Saturday, August 22, 2009

8/23 pubmed: adipose stem cell

Please add updates@feedmyinbox.com to your address book to make sure you receive these messages in the future.
pubmed: adipose stem cell Feed My Inbox

Adipose tissue mesenchymal stem cell expansion in animal serum-free medium supplemented with autologous human platelet lysate.
August 22, 2009 at 12:51 pm

Related Articles

Adipose tissue mesenchymal stem cell expansion in animal serum-free medium supplemented with autologous human platelet lysate.

Transfusion. 2009 Aug 18;

Authors: Blande IS, Bassaneze V, Lavini-Ramos C, Fae KC, Kalil J, Miyakawa AA, Schettert IT, Krieger JE

BACKGROUND: Mesenchymal stem cells (MSCs) have been considered for human regenerative therapy applications, and safe culture and expansion protocols are needed especially in the context of interspecies contamination. Human platelet lysate (PL) has been proposed as animal serum substitute during in vitro MSC expansion. In this work, a simplified and efficient method to obtain autologous PL to replace animal serum in cell culture applications is described. STUDY DESIGN AND METHODS: PL obtained by freezing and centrifugation procedures was tested as medium supplement for human adipose mesenchymal stem cell (hASC) culture. Differential proliferation, immunophenotypic changes, and differentiation under PL or fetal bovine serum (FBS) were assessed. RESULTS: In contrast to 10% FBS supplementation, cell population doubling time was significantly lower when hASCs were cultured with the same concentration of PL (PL 22.9 +/- 1.5 hr vs. FBS 106.7 +/- 6.5 hr, t test, p < 0.05). Furthermore, hASCs maintained with 2.5% PL supplementation also showed satisfactory results. Immunophenotypic analysis revealed no differences between hASCs cultivated with PL or FBS supplementation and both cultures retained the potential to differentiate into adipose cells. These results demonstrate that autologous PL obtained from the same donor can be used as animal serum substitute in hASC culture. CONCLUSIONS: Taken together, evidence is provided that platelets provided by a single donor are sufficient to obtain PL for hASC propagation for clinical-scale applications mitigating the potential untoward side effects associated with the use of animal-derived reagents.

PMID: 19694997 [PubMed - as supplied by publisher]


Histone deacetylase inhibitors decrease proliferation potential and multilineage differentiation capability of human mesenchymal stem cells.
August 22, 2009 at 12:51 pm

Related Articles

Histone deacetylase inhibitors decrease proliferation potential and multilineage differentiation capability of human mesenchymal stem cells.

Cell Prolif. 2009 Aug 17;

Authors: Lee S, Park JR, Seo MS, Roh KH, Park SB, Hwang JW, Sun B, Seo K, Lee YS, Kang SK, Jung JW, Kang KS

Abstract Objectives: Histone deacetylase (HDAC) is an important therapeutic target in cancer. Two of the main anticancer mechanisms of HDAC inhibitors are induction of terminal differentiation and inhibition of cell proliferation. To investigate the role of HDAC in maintenance of self-renewal and cell proliferation, we treated mesenchymal stem cells (MSCs) that originated from adipose tissue or umbilical cord blood with valproic acid (VPA) and sodium butyrate (NaBu). Materials and methods: Human MSCs were isolated from mammary fat tissue and cord blood. We performed MTT assay and flow cytometry-based cell cycle analysis to assess self-renewal of MSCs. In vitro differentiation assays into osteogenic, adipogenic, neurogenic and chondrogenic lineages were conducted to investigate MSC multipotency. Immunocytochemistry, Western blot and reverse transcription-polymerase chain reaction were used to interrogate molecular pathways. Results: VPA and NaBu flattened the morphology of MSCs and inhibited their growth. VPA and NaBu activated the transcription of p21(CIP1/WAF1) by increasing the acetylation of histone H3 and H4 and eventually blocked the cell cycle at G2/M phase. The expression level of p16(INK4A), a cdk inhibitor that is closely related to cellular senescence, was not changed by HDAC inhibitor treatment. We performed controlled differentiation into bone, fat, cartilage and nervous tissue to elucidate the role of HDAC in the pluripotency of MSC to differentiate into functional tissues. VPA and NaBu decreased the efficiency of adipogenic, chondrogenic, and neurogenic differentiation as visualized by specific staining and reverse transcription-polymerase chain reaction. In contrast, osteogenic differentiation was elevated by HDAC inhibitor treatment. Conclusion: HDAC activity is essential for maintaining the self-renewal and pluripotency of MSCs.

PMID: 19689470 [PubMed - as supplied by publisher]


In Vivo Physiologic Aransdifferentiation of Adult Adipose Cells.
August 22, 2009 at 12:51 pm

Related Articles

In Vivo Physiologic Aransdifferentiation of Adult Adipose Cells.

Stem Cells. 2009 Aug 17;

Authors: De Matteis R, Zingaretti MC, Murano I, Vitali A, Frontini A, Giannulis I, Barbatelli G, Marcucci F, Bordicchia M, Sarzani R, Raviola E, Cinti S

Grafts of adipose tissue from adult ROSA26 mice from different sites of the body, irrespective of the sex of the donor, share with the mammary fat the property of giving rise to milk-secreting epithelial cells when exposed to the microenvironment of mammary gland in pregnant and lactating females. To rule out the possibility that the labeled mammary glandular tissue was derived from stem cells associated with the stroma vascular part of the grafts, we injected into the mammary gland a pure suspension of adipocytes obtained by treating a fragment of adipose tissue with collagenase. X-gal positive cells became inserted into the alveoli of the native gland and electron microscopy showed that the labeled cells had transformed into milk-secreting glandular cells. At the site of the adipocyte injection the labeled alveoli contained a mixture of X-gal positive and negative cells and, occasionally, a single epithelial cell was stained in an otherwise unlabeled alveolus. This suggests that growing ducts individually recruit adjacent adipocytes that transdifferentiate into secretory epithelial cells as they became part of the glandular alveoli. After dissociation, the isolated adipocytes retained the morphology and protein markers typical of differentiated fat cells but expressed high level of stem cell genes and the reprogramming transcription factor Klf4.Thus, the well-documented osteogenic, chondrogenic, myogenic and angiogenic transformation of preadipocytes associated with the stroma-vascular component of the adipose tissue, may reflect an intrinsic capability of adipocytes to reprogram their gene expression and transform into different cytotypes.

PMID: 19688834 [PubMed - as supplied by publisher]


Homing of adipose-derived stem cells to radiofrequency catheter ablated canine atrium and differentiation into cardiomyocyte-like cells.
August 22, 2009 at 12:51 pm

Related Articles

Homing of adipose-derived stem cells to radiofrequency catheter ablated canine atrium and differentiation into cardiomyocyte-like cells.

Int J Cardiol. 2009 Aug 14;

Authors: Kim U, Shin DG, Park JS, Kim YJ, Park SI, Moon YM, Jeong KS

BACKGROUND AND OBJECTIVES: The purpose was to determine whether human adipose-derived stem cells (h-ASCs) can home to the radiofrequency ablated myocardial lesions when injected intravenously and differentiate into cardiomyocyte. METHODS: Human adipose tissues were obtained from patients and h-ASCs were isolated and cultured. The phenotype of isolated h-ASCs was identified by flow cytometry. Radiofrequency catheter ablation (RFCA) was performed with ten ablation pulses (40 W, 60 s each) to induce heat-mediated lesions at the free walls of the right atria of 14 dogs. Twenty-four hours after ablation, h-ASCs (1x10(7) cells) labeled with superparamagnetic iron oxide particles (SPIOs) were infused intravenously in 10 dogs as cell-therapy group and only saline without cells was infused in 4 dogs as control. The hearts were explanted 4 weeks later. RESULTS: h-ASCs were identified by flow cytometry as mesenchymal stem cell as positive for CD 13, CD29, CD44, CD90, CD166 and HLA-ABC and immunophenotyping revealed no immunologic responses. SPIO-labeled cells were identified in areas surrounding the RFCA-induced lesions by Prussian blue staining. Immunohistochemistry staining showed positive for anti-alpha-actinin, anti-cardiac troponin-I, anti-connexin 43 and anti-VEGFR-2. No lymphocyte infiltration, immunorejection or neoplasm-like cells were found in the h-ASC-positive areas. However, multiple iron-labeled h-ASCs were detected in lungs and spleens of cell-therapy group. CONCLUSION: h-ASCs can home into injured atrial tissue and express a cardiomyocyte-like phenotype, suggesting that intravenous delivery of stem cells might be feasible. Functional studies and quantification of delivered stem cells are needed for better evaluation and understanding of differentiation into cardiomyocytes.

PMID: 19683815 [PubMed - as supplied by publisher]


Influence of decantation, washing and centrifugation on adipocyte and mesenchymal stem cell content of aspirated adipose tissue: a comparative study.
August 22, 2009 at 12:51 pm

Related Articles

Influence of decantation, washing and centrifugation on adipocyte and mesenchymal stem cell content of aspirated adipose tissue: a comparative study.

J Plast Reconstr Aesthet Surg. 2009 Aug 11;

Authors: Condé-Green A, Gontijo de Amorim NF, Pitanguy I

BACKGROUND: In the last decade, controversy has arisen regarding the influence of fat harvesting, processing and injection techniques on adipose tissue graft. The aim of this study is to compare the influence of three widely used fat processing techniques in plastic surgery on the viability and number of adipocytes and mesenchymal stem cells (MSCs) of aspirated fat. METHODS: A prospective cross-sectional study was conducted in 20 adult healthy female patients in whom material obtained by liposuction of the lower abdomen was separated and processed by decantation, washing or centrifugation. The morphology and quantity of adipocytes were determined by histological analysis. The viability and number of MSCs in the middle layer of each lipoaspirate and the pellet derived from centrifuged samples were obtained by multi-colour flow cytometry. RESULTS: Cell count per high-powered field of intact nucleated adipocytes was significantly greater in decanted lipoaspirates, whereas centrifuged samples showed a greater majority of altered adipocytes. MSC concentration was significantly higher in washed lipoaspirates compared to decanted and centrifuged samples. However, the pellet collected at the bottom of the centrifuged samples showed the highest concentration of MSCs. CONCLUSION: Based on the theory of cell survival stating the importance of adipocytes' integrity for graft survival and the theory claiming the importance of regenerative MSCs in the maintenance and stabilisation of fat transplant, washing may turn out to be the best processing technique for adipose tissue graft take. While eliminating most contaminants during the process, it preserved and maintained the quantity, integrity and viability of the most important components of aspirated adipose tissue.

