the result of wear and tear on the joint through a number of years or as a direct result of a specific injury. With enough damage to the cartilage that protects the joint, there is a high risk opposing bones will rub directly against each other. This direct contact causes damage to the ends of the bones and a significant inflammatory response and pain.

Traditional treatment approaches only help to manage the pain and not cure the condition. Stem cell therapy is believed to be more fitting since the goal is to repair the condition and reduce the bone-on-bone contact.

This is a degenerative condition of the individual bones of the spine, called vertebrae. Most commonly, spondylolisthesis occurs when the vertebrae slips over one another or becomes dislocated. Patients may experience nonspecific low back pain because a large number of individuals with this anatomical distortion do not present to their physician with related symptoms, including pain.

With spondylolisthesis the nerves around the weakened vertebrae can become compressed, resulting in pain and potentially muscle weakness. These symptoms can include:

Estimates are that 12% of the population has had difficulties with spondylolisthesis.

This common condition is characterized by a narrowing of the spinal canal. With spinal stenosis there is a restriction from this narrowing that results in neurogenic claudication. The spine is a row of 26 bones that allows movement and bending. Through the center is an opening, or canal, that protects the spinal cord. The narrowing with spinal stenosis can occur in the center, in the canals, or the spaces between the vertebrae. This narrowing puts pressure on the nerves in the spinal cord and can result in pain or numbness in the legs or shoulders, depending upon where the restriction is located.

Spinal stenosis is more common in individuals over the age of 50 years, but may occur in younger people who suffer an injury to the spine or are born with a narrowing of the spinal canal. Generally, pain specialists suggest using conservative or lifestyle changes to manage pain from this condition. If that doesnt work, patients may find relief with regenerative medicine techniques.

Spinal deformities are genetically linked issues that are related to the natural curvature of the spine. They generally involve the entire length of the spinal column and are relatively uncommon.

Conditions can affect the cervical, thoracic, or lumbar spinal regions and symptoms will vary widely depending upon the location. Some of these conditions are visible at birth, while others are only diagnosed when signs and symptoms develop.

A compression fracture is typically caused by osteoporosis and has a higher prevalence rate among post-menopausal women and in those individuals with a long history of corticosteroid use. These fractures result in a decrease in height of the vertebrae of at least 15 to 20%.

In one study, which examined 7,000 women over the age of 65, researchers found that 5% had suffered a compression fracture over a four-year period. Previous studies have suggested that nearly 4% of adults evaluated in a primary care setting could attribute back pain symptoms to a compression fracture.

Degenerative disc disease is a condition that results in symptoms from changes to the vertebral discs in adults as they age. The aging process increases the risk of tears to the disc, which is a likely cause of this type of pain. Spinal discs are soft and compressible that helps cushion the spinal column, which allows the spine to flex, twist, and bend. Although it can occur anywhere along the spine, it most often occurs in the lower back and neck. The pain can occur throughout the spine but in some instances affects the intervertebral disc.

Treatment modalities include physical therapy, pain medications, spinal fusion surgery, and steroid injections. New advances in regenerative medicine and stem cell therapy can help patients suffering from degenerative disc disease. Following extraction of the patients stem cells, usually from the bone marrow in the hip, the cells are engineered, concentrated, and injected into the site of the injury.

A herniated disc is characterized by damage to the intervertebral discs, which cause them to bulge from the intervertebral space or to rupture completely. The daily stress of movement, poor posture, injuries, and age can cause them to bulge, rupture, or herniate. The expansion of the disc material puts pressure on the surrounding nerves and spinal column, which is believed to be the source of pain. Herniated discs are more commonly found in aging people. Treatments include physical therapy, which has shown promise in relieving pain and improving the ability to function daily, however, it requires a significant time commitment in the therapists office and in daily home exercises.