PMID: 19679523 [PubMed - as supplied by publisher]


Adipose-Derived Mesenchymal Stem Cells Ameliorate Chronic Experimental Autoimmune Encephalomyelitis.
August 22, 2009 at 12:51 pm

Related Articles

Adipose-Derived Mesenchymal Stem Cells Ameliorate Chronic Experimental Autoimmune Encephalomyelitis.

Stem Cells. 2009 Aug 12;

Authors: Constantin G, Marconi S, Rossi B, Angiari S, Calderan L, Anghileri E, Gini B, Bach SD, Martinello M, Bifari F, Galiè M, Turano E, Budui S, Sbarbati A, Krampera M, Bonetti B

Mesenchymal stem cells (MSC) represent a promising therapeutic approach for neurological autoimmune diseases; previous studies have shown that treatment with bone marrow-derived MSC induces immune modulation and reduces disease severity in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Here we show that intravenous administration of adipose-derived MSC (ASC) before disease onset significantly reduces the severity of EAE by immune modulation and decreases spinal cord inflammation and demyelination. ASC preferentially home into lymphoid organs, but migrates also inside the CNS. Most importantly, administration of ASC in chronic established EAE significantly ameliorates the disease course and reduces both demyelination and axonal loss, and induce a Th2-type cytokine shift in T cells. Interestingly, a relevant subset of ASC expresses activated alpha4 integrins and adheres to inflamed brain venules in intravital microscopy experiments. Bioluminescence imaging shows that alpha4 integrins control ASC accumulation in inflamed CNS. Importantly, we found that ASC cultures produce basic fibroblast growth factor, brain-derived growth factor and platelet-derived growth factor-AB. Moreover, ASC infiltration within demyelinated areas is accompanied by increased number of endogenous oligodendrocyte progenitors. In conclusion, we show that ASC have clear therapeutic potential by a bimodal mechanism, by suppressing the autoimmune response in early phases of disease as well as by inducing local neuro-regeneration by endogenous progenitors in animals with established disease. Overall our data suggest that ASC represent a valuable tool for stem cell-based therapy in chronic inflammatory diseases of the CNS.

PMID: 19676124 [PubMed - as supplied by publisher]


Cellular cardiac regenerative therapy in which patients?
August 22, 2009 at 12:51 pm

Related Articles

Cellular cardiac regenerative therapy in which patients?

Expert Rev Cardiovasc Ther. 2009 Aug;7(8):911-9

Authors: Chachques JC

Cell-based myocardial regenerative therapy is undergoing experimental and clinical trials in order to limit the consequences of decreased contractile function and compliance of damaged ventricles owing to ischemic and nonischemic myocardial diseases. A variety of myogenic and angiogenic cell types have been proposed, such as skeletal myoblasts, mononuclear and mesenchymal bone marrow cells, circulating blood-derived progenitors, adipose-derived stromal cells, induced pluripotent stem cells, umbilical cord cells, endometrial mesenchymal stem cells, adult testis pluripotent stem cells and embryonic cells. Current indications for stem cell therapy concern patients who have had a left- or right-ventricular infarction or idiopathic dilated cardiomyopathies. Other indications and potential applications include patients with diabetic cardiomyopathy, Chagas heart disease (American trypanosomiasis), ischemic mitral regurgitation, left ventricular noncompacted myocardium and pediatric cardiomyopathy. Suitable sources of cells for cardiac implant will depend on the types of diseases to be treated. For acute myocardial infarction, a cell that reduces myocardial necrosis and augments vascular blood flow will be desirable. For heart failure, cells that replace or promote myogenesis, reverse apoptopic mechanisms and reactivate dormant cell processes will be useful. It is important to note that stem cells are not an alternative to heart transplantation; selected patients should be in an early stage of heart failure as the goal of this regenerative approach is to avoid or delay organ transplantation. Since the cell niche provides crucial support needed for stem cell maintenance, the most interesting and realistic perspectives include the association of intramyocardial cell transplantation with tissue-engineered scaffolds and multisite cardiac pacing in order to transform a passive regenerative approach into a 'dynamic cellular support', a promising method for the creation of 'bioartificial myocardium'.

PMID: 19673669 [PubMed - in process]


Culture effects of epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) on cryopreserved human adipose-derived stromal/stem cell proliferation and adipogenesis.
August 22, 2009 at 12:51 pm

Related Articles

Culture effects of epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) on cryopreserved human adipose-derived stromal/stem cell proliferation and adipogenesis.

J Tissue Eng Regen Med. 2009 Aug 10;

Authors: Hebert TL, Wu X, Yu G, Goh BC, Halvorsen YD, Wang Z, Moro C, Gimble JM

Previous studies have demonstrated that EGF and bFGF maintain the stem cell properties of proliferating human adipose-derived stromal/stem cells (hASCs) in vitro. While the expansion and cryogenic preservation of isolated hASCs are routine, these manipulations can impact their proliferative and differentiation potential. This study examined cryogenically preserved hASCs (n = 4 donors), with respect to these functions, after culture with basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF) at varying concentrations (0-10 ng/ml). Relative to the control, cells supplemented with EGF and bFGF significantly increased proliferation by up to three-fold over 7-8 days. Furthermore, cryopreserved hASCs expanded in the presence of EGF and bFGF displayed increased oil red O staining following adipogenic induction. This was accompanied by significantly increased levels of several adipogenesis-related mRNAs: aP2, C/EBPalpha, lipoprotein lipase (LPL), PPARgamma and PPARgamma co-activator-1 (PGC1). Adipocytes derived from EGF- and bFGF-cultured hASCs exhibited more robust functionality based on insulin-stimulated glucose uptake and atrial natriuretic peptide (ANP)-stimulated lipolysis. These findings indicate that bFGF and EGF can be used as culture supplements to optimize the proliferative capacity of cryopreserved human ASCs and their adipogenic differentiation potential. Copyright (c) 2009 John Wiley & Sons, Ltd.

PMID: 19670348 [PubMed - as supplied by publisher]


Isolation method for a stem cell population with neural potential from skin and adipose tissue.
August 22, 2009 at 12:51 pm

Related Articles

Isolation method for a stem cell population with neural potential from skin and adipose tissue.

Neurol Res. 2009 Aug 5;

Authors: Vindigni V, Michelotto L, Lancerotto L, Puppa AD, D'Avella D, Abatangelo G, Cortivo R, Zavan B

OBJECTIVE: In recent years, research on stem cells has been focused on the development of personalized cell-based therapies. Owing to their homing properties, adult human stem cells are a promising source of autologous cells to be used as therapeutic vehicles. Multiple potential sources for clinically useful stem and progenitor cells have been identified, including autologous and allogenic embryonic, fetal and adult somatic cells from neural, adipose and mesenchymal tissue. In the present report, we describe a simple protocol to obtain an enriched culture of adult stem cells organized in neurospheres from two post-natal tissues: skin and adipose tissue. METHODS: Adult stem cells isolated from skin and adipose tissue derived from the same adult donor were amplified under varying conditions related to the coating of the chamber slide and the presence of serum and/or growth factors, such as with EGF and FGF2. Neurospheres were then expanded and evaluated in terms of proliferation and gene expression. RESULTS: Adipose and skin derived neurospheres were comparable in size, quantity of cells and genes expressed. Cells from both types of tissue grew optimally without slide coating, in the presence of serum and with the combined addition of FGF2 and EGF. DISCUSSION: We describe a method for isolating and improving a population of multipotent adult precursor cells from the two most accessible adult tissue sources: skin and adipose tissue. This autologous adult stem cell population could be used for cell replacement or cell therapies.

PMID: 19660183 [PubMed - as supplied by publisher]


Sequential hepatogenic trans-differentiation of adipose tissue-derived stem cells: relevance of different extracellular signaling molecules, transcription factors involved and expression of new key marker genes.
August 22, 2009 at 12:51 pm

Related Articles

Sequential hepatogenic trans-differentiation of adipose tissue-derived stem cells: relevance of different extracellular signaling molecules, transcription factors involved and expression of new key marker genes.

Cell Transplant. 2009 Aug 5;

Authors: A BC, R J, V M, A L, F C, Jv C, Mj GL

Adipose tissue contains a mesenchymal stem cell (MSC) population known as adipose-derived stem cells (ASCs) capable of differentiating into different cell types. Our aim was to induce hepatic trans-differentiation of ASCs by sequential exposure to several combinations of cytokines, growth factors and hormones. The most efficient hepatogenic protocol includes fibroblastic growth factors (FGF) 2 and 4 and epidermal growth factor (EGF) (Step 1), hepatocyte growth factor (HGF), FGF2, FGF4 and nicotinamide (Nic) (Step 2), and oncostatin M (OSM), dexamethasone (Dex) and insulin-tranferrin-selenium (Step 3). This protocol activated transcription factors (GATA6, Hex, CCAAT/enhancer binding protein alpha and beta, CEBPalpha and beta, peroxisome proliferator-activated receptor gamma, coactivator 1 alpha, PGC1alpha, and hepatocyte nuclear factor 4 alpha, HNF4alpha), which promoted a characteristic hepatic phenotype, as assessed by new informative markers for the step-by-step hepatic trans-differentiation of hMSC (early markers: albumin, ALB, alpha-2-macroglobuline, alpha2M, complement protein C3, C3, and selenoprotein P1, SEPP1; late markers: cytochrome P450 3A4, CYP3A4, apolipoprotein E, APOE, Acyl-CoA synthetase long-chain family member 1, ACSL1, and Angiotensin II receptor, type 1, AGTR1).The loss of adipose adult stem cell phenotype was detected by losing expression of Thy1 and inhibitor of DNA binding 3 (Id3). The re-expression of phosphoenolpyruvate corboxykinase, PEPCK, apolipoprotein C3, APOCIII, Aldolase B, ALDOB, and cytochrome P450 1A2, CYP1A2, was achieved by transduction with a recombinant adenovirus for HNF4alpha and finally hepatic functionality was also assessed by analyzing specific biochemical markers. We conclude that ASCs could represent an alternative tool in clinical therapy for liver dysfunction and regenerative medicine.

PMID: 19660180 [PubMed - as supplied by publisher]


[Stem cell therapy for corneal defects]
August 22, 2009 at 12:51 pm

Related Articles

[Stem cell therapy for corneal defects]

Klin Monatsbl Augenheilkd. 2009 Jul;226(7):541-5

Authors: Schilimow A, Wiechens B

BACKGROUND: The healing process of corneal defects requires functioning limbus stem cells. Their loss will lead to secondary wound healing problems. Stem cell research offers new treatment options. MATERIAL AND METHODS: A Medline search of the U. S. National Library of Medicine was carried out. RESULTS: The autologous limbus stem cell transplantation is currently the treatment of choice. Amniotic membrane transplantation, previously settled with limbus stem cells, is a clinically proven method. In animal experiments bone marrow-derived mesenchymal stem cells or epidermal stem cells can be used to improve healing of corneal defects. Adipose-derived stem cells may support the regenerative ability of the cornea as well. Moreover, membrane transplantation of epithelial cells from the buccal mucosa cultivated in vitro was clinically tested. DISCUSSION AND CONCLUSIONS: Limbus stem cell failure of both eye is the limiting factor for autologous limbus stem cell transplantation. Epithelial cells, epidermal stem cells, bone marrow- or adipose-derived mesenchymal stem cells promote the regeneration of the cornea and have become established for the treatment of corneal defects. Additionally, mesenchymal stem cells offer the advantage of immunosuppressive and anti-inflammatory effects.

PMID: 19644800 [PubMed - in process]


Targeting rat brainstem glioma using human neural stem cells and human mesenchymal stem cells.
August 22, 2009 at 12:51 pm

Related Articles

Targeting rat brainstem glioma using human neural stem cells and human mesenchymal stem cells.

Clin Cancer Res. 2009 Aug 1;15(15):4925-34

Authors: Lee DH, Ahn Y, Kim SU, Wang KC, Cho BK, Phi JH, Park IH, Black PM, Carroll RS, Lee J, Kim SK

PURPOSE: Brainstem gliomas are usually inoperable and have a dismal prognosis. Based on the robust tropisms of neural stem cells (NSC) and mesenchymal stem cells (MSC) to brain tumors, we compared the tumor-tropic migratory capacities of these stem cells and evaluated the therapeutic potential of genetically engineered human NSCs encoding cytosine deaminase (CD) and IFNbeta against brainstem gliomas. EXPERIMENTAL DESIGN: The directed migratory capacities of NSCs and MSCs to brainstem glioma (F98) were evaluated both in vitro and in vivo. The human NSCs (HB1.F3) and various human MSCs, such as bone marrow-derived MSCs (HM3.B10), adipose tissue-derived MSCs, and umbilical cord blood-derived MSCs, were tested. Human fibroblast cells (HFF-1) were used as the negative control. As a proof of concept, the bioactivity of HB1.F3-CD-IFNbeta was analyzed with a cell viability assay, and animals with brainstem gliomas were injected with HB1.F3-CD-IFNbeta cells followed by systemic 5-fluorocytosine treatment. RESULTS: In an in vitro modified Transwell migration assay and in vivo stem cell injection into established brainstem gliomas in rats, all the stem cells showed a significant migratory capacity compared with that of the control (P < 0.01). Histologic analysis showed a 59% reduction in tumor volume in the HB1.F3-CD-IFNbeta-treated group (P < 0.05). Apoptotic cells were increased 2.33-fold in animals treated with HB1.F3-CD-IFNbeta compared with the respective control groups (P < 0.01). CONCLUSION: The brainstem glioma-tropic migratory capacities of MSCs from various sources were similar to those of NSCs. Genetically engineered NSCs show therapeutic efficacy against brainstem gliomas.

PMID: 19638465 [PubMed - in process]


[Cell sheet fabrication of hepatocyte-like cells differentiated from adipose tissue mesenchymal stem cells]
August 22, 2009 at 12:51 pm

Related Articles

[Cell sheet fabrication of hepatocyte-like cells differentiated from adipose tissue mesenchymal stem cells]

Sheng Wu Gong Cheng Xue Bao. 2009 Apr;25(4):599-604

Authors: Lu Y, Qiu F, Chen Y, Zhao X

Adult pluripotent stem cells, such as mesenchymal stem cells derived from bone marrow and adipose tissue are capable of multilineage differentiation. Although autologous stem cell transplantation is an effective alternative to organ transplantation, the loss of cell viability and differentiation confinement of implanted cells has largely impaired the therapeutic efficacy. To produce biomaterial-free liver construct to integrate into living tissue, we isolated adipose mesenchymal stem cells and subjected them to a delicate culture configuration to mediate the hepatocyte differentiation. The differentiated hepatocyte-like cells were then inoculated onto poly (N-isopropylacrylamide) (PNIPAAm) grafted cell culture dish. By lowering the culture temperature to 20 degrees C, cells detached from the dish surface into a complete cell sheet. Hematoxylin and eosin staining and immunohistochemistry results showed that cell sheet was composed of 2-3 layers of cells and extracellular matrix was maintained intact. As compared with traditional cell harvest using trypsin digestion, cell sheet fabrication causes no damage to cell membrane and extracellular matrix. Hence, cell sheets would form a better interaction with tissues in situ, and a higher cell viability and therapeutic efficiency would be expected.

PMID: 19637638 [PubMed - in process]


Cardiomyoblast-like cells differentiated from human adipose tissue-derived mesenchymal stem cells improve left ventricular dysfunction and survival in a rat myocardial infarction model.
August 22, 2009 at 12:51 pm

Related Articles

Cardiomyoblast-like cells differentiated from human adipose tissue-derived mesenchymal stem cells improve left ventricular dysfunction and survival in a rat myocardial infarction model.

Tissue Eng Part C Methods. 2009 Jul 22;

Authors: Okura H, Matsuyama A, Lee CM, Saga A, Kakuta-Yamamoto A, Nagao A, Sougawa N, Sekiya N, Takekita K, Shudo Y, Miyagawa S, Komoda H, Okano T, Sawa Y

Adipose tissue-derived mesenchymal stem cells (ADMSCs) are multipotent cells. Here we examined whether human ADMSCs (hADMSCs) could differentiate into cardiomyoblast-like cells (CLC) by induction with dimethylsulfoxide (DMSO) and whether the cells would be utilized to treat cardiac dysfunction. DMSO induced the expression of various cardiac markers in hADMSCs, such as alpha-cardiac actin, cardiac myosin light chain, and myosin heavy chain; none of which were detected in noncommitted hADMSCs. The induced cells were thus designated as hADMSC-derived cardiomyoblast-like cells (hCLCs). To confirm their beneficial effect on cardiac function, hCLC cell patches were transplanted onto the Nude rat MI model, and compared to noncommitted hADMSC-patch transplants and sham operations. Echocardiography demonstrated significant short-term improvement of cardiac function in both patch-transplanted groups. However, long-term follow-up showed rescue and maintenance of cardiac function in the hCLC patch-transplanted group only, but not in the noncommitted-hADMSC patch-transplanted animals. The hCLCs, but not the hADMSCs, engrafted into the scarred myocardium and differentiated into human cardiac troponin I-positive cells, and thus regarded as cardiomyocytes. Transplantation of the hCLC-patches also resulted in recovery of cardiac function and improvement of long-term survival rate. Thus, transplantation of hADMSC-derived CLC patches is potentially effective therapeutic strategy for future cardiac tissue regeneration.

PMID: 19624256 [PubMed - as supplied by publisher]


Hypoxia-enhanced wound-healing function of adipose-derived stem cells: increase in stem cell proliferation and up-regulation of VEGF and bFGF.
August 22, 2009 at 12:51 pm

Related Articles

Hypoxia-enhanced wound-healing function of adipose-derived stem cells: increase in stem cell proliferation and up-regulation of VEGF and bFGF.

Wound Repair Regen. 2009 Jul-Aug;17(4):540-7

Authors: Lee EY, Xia Y, Kim WS, Kim MH, Kim TH, Kim KJ, Park BS, Sung JH

Adipose-derived stem cells (ADSCs) have been shown to induce wound-healing effects. Because inflammation near the wound area induces oxygen deficiency, it is interesting to elucidate the effect of hypoxia on the function of ADSCs. In this work, we asked: (1) does hypoxia alter the wound-healing function of ADSCs? and (2) what are the major factors responsible for the alteration in the wound-healing function? Effect of hypoxia on the proliferation of ADSCs was first examined that hypoxia (2% O(2)) enhanced the proliferation of ADSCs in either the presence of serum or in the absence of serum. The conditioned medium of ADSCs harvested under hypoxia (hypoCM) significantly promoted collagen synthesis and the migration of human dermal fibroblasts, compared with that in normoxia (norCM). In the animal studies, hypoCM significantly reduced the wound area compared with norCM. Furthermore, mRNA and protein measurements showed that hypoxia up-regulated growth factors such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF). Inhibition of VEGF and bFGF using neutralizing antibodies reversed the migration of the wounded human dermal fibroblasts and the healing of wounds in animal experiment. Collectively, these results suggest that hypoxia increases the proliferation of ADSCs and enhances the wound-healing function of ADSCs, at least partly, by up-regulating the secretion of VEGF and bFGF.

PMID: 19614919 [PubMed - in process]


Promotion of stem cell proliferation by vegetable peptone.
August 22, 2009 at 12:51 pm

Related Articles

Promotion of stem cell proliferation by vegetable peptone.