Surgical treatment options may be suggested to cut out or remove the bulging or herniated material from the spinal column. The removal of the herniated disc carries a number of different risks related to the area of the spinal column where the disc is located and the weakened area of the column following surgery. Surgery is not always successful and there is a slight risk of damage to the spine or nerves, and risk of infection. New techniques from regenerative medicine using a patients own stem cells has shown good results with regeneration and rebuilding of the network of cells that make up the injured disc.

This is a common form of chronic foot pain that occurs between the ball of the foot and the heel. There is a thick connective tissue on the bottom of the foot, called the plantar fascia, which connects the ball of the foot to the heel. This plantar fascia supports the arch of the foot and can become strained from a number of different sources, including overuse, tight calf muscles, and poor foot placement. The damage forms tiny tears along the ligament, which is the likely source of pain. Treatments usually target the underlying condition, and then the symptoms of pain.

Regenerative medical treatments are an ideal choice for patients who have chronic pain in the plantar fascia and have corrected the underlying biomechanical issue that caused the initial condition. These therapies will promote healing of the damaged tissue. In fact, several studies provide realistic support for the use of platelet rich plasma therapy as an effective method of treatment to reduce or eliminate the pain associated with plantar fasciitis.

The sacroiliac joint is a large joint area that is located at the base of the spine. The joint connects the spine to the hip, or pelvis. In many cases, the individual can identify an injury that transpired previous to the onset of pain. Causes of this type of pain include:

There is limited evidence that current treatments are successful. Once a doctor relieves the underlying cause of pain, regenerative medicine may affect some degree of pain relief. This pain relief appears to last longer than that of steroid injections.

Also known as sciatica, lumbar radiculopathy occurs when a herniated disc, often between L5 and S1, pushes against the nerve. Patients experience pain that travels down the leg. The primary goal is to reduce the size of the disc and reduce the compression on the nerve root, thus reducing the pain.

There are a number of different treatment options for patients who suffer from lumbar radiculopathy. However, if they are unsuccessful, or if patients do not receive relief from their pain, they can be a candidate for stem cell injections.

In cervical radiculopathy, patients experience chronic pain originating from the cervical spine, or the neck area. When a disc in the neck pushes against a nerve root exiting the cervical spine, it causes pain to travel down the arms.

Radiculopathy in younger individuals can be from a herniated disc or neck injury. Older adults may suffer but physicians expect to also find osteophyte formation causing narrowing of the foramen, a reduced disc height and degenerative changes in the intervertebral joints.

Some patients who experience severe, unremitting back pain choose to undergo surgery to gain relief. Unfortunately, some patients may continue to suffer pain following surgical repair, which is recognized as a failed back surgery. Causes include:

Individuals with a history of other emotional disturbances, such as difficulty falling or staying asleep, depression, or anxiety are at an increased risk of developing chronic pain conditions following a back surgery.

Pain symptoms of a failed back surgery are usually dull, aching pain that is diffuse across the back and legs. Some patients do suffer from stabbing, pricking, or sharp pain in the limbs. When other treatments have failed to relieve pain following a failed back surgery, regenerative medicine treatment options might be considered.

Further, regenerative medicine therapies are relatively new and historically have very few studies documenting their effectiveness on different types of pain conditions. The FDA has not approved the use of adult stem cells to treat aging or to prevent, treat, or cure any disease or medical condition mentioned. Because of this, your insurance may not cover the cost of these procedures.

In addition, many regenerative medicine studies and treatments involve the use of living stem cells.Both legal and ethical issues are inherent in the use of embryonic stem cells. Although stem cell research holds great promise for the development of successful treatment modalities for conditions that thus far have no permanent treatment, research also raises both ethical and political controversies. However, reprogramming adult stem cells to produce pluripotent stem cells avoids these ethical issues that are specific to embryonic stem cell research. Adult or pluripotent stem cells are used for the majority of all regenerative pain medicine approaches, but always talk to your doctor if you have further questions.

To learn more about regenerative medicine for your pain condition, you can find a pain doctor in your area by clicking the button below or looking for one in your area by using the tips here: https://paindoctor.com/pain-management-doctors/.

See original here:
Regenerative Medicine Uses And Future Potential ...