Cell Prolif. 2009 Jul 14;

Authors: Lee J, Lee J, Hwang H, Jung E, Huh S, Hyun J, Park D

Abstract Objectives: Technical limitations and evolution of therapeutic applications for cell culture-derived products have accelerated elimination of animal-derived constituents from such products to minimize inadvertent introduction of microbial contaminants, such as fungi, bacteria or viruses. The study described here was conducted to investigate the proliferative effect of vegetable peptone on adult stem cells in the absence of serum, and its possible mechanisms of action. Materials and methods: Cell viability and proliferation were determined using the MTT assay and Click-iT EdU flow cytometry, respectively. In addition, changes in expression of cytokine genes were analysed using MILLIPLEX human cytokine enzyme-linked immunosorbent assay kit. Results: Viability of cord blood-derived mesenchymal stem cells (CB-MSC) and adipose tissue-derived stem cells (ADSC) increased significantly when treated with the peptone. In addition, median value of the group treated with peptone shifted to the right when compared to the untreated control group. Furthermore, quantitative analysis of the cytokines revealed that production of vascular endothelial growth factor (VEGF), transforming growth factor-beta1 (TGF-beta1), and interleukin-6 (IL-6) increased significantly in response to treatment with our vegetable peptone in both CB-MSCs and ADSCs. Conclusions: Our findings revealed that the vegetable peptone promotes proliferation of CB-MSCs and ADSCs. In addition, results of this study suggest that induction of stem cell proliferation by vegetable peptone is likely to be related to its induction of VEGF, TGF-beta1, and IL-6 expression.

PMID: 19614679 [PubMed - as supplied by publisher]


Expression profile of mRNAs encoding core circadian regulatory proteins in human subcutaneous adipose tissue: correlation with age and body mass index.
August 22, 2009 at 12:51 pm

Related Articles

Expression profile of mRNAs encoding core circadian regulatory proteins in human subcutaneous adipose tissue: correlation with age and body mass index.

Int J Obes (Lond). 2009 Jul 14;

Authors: Wu X, Xie H, Yu G, Hebert T, Goh BC, Smith SR, Gimble JM

Objective:Circadian mechanisms underlie the physiology of mammals as an adaptation to the earth's rotation on its axis. Highly conserved core circadian regulatory proteins (CCRPs) maintain an oscillatory expression profile in the central and peripheral tissues. The CCRP include both a positive and negative arm, as well as downstream transcriptional regulators. Recent studies in murine models have determined that the mRNAs encoding the CCRP are present in multiple adipose tissue depots and exhibit a robust oscillatory expression profile. This study set out to examine the expression of CCRP mRNAs in human subcutaneous adipose tissues.Design:Retrospective analysis of total RNA isolated from subcutaneous adipose tissue.Subjects:A total of 150 healthy female and male lean (body mass index (BMI) <25), overweight (BMI between 25 and 29.99) or obese (BMI >30) subjects of varied ethnic backgrounds undergoing elective liposuction or surgical procedures.Results:The expression of the CCRP mRNAs displayed a significant correlation between each other and mRNAs representative of adipogenic biomarkers. Hierarchical cluster analyses of mRNAs isolated from the cohort of female Caucasian subjects (n=116) identified three major clusters based on expression of downstream CCRP mRNAs. The mRNAs encoding D site of albumin promoter-binding protein (DBP), E4 promoter-binding protein 4 (E4BP4), PPARgamma coactivator-1beta (PGC-1beta) and Rev-erbalpha were negatively correlated with BMI in a lean cluster (n=66), positively correlated with BMI in a younger overweight/obese cluster (n=19), and not significantly correlated with BMI in an older, overweight/obese cluster (n=31).Conclusions:These data confirm and extend findings that link the CCRP and circadian mechanisms to the risk of obesity.International Journal of Obesity advance online publication, 14 July 2009; doi:10.1038/ijo.2009.137.

PMID: 19597517 [PubMed - as supplied by publisher]


Isolation of Mesenchymal Stem Cells from Human Placental Decidua Basalis and Resistance to Hypoxia and Serum Deprivation.
August 22, 2009 at 12:51 pm

Related Articles

Isolation of Mesenchymal Stem Cells from Human Placental Decidua Basalis and Resistance to Hypoxia and Serum Deprivation.

Stem Cell Rev Rep. 2009 May 23;

Authors: Huang YC, Yang ZM, Chen XH, Tan MY, Wang J, Li XQ, Xie HQ, Deng L

Mesenchymal stem cells (MSCs) are the most promising seed cells for cell therapy and tissue engineering, which can be isolated from various sources of human adult tissues such as bone marrow and adipose tissue. However, cells from these tissues must be obtained through invasive procedures and sometimes the individual difference is hard to control. Hence, the search continues for an ethically conducive, easily accessible and controllable source of stem cells. We herein report the isolation of a population of stem cells from the human placental decidua basalis (termed as PDB-MSCs), a maternal portion of placenta. PDB-MSCs were further shown to express markers common to MSCs and positive for SSEA-1, SSEA-3, SSEA-4, TRA-1-60, TRA-1-81 and Oct-4. In order to facilitate the further utility in ischemic diseases, we tested the apoptosis of PDB-MSCs in hypoxia and serum deprivation, two components of ischemia in vivo. Taken together, our findings indicate that PDB-MSCs are resistant to hypoxia and serum deprivation, which may relate to Bcl-2.

PMID: 19590988 [PubMed - as supplied by publisher]


Characterization of human adult stem-cell populations isolated from visceral and subcutaneous adipose tissue.
August 22, 2009 at 12:51 pm

Related Articles

Characterization of human adult stem-cell populations isolated from visceral and subcutaneous adipose tissue.

FASEB J. 2009 Jul 7;

Authors: Baglioni S, Francalanci M, Squecco R, Lombardi A, Cantini G, Angeli R, Gelmini S, Guasti D, Benvenuti S, Annunziato F, Bani D, Liotta F, Francini F, Perigli G, Serio M, Luconi M

Adipose tissue is a dynamic endocrine organ with a central role in metabolism regulation. Functional differences in adipose tissue seem associated with the regional distribution of fat depots, in particular in subcutaneous and visceral omental pads. Here, we report for the first time the isolation of human adipose-derived adult stem cells from visceral omental and subcutaneous fat (V-ASCs and S-ASCs, respectively) from the same subject. Immunophenotyping shows that plastic culturing selects homogeneous cell populations of V-ASCs and S-ASCs from the corresponding stromal vascular fractions (SVFs), sharing typical markers of mesenchymal stem cells. Electron microscopy and electrophysiological and real-time RT-PCR analyses confirm the mesenchymal stem nature of both V-ASCs and S-ASCs, while no significant differences in a limited pattern of cytokine/chemokine expression can be detected. Similar to S-ASCs, V-ASCs can differentiate in vitro toward adipogenic, osteogenic, chondrogenic, muscular, and neuronal lineages, as demonstrated by histochemical, immunofluorescence, real-time RT-PCR, and electrophysiological analyses, suggesting the multipotency of such adult stem cells. Our data demonstrate that both visceral and subcutaneous adipose tissues are a source of pluripotent stem cells with multigermline potential. However, the visceral rather than the subcutaneous ASC could represent a more appropriate in vitro cell model for investigating the molecular mechanisms implicated in the pathophysiology of metabolic disorders such as obesity.-Baglioni, S., Francalanci, M., Squecco, R., Lombardi, A., Cantini, G., Angeli, R., Gelmini, S., Guasti, D., Benvenuti, S., Annunziato, F., Bani, D., Liotta, F., Francini, F., Perigli, G., Serio, M., Luconi, M. Characterization of human adult stem-cell populations isolated from visceral and subcutaneous adipose tissue.

PMID: 19584303 [PubMed - as supplied by publisher]


Effect of oxygen concentration, culture format and donor variability on in vitro chondrogenesis of human adipose tissue-derived stem cells.
August 22, 2009 at 12:51 pm

Related Articles

Effect of oxygen concentration, culture format and donor variability on in vitro chondrogenesis of human adipose tissue-derived stem cells.

Regen Med. 2009 Jul;4(4):539-48

Authors: Pilgaard L, Lund P, Duroux M, Fink T, Ulrich-Vinther M, Søballe K, Zachar V

BACKGROUND: The chondrogenic differentiation potential of the easily accessible adipose tissue-derived stem cells (ASCs) is of particular interest within the field of tissue engineering for treating cartilage defects. However, no consensus has been reached as to which oxygen tension is more beneficial for the differentiation process. MATERIALS & METHODS: In this investigation, the impact of available oxygen was investigated to identify optimal conditions for human ASC chondrogenesis in vitro. Four physiologically relevant oxygen concentrations of 15, 10, 5 and 1% were compared with ambient air condition, and the ASCs originating from six unrelated donors were subjected to chondrogenic induction in high-density pellet cultures. RESULTS: The qualitative and quantitative assessment of accumulated extracellular matrix and the gene-expression analysis revealed marked interindividual differences, nevertheless the chondrogenic process was optimally supported in high-density pellet setup at ambient or 15% oxygen concentrations, irrespective of the origin of cells. The histochemical analysis based on alcian blue staining demonstrated that the differentiation took place in a gradient-like fashion, displaying highest levels in restricted regions, most often adjacent to the periphery. The two lowest hypoxic conditions, at 5 and 1% oxygen, seemed to have an inhibitory effect. CONCLUSION: The micropellet cultures at ambient or 15% oxygen concentration provided the most suitable environment for inducing chondrogenesis in ASCs. Furthermore, in light of the fact that the induction appeared in a zone-dependent manner, this format lends itself as a suitable model for further analysis of the relationship between chondrogenic differentiation and the gradient of nutrients.

PMID: 19580403 [PubMed - in process]


Hematopoietic stem cell origin of adipocytes.
August 22, 2009 at 12:51 pm

Related Articles

Hematopoietic stem cell origin of adipocytes.

Exp Hematol. 2009 Sep;37(9):1108-20, 1120.e1-4

Authors: Sera Y, LaRue AC, Moussa O, Mehrotra M, Duncan JD, Williams CR, Nishimoto E, Schulte BA, Watson PM, Watson DK, Ogawa M

OBJECTIVE: It has generally been believed that adipocytes are derived from mesenchymal stem cells via fibroblasts. We recently reported that fibroblasts/myofibroblasts in a number of tissues and organs are derived from hematopoietic stem cells (HSCs). In the present study, we tested the hypothesis that HSCs also give rise to adipocytes. MATERIALS AND METHODS: Using transplantation of a single enhanced green fluorescent protein-positive (EGFP(+)) HSC and primary culture, we examined generation of adipocytes from HSCs. RESULTS: Adipose tissues from clonally engrafted mice showed EGFP(+) adipocytes that stained positive for leptin, perilipin, and fatty acid binding protein 4. A diet containing rosiglitazone, a peroxisome proliferator-activated receptor-gamma agonist, significantly enhanced the number of EGFP(+) adipocytes. When EGFP(+) bone marrow cells from clonally engrafted mice were cultured under adipogenic conditions, all of the cultured cells stained positive with Oil Red O and Sudan Black B and exhibited the presence of abundant mRNA for adipocyte markers. Finally, clonal culture- and sorting-based studies of Mac-1 expression of hematopoietic progenitors suggested that adipocytes are derived from HSCs via progenitors for monocytes/macrophages. CONCLUSION: Together, these studies clarify the current controversy regarding the ability of HSCs to give rise to adipocytes. Furthermore, our primary culture method that generates adipocytes from uncommitted hematopoietic cells should contribute to the studies of the mechanisms of early adipocytic differentiation and may lead to development of therapeutic solutions for many general obesity issues.

PMID: 19576951 [PubMed - in process]


The Human Lipodystrophy Gene Product BSCL2/Seipin Plays a Key Role in Adipocyte Differentiation.
August 22, 2009 at 12:51 pm

Related Articles

The Human Lipodystrophy Gene Product BSCL2/Seipin Plays a Key Role in Adipocyte Differentiation.

Endocrinology. 2009 Jul 2;

Authors: Chen W, Yechoor VK, Chang BH, Li MV, March KL, Chan L

Mutations in the Berardinelli-Seip congenital lipodystrophy 2 gene (BSCL2) are the underlying defect in patients with congenital generalized lipodystrophy type 2. BSCL2 encodes a protein called seipin, whose function is largely unknown. In this study, we investigated the role of Bscl2 in the regulation of adipocyte differentiation. Bscl2 mRNA is highly upregulated during standard hormone-induced adipogenesis in 3T3-L1 cells in vitro. However, this upregulation does not occur during mesenchymal stem cell (C3H10T1/2 cells) commitment to the preadipocyte lineage. Knockdown of Bscl2 by short hairpin RNA (shRNA) in C3H10T1/2 cells has no effect on BMP4-induced preadipocyte commitment. However, knockdown in 3T3-L1 cells prevents adipogenesis induced by a standard hormone cocktail but adipogenesis can be rescued by the addition of PPARgamma agonist pioglitazone at an early stage of differentiation. Interestingly, pioglitazone-induced differentiation in the absence of standard hormone is not associated with upregulated Bscl2 expression. On the other hand, shRNA-knockdown of Bscl2 largely blocks pioglitazone-induced adipose differentiation. These experiments suggest that Bscl2 may be essential for normal adipogenesis; it works upstream or at the level of PPARgamma, enabling the latter to exert its full activity during adipogenesis. Loss of Bscl2 function thus interferes with the normal transcriptional cascade of adipogenesis during fat cell differentiation, resulting in near total loss of fat or lipodystrophy.

PMID: 19574402 [PubMed - as supplied by publisher]


Adipose tissue of control and ex-obese patients exhibit differences in blood vessel content and resident mesenchymal stem cell population.
August 22, 2009 at 12:51 pm

Related Articles

Adipose tissue of control and ex-obese patients exhibit differences in blood vessel content and resident mesenchymal stem cell population.

Obes Surg. 2009 Sep;19(9):1304-12

Authors: Baptista LS, da Silva KR, da Pedrosa CS, Claudio-da-Silva C, Carneiro JR, Aniceto M, de Mello-Coelho V, Takiya CM, Rossi MI, Borojevic R

BACKGROUND: The normal function of white adipose tissue is disturbed in obesity. After weight loss that follows bariatric surgery, ex-obese patients undergo plastic surgery to remove residual tissues and it is not known whether their adipose tissue returns to its original state. The aim of this study was to compare the white adipose tissue composition of ex-obese with control patients with regard to blood vessels and resident mesenchymal stem cells (MSC). METHODS: Quantification of blood vessels was performed on histological sections of adipose tissue stained with hematoxylin and eosin and for von Willebrand antigen. MSC were induced to the adipogenic and osteogenic lineages by specific inductive culture media. Expression of PPARgamma2 was analyzed by reverse transcription polymerase chain reaction. RESULTS: Ex-obese adipose tissue showed a higher number (p = 0.0286) of small (107.3 +/- 22.0) and large (22.5 +/- 6.4) blood vessels, when compared to control patients (42.0 +/- 24.4 and 7.2 +/- 2.2, respectively) and they also occupied a larger area (control versus ex-obese, p = 0.0286). Adipose tissue MSC from both groups of patients expressed PPARgamma2 and were equally able to differentiate to the osteogenic lineage, but ex-obese MSC showed a higher adipogenic potential when induced in vitro (p < 0.05). CONCLUSIONS: The higher number of adipose tissue blood vessels in ex-obese patients explains the excessive bleeding observed during their plastic surgery. The presence of more committed cells to the adipogenic lineage may favor the easy weight regain that occurs in ex-obese patients. These results show that, after extensive weight loss, adipose tissue cell composition was not totally restored.

PMID: 19562421 [PubMed - in process]


Early translation of adipose-derived cell therapy for cardiovascular disease.
August 22, 2009 at 12:51 pm

Related Articles

Early translation of adipose-derived cell therapy for cardiovascular disease.

Cell Transplant. 2009;18(3):245-54

Authors: Sanz-Ruiz R, Fernández-Santos E, Domínguez-Muñoa M, Parma R, Villa A, Fernández L, Sánchez PL, Fernández-Avilés F

Over the past decade, cell therapy has emerged as a new approach to reversing myocardial ischemia. Several types of adult stem cells have been studied in both preclinical and clinical conditions for this purpose: bone marrow cells, circulating cells, and myoblasts. Nevertheless, the quest for the ideal "anti-ischemic" cell is still ongoing. Recently, the existence of a population of stem cells located in adipose tissue (adipose-derived stem cells) has been observed. These are able to differentiate into multiple cell lineages including cardiomyocytic differentiation. In this review we discuss the basic principles of adipose-derived stem cells (types and characteristics, harvesting, and expansion), the initial experimental studies, and the currently ongoing clinical trials.

PMID: 19558773 [PubMed - indexed for MEDLINE]


Resolution of refractory chronic autoimmune thrombocytopenic purpura following mesenchymal stem cell transplantation: a case report.
August 22, 2009 at 12:51 pm

Related Articles

Resolution of refractory chronic autoimmune thrombocytopenic purpura following mesenchymal stem cell transplantation: a case report.

Transplant Proc. 2009 Jun;41(5):1827-30

Authors: Fang B, Song YP, Li N, Li J, Han Q, Zhao RC

Herein we have reported the case of a man with chronic autoimmune thrombocytopenia (AITP) refractory to conventional therapy who underwent T-cell-depleted autologous peripheral blood stem cell (PBSC) transplantation. A complete, steroid-independent, platelet remission was obtained, but unfortunately he subsequently relapsed after 3 months. Finally, we administered human adipose tissue-derived mesenchymal stem cells (AMSC) with resolution to the AITP.

PMID: 19545737 [PubMed - in process]


Stem cell transplant into preimplantation embryo yields myocardial infarction-resistant adult phenotype.
August 22, 2009 at 12:51 pm

Related Articles

Stem cell transplant into preimplantation embryo yields myocardial infarction-resistant adult phenotype.

Stem Cells. 2009 Jul;27(7):1697-705

Authors: Yamada S, Nelson TJ, Behfar A, Crespo-Diaz RJ, Fraidenraich D, Terzic A

Stem cells are an emerging strategy for treatment of myocardial infarction, limited however to postinjury intervention. Preventive stem cell-based therapy to augment stress tolerance has yet to be considered for lifelong protection. Here, pluripotent stem cells were microsurgically introduced at the blastocyst stage of murine embryo development to ensure stochastic integration and sustained organ contribution. Engineered chimera displayed excess in body weight due to increased fat deposits, but were otherwise devoid of obesity-related morbidity. Remarkably, and in sharp contrast to susceptible nonchimeric offspring, chimera was resistant to myocardial infarction induced by permanent coronary occlusion. Infarcted nonchimeric adult hearts demonstrated progressive deterioration in ejection fraction, while age-matched 12-14-months-old chimera recovered from equivalent ischemic insult to regain within one-month preocclusion contractile performance. Electrical remodeling and ventricular enlargement with fibrosis, prominent in failing nonchimera, were averted in the chimeric cohort characterized by an increased stem cell load in adipose tissue and upregulated markers of biogenesis Ki67, c-Kit, and stem cell antigen-1 in the myocardium. Favorable outcome in infarcted chimera translated into an overall benefit in workload capacity and survival. Thus, prenatal stem cell transplant yields a cardioprotective phenotype in adulthood, expanding cell-based indications beyond traditional postinjury applications to include pre-emptive therapy.

PMID: 19544428 [PubMed - in process]


Locally Delivered Growth Factor Enhances the Angiogenic Efficacy of Adipose-Derived Stromal Cells Transplanted to Ischemic Limbs.
August 22, 2009 at 12:51 pm

Related Articles

Locally Delivered Growth Factor Enhances the Angiogenic Efficacy of Adipose-Derived Stromal Cells Transplanted to Ischemic Limbs.

Stem Cells. 2009 May 7;27(8):1976-1986

Authors: Bhang SH, Cho SW, Lim JM, Kang JM, Lee TJ, Yang HS, Song YS, Park MH, Kim HS, Yoo KJ, Jang Y, Langer R, Anderson DG, Kim BS

Ischemia is a potentially fatal medical event that is associated with as many as 30% of all deaths. Stem cell therapy offers significant therapeutic promise, but poor survival following transplantation to ischemic tissue limits its efficacy. Here we demonstrate that nanosphere-mediated growth factor delivery can enhance the survival of transplanted human adipose-derived stromal cells (hADSCs) and secretion of human angiogenic growth factors per cell, and substantially improve therapeutic efficacy of hADSCs. In vitro, in hypoxic (1% oxygen) and serum-deprived conditions that simulate in vivo ischemia, fibroblast growth factor-2 (FGF2) significantly reduced hADSC apoptosis and enhanced angiogenic growth factor secretion. In vivo, hADSCs delivered intramuscularly into ischemic hind limbs in combination with FGF2 resulted in significant improvements in limb survival and blood perfusion, as well as survival of the transplanted hADSCs and secretion of human angiogenic growth factors (i.e., vascular endothelial growth factor, hepatocyte growth factor, and FGF2). Interestingly, the majority of transplanted hADSCs were localized adjacent to the microvessels rather than being incorporated into them, suggesting that their major contribution to angiogenesis might be to increase paracrine secretion of angiogenic growth factors. This study demonstrates the potential of hADSCs in combination with growth factors for use in the treatment of ischemia. STEM CELLS 2009;27:1976-1986.

PMID: 19544425 [PubMed - as supplied by publisher]


Identification of Non-Epithelial Multipotent Cells in the Embryonic Olfactory Mucosa.
August 22, 2009 at 12:51 pm

Related Articles

Identification of Non-Epithelial Multipotent Cells in the Embryonic Olfactory Mucosa.

Stem Cells. 2009 May 21;

Authors: Tomé M, Lindsay SL, Riddell JS, Barnett SC

Olfactory mucosal tissue, a potential source of stem cells, is currently being assessed in the clinic as a candidate tissue for transplant-mediated repair of spinal cord injury. We examined the ability of embryonic rat olfactory mucosal tissue to generate stem cells using culture conditions known to promote neural stem cell proliferation. Primary spheres formed which proliferated and exhibited two main morphologies: i) CNS neurosphere-like, (OM-I) and ii) small, tight spheroid-like (OM-II). The OM-I spheres expressed the neural stem cell marker nestin, but also markers of peripheral glia, neurons and connective tissue. Further studies demonstrated the presence of multipotential mesenchymal-like stem cells within OM-I spheres which differentiated into bone, adipose and smooth muscle cells. In contrast, the OM-II spheres contained mainly cytokeratin-expressing cells. Immunolabelling of rat olfactory tissue with Stro-1, CD90 and CD105 showed the presence of the multipotent mesenchymal cells in the lamina propria while cytokeratin was expressed by the epithelial cells of the olfactory epithelium (OE). In addition a comparable pattern of immunoreactivity was detected in human tissue using Stro-1 and cytokeratin suggesting the presence of similar cells in this tissue. The identification of a non-epithelial multipotent cell in the olfactory mucosa (OM) may explain the varied reports on olfactory stem cell differentiation capacity in vitro and in vivo and illustrates the cellular complexity of this tissue as a potential source of stem cells for transplantation and translation to the clinic.

PMID: 19544421 [PubMed - as supplied by publisher]


Insulin-Secreting Cells from Human Eyelid-Derived Stem Cells Alleviate Type I Diabetes in Immunocompetent Mice.
August 22, 2009 at 12:51 pm

Related Articles

Insulin-Secreting Cells from Human Eyelid-Derived Stem Cells Alleviate Type I Diabetes in Immunocompetent Mice.

Stem Cells. 2009 May 14;27(8):1999-2008

Authors: Kang HM, Kim J, Park S, Kim J, Kim H, Kim KS, Lee EJ, Seo SI, Kang SG, Lee JE, Lim H

Various attempts have been made to develop stem cell-based therapy to alleviate type I diabetes using animal models. However, it has been a question whether human insulin produced from explanted cells is solely responsible for the normoglycemia of diabetic animals. In this study, we isolated neural crest-like stem cells from the human eyelid fat and examined their therapeutic potentials for diabetes. The human eyelid adipose-derived stem cells (HEACs) displayed characteristics of neural crest cells. Using a two-step culture condition combined with nicotinamide, activin, and/or GLP-1, we differentiated HEACs into insulin-secreting cells and examined in vivo effects of differentiated cells by transplantation experiments. Following differentiation in vitro, HEACs released insulin and c-peptide in a glucose-dependent manner. Upon their transplantation under kidney capsules of streptozotocin-treated immunocompetent mice, we observed normalization of hyperglycemia in 10 of 20 recipient mice until sacrifice after 2 months. Only the human, but not the mouse, insulin and c-peptide were detected in the blood of recipient mice. Removal of the kidneys transplanted with HEACs resulted in a sharp increase of blood glucose level. Removed kidney tissues showed distinct expression of various human genes including insulin, and colocalization of the human insulin and the human nuclear protein in many cells. However, they showed diminished or null expression of some immune-related genes. In conclusion, human insulin alone produced from eyelid-derived stem cells following differentiation into insulin-secreting cells and transplantation could normalize type I diabetes in mice. STEM CELLS 2009;27:1999-2008.

PMID: 19544420 [PubMed - as supplied by publisher]


Enhanced ITM2A expression inhibits chondrogenic differentiation of mesenchymal stem cells.
August 22, 2009 at 12:51 pm

Related Articles

Enhanced ITM2A expression inhibits chondrogenic differentiation of mesenchymal stem cells.

Differentiation. 2009 Jun 19;

Authors: Boeuf S, Börger M, Hennig T, Winter A, Kasten P, Richter W

Mesenchymal stem cells (MSC) from bone marrow or adipose tissue (ASC) are broadly discussed as a cell population able to support cartilage regeneration and thus represent interesting candidates for cell-based tissue engineering in cartilage. ASC could represent an easily accessible and therefore particularly suitable source of cells. Their chondrogenic differentiation potential is, however, lower than that of MSC. The aim of this work was to characterise ASC in comparison to MSC in order to identify genes which may be involved in mechanisms causing the altered chondrogenic potential of ASC. Representational difference analysis was used to identify genes with higher expression in undifferentiated ASC than in MSC. Expression levels of identified genes were confirmed by real-time RT-PCR. Integral membrane protein 2A (ITM2A) was higher expressed in expanded ASC than in MSC in a donor-independent manner. During early chondrogenic differentiation in spheroid cultures ITM2A levels remained low in MSC and a transient down-regulation occurred in ASC correlating with successful chondrogenesis. Persisting ITM2A levels were found in non-differentiating ASC. Consistent with this finding, forced expression of ITM2A in the mouse mesenchymal stem cell line C3H10T1/2 prevented chondrogenic induction. In conclusion, ITM2A may in early stages of differentiation be associated with an inhibition of the initiation of chondrogenesis and elevated expression of ITM2A in ASC may therefore be linked to the poorer chondrogenic differentiation potential of these cells.

PMID: 19541402 [PubMed - as supplied by publisher]


[Use of mesenchymal stem cells from adult bone marrow for injured tissue repair]
August 22, 2009 at 12:51 pm

Related Articles

[Use of mesenchymal stem cells from adult bone marrow for injured tissue repair]

Orv Hetil. 2009 Jul 5;150(27):1259-65

Authors: Salamon A, Toldy E

Mesenchymal stem cells are known as being multipotent and exhibit the potential for differentiation into different cells/tissue lineages, including cartilage, bone, adipose tissue, tendon, and ligament. These pluripotent mesenchymal progenitor cells are denoted as stromal or mesenchymal stem cells. Bone marrow contains two main cell types: hematopoietic cells and stromal cells. The stem cells for non hematopoietic tissues are referred as mesenchymal cells because of their ability to differentiate as mesenchymal or stromal cells. Mesenchymal cells are easily obtainable from bone marrow by means of minimally invasive approach and can be expanded in culture and permitted to differentiate into the desired lineage. The differentiation can be reached by the application of bioactive signaling molecules, specific growth factors. The transforming growth factor beta (TGF-beta) superfamily member proteins such as the bone morphogenetic proteins (BMP-s) are the most important factors of chondrogenic and osteogenic differentiation of mesenchymal stem cells. From the series of recently identified factors, BMP 2,4 and 7 may play an important role in chondrogenic and osteogenic differentiation proteins. Little is known, however, about the signaling pathway involved in tenogenesis of mesenchymal stem cells, but there are some encouraging data about fibroblastic differentiation. The success of growth factor therapy needs a delivery system with biomaterials. Mesenchymal stem cells have become promising vehicles for gene therapy, cell therapy and tissue engineering. In present review, authors deal with the experimental investigations and with the clinical application of the adult bone marrow derived mesenchymal stem cells with bioactive molecules, growth factors.

PMID: 19531459 [PubMed - indexed for MEDLINE]


The wound-healing and antioxidant effects of adipose-derived stem cells.
August 22, 2009 at 12:51 pm

Related Articles

The wound-healing and antioxidant effects of adipose-derived stem cells.

Expert Opin Biol Ther. 2009 Jul;9(7):879-87

Authors: Kim WS, Park BS, Sung JH

BACKGROUND: The aim of tissue engineering is to repair and regenerate damaged organs using a combination of cells, biomaterials and growth factors. Mesenchymal stem cells within the stromal-vascular fraction of subcutaneous adipose tissue, that is adipose-derived stem cells (ADSCs) have been used in skin repair with satisfactory results. The production and secretion of growth factors has been reported to be an essential function of ADSCs, and diverse regenerative effects of ADSCs in the skin have been demonstrated. OBJECTIVE: Recent research developments concerning the wound-healing and antioxidant effects of ADSCs are briefly described. METHODS: Various experimental results regarding the wound-healing and antioxidant effect of ADSCs are introduced, and the mechanisms and identification of active proteins involved in these function are further discussed. RESULTS/CONCLUSION: Evidence of ADSC differentiation of skin has not been reported in vivo, but ADSCs accelerate wound-healing and exhibit antioxidant effects under various conditions. The wound-healing and antioxidant effects of ADSCs are mainly mediated by the activation of dermal fibroblasts and keratinocytes via the paracrine mechanism. Since ADSCs are easily obtained in large quantities and have an advantage over other stem cell sources, ADSCs and their secretory factors show promise for use in skin repair and regeneration.

PMID: 19522555 [PubMed - in process]


Maximizing results for lipofilling in facial reconstruction.
August 22, 2009 at 12:51 pm

Related Articles

Maximizing results for lipofilling in facial reconstruction.

Clin Plast Surg. 2009 Jul;36(3):487-92

Authors: Barret JP, Sarobe N, Grande N, Vila D, Palacin JM

Lipostructure (also known as structural fat grafts, lipofilling, or fat grafting) has become a technique with a good reputation and reproducible results. The application of this technology in patients undergoing reconstruction is a novel surgical alternative. Obtaining good results in this patient population is very difficult, but the application of small fat grafts with a strict Coleman technique produces long-term cosmetic effects. Adult-derived stem cells have been pointed out as important effectors of this regenerative technology, and future research should focus in this direction.

PMID: 19505616 [PubMed - indexed for MEDLINE]


Cartilage regeneration of adipose-derived stem cells in a hybrid scaffold from fibrin-modified PLGA.
August 22, 2009 at 12:51 pm

Related Articles

Cartilage regeneration of adipose-derived stem cells in a hybrid scaffold from fibrin-modified PLGA.

Cell Transplant. 2009;18(2):159-70

Authors: Wei Y, Hu H, Wang H, Wu Y, Deng L, Qi J

Adipose-derived stem cells (ASCs) appear to be a useful stem cell population, which has been shown to possess multipotentiality. The aim of this study was to evaluate the utility of ASCs in tissue-engineered cartilage using a hybrid scaffold from fibrin-modified PLGA scaffold. ASCs were isolated from rabbit adipose tissue. The PLGA scaffold was prepared by low-temperature deposition technology and the hybrid scaffold was fabricated by a freeze-drying method. When ASCs were seeded onto fibrin-modified PLGA scaffold in vitro, enhanced cellular viability was observed compared to unmodified PLGA scaffold. The analysis of proteoglycan and collagen II revealed that fibrin-modified scaffold succeeded in inducing ASCs to differentiate into chondrocytes in vitro. A preliminary study on cartilage regeneration was also performed in vivo. Observation of histology and immunoblotting demonstrated that ASCs containing the hybrid scaffold promoted cartilage regeneration in the defects of articular cartilage much better than other groups. These results indicated that ASCs containing the hybrid scaffold are a more effective way to potentially enhance articular cartilage regeneration.

PMID: 19499704 [PubMed - indexed for MEDLINE]


Clinical study of the efficiency of combined cell transplant on the basis of multipotent mesenchymal stromal adipose tissue cells in patients with pronounced deficit of the maxillary and mandibulary bone tissue.
August 22, 2009 at 12:51 pm

Related Articles

Clinical study of the efficiency of combined cell transplant on the basis of multipotent mesenchymal stromal adipose tissue cells in patients with pronounced deficit of the maxillary and mandibulary bone tissue.

Bull Exp Biol Med. 2008 Oct;146(4):522-5

Authors: Kulakov AA, Goldshtein DV, Grigoryan AS, Rzhaninova AA, Alekseeva IS, Arutyunyan IV, Volkov AV

The use of synthetic osteoplastic materials not always provides the required amount of the bone tissue. Transplantation of tissue-engineering constructs containing osteogenic precursor cells can be an alternative high-technology implantation method. Here we present the results of a pilot clinical study demonstrating safety of this method, accelerated healing of the operation wound, formation of young bone tissue after transplantation, and the possibility of mounting implants after 3 months in case of sufficient amount of the bone for primary fixation.

PMID: 19489333 [PubMed - indexed for MEDLINE]


Therapeutic potential and related signal pathway of adipose-derived stem cell transplantation for rat liver injury.
August 22, 2009 at 12:51 pm

Related Articles

Therapeutic potential and related signal pathway of adipose-derived stem cell transplantation for rat liver injury.

Hepatol Res. 2009 Mar 25;

Authors: Liang L, Ma T, Chen W, Hu J, Bai X, Li J, Liang T

Aim: Liver transplantation is the only currently effective therapy for end-stage chronic liver disease and severe acute liver failure, but its use is limited by high cost and a shortage of allografts. Here we explored the effectiveness of transplanting adipose-derived stem cells (ADSCs) into rats with experimentally induced liver injury. Methods: ADSCs obtained from rats were hepatogenic induced in vitro with MAPK pathways inhibitors preconditioning. In vivo, ADSCs were transplanted into rats via different routes and serum liver function markers from post-operative rats were tested. Results: When grown in adipogenic induction medium, ADSCs were able to differentiate into adipocytes. In hepatogenic induction medium, ADSCs were able to differentiate into hepatocyte-like cells, with appropriate changes in morphology and appropriately elevated expression of hepatocyte-specific markers. ERK1/2 phosphorylation activity was also significantly upregulated during the hepatogenic differentiation process, and was blocked by the ERK/MAPK pathway-specific inhibitor PD98059. In a rat liver injury model, intravenously injected ADSCs successfully engrafted into recipient livers. We found that injection via the hepatic portal vein was more efficient than via the dorsal vein of the penis. ADSC transplantation into damaged livers significantly decreased the level of serum liver enzymes such as alanine aminotransferase and aspartate aminotransferase, and improved serum albumin level. Both the number of engrafted cells and the improvement of liver function reached a peak two weeks after transplantation. Conclusion: Transplanted ADSCs appear to be therapeutically effective in the rat liver injury model, which may ultimately provide a therapeutic alternative to liver transplantation in human patients.

PMID: 19473439 [PubMed - as supplied by publisher]


Adipose-derived stem cells for tissue repair and regeneration: ten years of research and a literature review.
August 22, 2009 at 12:51 pm

Related Articles

Adipose-derived stem cells for tissue repair and regeneration: ten years of research and a literature review.

J Nippon Med Sch. 2009 Apr;76(2):56-66

Authors: Mizuno H

Stem cell based therapies for the repair and regeneration of various tissues and organs offer a paradigm shift that may provide alternative therapeutic solutions for a number of diseases. Although embryonic stem cells and induced pluripotent stem cells are theoretically highly beneficial, there are various limitations to their use imposed by cell regulations, ethical considerations, and genetic manipulation. Adult stem cells, on the other hand, are more easily available, with neither ethical nor immunoreactive considerations, as long as they are of autologous tissue origin. Much research has focused on mesenchymal stem cells isolated from bone marrow stroma which have been shown to possess adipogenic, osteogenic, chondrogenic, myogenic, and neurogenic potential in vitro. However bone marrow procurement is extremely painful for patients and yields low numbers of harvested cells. When compared with bone marrow-derived mesenchymal stem cells, adipose-derived stem cells are equally capable of differentiating into cells and tissues of mesodermal origin. Because human adipose tissue is ubiquitous and easily obtainable in large quantities under local anesthesia with little patient discomfort, it may provide an alternative source of stem cells for mesenchymal tissue regeneration and engineering. Based on our previous experimental findings, this review highlights the molecular characteristics, the potential for differentiation, the potential for wound healing, and the future role of adipose-derived stem cells in cell-based therapies and tissue engineering.

PMID: 19443990 [PubMed - indexed for MEDLINE]


Robust functional vascular network formation in vivo by cooperation of adipose progenitor and endothelial cells.
August 22, 2009 at 12:51 pm

Related Articles

Robust functional vascular network formation in vivo by cooperation of adipose progenitor and endothelial cells.

Circ Res. 2009 Jun 19;104(12):1410-20

Authors: Traktuev DO, Prater DN, Merfeld-Clauss S, Sanjeevaiah AR, Saadatzadeh MR, Murphy M, Johnstone BH, Ingram DA, March KL

Rapid induction and maintenance of blood flow through new vascular networks is essential for successfully treating ischemic tissues and maintaining function of engineered neo-organs. We have previously shown that human endothelial progenitor cells (EPCs) form functioning vessels in mice, but these are limited in number and persistence; and also that human adipose stromal cells (ASCs) are multipotent cells with pericytic properties which can stabilize vascular assembly in vitro. In this study, we tested whether ASCs would cooperate with EPCs to coassemble vessels in in vivo implants. Collagen implants containing EPCs, ASCs, or a 4:1 mixture of both were placed subcutaneously into NOD/SCID mice. After a range of time periods, constructs were explanted and evaluated with regard to vascular network assembly and cell fate; and heterotypic cell interactions were explored by targeted molecular perturbations. The density and complexity of vascular networks formed by the synergistic dual-cell system was many-fold higher than found in implants containing either ASCs or EPCs alone. Coimplantation of ASCs and EPCs with either pancreatic islets or adipocytes produced neoorgans populated by these parenchymal cells, as well as by chimeric human vessels conducting flow. This study is the first to demonstrate prompt and consistent assembly of a vascular network by human ASCs and endothelial cells and vascularization by these cells of parenchymal cells in implants. Mixture of these 2 readily available, nontransformed human cell types provides a practical approach to tissue engineering, therapeutic revascularization, and in vivo studies of human vasculogenesis.

PMID: 19443841 [PubMed - indexed for MEDLINE]


Adult stem cells as an alternative source of multipotential (pluripotential) cells in regenerative medicine.
August 22, 2009 at 12:51 pm

Related Articles

Adult stem cells as an alternative source of multipotential (pluripotential) cells in regenerative medicine.

Curr Stem Cell Res Ther. 2009 May;4(2):107-17

Authors: Kuçi S, Kuçi Z, Latifi-Pupovci H, Niethammer D, Handgretinger R, Schumm M, Bruchelt G, Bader P, Klingebiel T

Embryonic stem cells are by definition the master cells capable of differentiating into every type of cells either in vitro or in vivo. Several lines of evidence suggest, however, that adult stem cells and even terminally differentiated somatic cells under appropriate microenvironmental cues are able to be reprogrammed and contribute to a much wider spectrum of differentiated progeny than previously anticipated. This has been demonstrated by using tissue- specific stem cells, which like embryonic stem cells do not express CD45 as an exclusive hematopoietic marker (skin, adipose, cord blood and bone marrow- derived stem cells). On the other side, there is a great number of reports which demonstrate that hematopoietic cells (CD45+) from different sources (peripheral blood, cord blood, bone marrow) are also able to cross the tissue boundaries and give rise to the cells of the other germ layers. Herein we discuss the differentiation and reprogramming potential of both hematopoietic and non- hematopoietic stem cells along endodermal, mesodermal and neuroectodermal lineage and their importance for regenerative medicine.

PMID: 19442195 [PubMed - indexed for MEDLINE]


Practical considerations concerning the use of stem cells for peripheral nerve repair.
August 22, 2009 at 12:51 pm

Related Articles

Practical considerations concerning the use of stem cells for peripheral nerve repair.

Neurosurg Focus. 2009 Feb;26(2):E2

Authors: Walsh S, Midha R

In this review the authors intend to demonstrate the need for supplementing conventional repair of the injured nerve with alternative therapies, namely transplantation of stem or progenitor cells. Although peripheral nerves do exhibit the potential to regenerate axons and reinnervate the end organ, outcome following severe nerve injury, even after repair, remains relatively poor. This is likely because of the extensive injury zone that prevents axon outgrowth. Even if outgrowth does occur, a relatively slow growth rate of regeneration results in prolonged denervation of the distal nerve. Whereas denervated Schwann cells (SCs) are key players in the early regenerative success of peripheral nerves, protracted loss of axonal contact renders Schwann cells unreceptive for axonal regeneration. Given that denervated Schwann cells appear to become effete, one logical approach is to support the distal denervated nerve environment by replacing host cells with those derived exogenously. A number of different sources of stem/precursor cells are being explored for their potential application in the scenario of peripheral nerve injury. The most promising candidate, transplant cells are derived from easily accessible sources such as the skin, bone marrow, or adipose tissue, all of which have demonstrated the capacity to differentiate into Schwann cell-like cells. Although recent studies have shown that stem cells can act as promising and beneficial adjuncts to nerve repair, considerable optimization of these therapies will be required for their potential to be realized in a clinical setting. The authors investigate the relevance of the delivery method (both the number and differentiation state of cells) on experimental outcomes, and seek to clarify whether stem cells must survive and differentiate in the injured nerve to convey a therapeutic effect. As our laboratory uses skin-derived precursor cells (SKPCs) in various nerve injury paradigms, we relate our findings on cell fate to other published studies to demonstrate the need to quantify stem cell survival and differentiation for future studies.

PMID: 19435443 [PubMed - indexed for MEDLINE]


Mesenchymal stem cells for therapeutic purposes.
August 22, 2009 at 12:51 pm

Related Articles

Mesenchymal stem cells for therapeutic purposes.

Transplantation. 2009 May 15;87(9 Suppl):S49-53

Authors: Sensebé L, Bourin P

Mesenchymal stem cells (MSC) are multipotent adult stem cells harboring a wide range of differentiations and non-human leukocyte antigen-restricted immunosuppressive properties that lead to an increasing use of MSC in immunomodulation and in regenerative medicine. To produce MSC, definitive standards are still lacking. Whatever the starting material used (e.g., bone marrow, adipose tissue, or cord blood), numerous parameters including cell plating density, number of passages, and culture medium, play a major role in the culture process and have to be determined. To date, the different production processes have been effective, and based on phenotypic analysis and differentiation potential, a first set of simple controls have been defined. However, controls of the final product should provide precise data on efficacy and safety. The next challenge will be to develop production processes that reach good manufacturing practices goals and to define more accurate control methods of cultivated MSC.

PMID: 19424006 [PubMed - indexed for MEDLINE]


Human adipose-derived stem cells: isolation, characterization and applications in surgery.
August 22, 2009 at 12:51 pm

Related Articles

Human adipose-derived stem cells: isolation, characterization and applications in surgery.

ANZ J Surg. 2009 Apr;79(4):235-44

Authors: Locke M, Windsor J, Dunbar PR

The ideal stem cell for use in functional tissue engineering needs to be abundantly available, harvested with minimal morbidity, differentiated reliably down various pathways and able to be transplanted safely and efficaciously. Adult human adipose tissue contains a population of mesenchymal stem cells, termed 'adipose-derived stem cells' (ASC), which seem to fulfil most, if not all, of these criteria. ASC can be harvested readily, safely, and in relative abundance by modern liposuction techniques. They are capable of differentiating into other mesenchymal tissue types, including adipocytes, chondrocytes, myocytes and osteoblasts. They also show angiogenic properties, with recent evidence of a potential role in healing radiotherapy-damaged tissue, possibly due to their secretion of vascular endothelial growth factor. Similarly, they may have a role in healing chronic wounds, and as such are being investigated in phase 1 trials for their ability to aid healing of recurrent Crohn's fistulae. Subsequently they have a wide range of potential clinical uses in all fields of surgery. This article reviews the current and potential clinical applications of ASC in relation to surgery, as well as methods for their isolation, differentiation and molecular characterization.

PMID: 19432707 [PubMed - indexed for MEDLINE]


Characterization of zinc-releasing three-dimensional bioactive glass scaffolds and their effect on human adipose stem cell proliferation and osteogenic differentiation.
August 22, 2009 at 12:51 pm

Related Articles

Characterization of zinc-releasing three-dimensional bioactive glass scaffolds and their effect on human adipose stem cell proliferation and osteogenic differentiation.

Acta Biomater. 2009 Apr 16;

Authors: Haimi S, Gorianc G, Moimas L, Lindroos B, Huhtala H, Räty S, Kuokkanen H, Sándor GK, Schmid C, Miettinen S, Suuronen R

While the addition of zinc ions to bioactive ceramics has been shown to enhance the proliferation and osteogenic differentiation of osteoblast-like cells, contradictory results have been found. Therefore, the effect of zinc-releasing ceramics on cell proliferation and differentiation into osteogenic lineages requires further clarification. The aim of this study was to evaluate the effects of zinc addition on the degradation profile of three-dimensional bioactive glass scaffold, and on the proliferation and osteogenesis of human adipose stem cells (hASCs) in these scaffolds. Bioactive glass scaffolds containing Na(2)O, K(2)O, MgO, CaO, B(2)O(3), TiO(2), P(2)O(5) and SiO(2) were prepared. The degradation was evaluated by weight loss measurement, scanning electron microscopy and elemental analysis. The degradation profile of bioactive glass was shown to slow down with the addition of zinc. Qualitative live/dead staining showed that zinc addition to bioactive glass inhibits cell spreading and proliferation of hASCs. However, zinc addition had no significant effect on DNA content, alkaline phosphatase activity and osteopontin concentration of hASCs when measured quantitatively. Our results suggest that the possible stimulatory effect of addition of zinc on hASC proliferation and osteogenesis was not detected because addition of zinc slowed down the degradation rate of the studied bioactive glass scaffolds.

PMID: 19428318 [PubMed - as supplied by publisher]


Adipose stem cell side population in the mouse.
August 22, 2009 at 12:51 pm

Related Articles

Adipose stem cell side population in the mouse.

J Tissue Eng Regen Med. 2009 Aug;3(6):430-41

Authors: Ramos TV, Wang T, Maki CB, Pascual M, Izadyar F

Adipose tissue has become a reliable source of adult stem cells, which appear to possess a yet-undetermined degree of plasticity. With the difficulties associated with harvesting adult bone marrow stem cells, adipose tissue may represent a valuable and easily acquired source of stem cells. Stem cells have been identified using the DNA binding dye Hoechst 33342 and flow cytometry in various tissues known as the side population (SP). The present study shows, for the first time, the presence of side population stem cells in adult adipose tissues. Flow cytometric identification and isolation of this subpopulation of stem cells revealed that in the mouse there are 2.5% of adipose SP cells within the stromal vascular fraction of adipose tissue. In culture, mouse adipose SP cells showed the capacity to undergo in vitro differentiation into osteogenic, chondrogenic and adipogenic lineages. In NOD/SCID mice, freshly sorted mouse adipose SP cells were able to engraft and assist in wound healing. This animal model study showed that adipose SP cells were able to regenerate epithelial layers and connective tissue with minor scar formation. The ability of this novel cell population within adipose tissue to undergo directional differentiation in vitro and to regenerate skin in vivo has potential impact for uses in surgical dermal applications.

PMID: 19415785 [PubMed - in process]


Ex vivo expansion of adipose tissue-derived stem cells in spinner flasks.
August 22, 2009 at 12:51 pm

Related Articles

Ex vivo expansion of adipose tissue-derived stem cells in spinner flasks.

Biotechnol J. 2009 Aug;4(8):1198-209

Authors: Zhu Y, Liu T, Song K, Fan X, Ma X, Cui Z

Recent reports indicate that adipose tissue is a novel source of multipotent stem cells that can be used in cell therapy and tissue engineering. However, using the traditional cultivation of adipose tissue-derived stem cells (ADSCs), it is hard to meet the needs of clinical applications. To obtain a large number of ADSCs while retaining their stemness, we seeded ADSCs in collagen/chitosan scaffolds and compared the proliferation of ADSCs in a 3-D static environment in dishes and a 3-D dynamic environment in spinner flask. The growth dynamic parameters of ADSCs were examined using a CCK-8 kit every other day, and the variations of glucose and lactic acid concentrations were analyzed every day. After 14 days, the cells were observed under a scanning electron microscope. The surface markers (CD29, CD34, CD44, CD45, CD73, CD105, CD166 and HLA-DR), the specific transcription factors (Nanog, Oct-4, Sox-2 and Rex-1) and the multi-differentiation potential (adipogenic, osteogenic and chondrogenic) were also assayed to identify the stemness of expanded cells. The results showed that the cells in scaffolds in spinner flask could be expanded by more than 26 times, and they presented better morphology and vitality and stronger differentiation ability than the cells cultivated in scaffolds statically. All the cells maintained stem cell characteristics after proliferation. Therefore, spinner flask cultivation is an easy-to-use, inexpensive system for expanding ADSCs in 3-D scaffolds.

PMID: 19404993 [PubMed - in process]


In vitro Adipogenic Differentiation of Preadipocytes Varies with Differentiation Stimulus, Culture Dimensionality, and Scaffold Composition.
August 22, 2009 at 12:51 pm

Related Articles

In vitro Adipogenic Differentiation of Preadipocytes Varies with Differentiation Stimulus, Culture Dimensionality, and Scaffold Composition.

Tissue Eng Part A. 2009 Apr 29;

Authors: Stacey DH, Hanson SE, Lahvis G, Gutowski KA, Masters KS

Despite the rapidly-growing body of work on stem cell-based adipose tissue engineering, there remains much to be learned about the role of the scaffold and culture environments in directing the adipogenic differentiation of cells. The present study examined how various culture environments and differentiation stimuli (traditional differentiation medium and co-culture with mature adipocytes) impacted the adipogenic differentiation of human preadipocytes, with studies progressing from 2-D to 3-D in vitro. Assays for adipogenic markers (leptin, adiponectin, glycerol) and Oil Red O staining were used to assess differentiation. After 16 days of 2-D culture, adipogenesis was substantially greater when preadipocytes were co-cultured with adipocytes rather than treated with differentiation medium. In a 3-D in vitro environment, the production of adipogenic markers was significantly elevated relative to 2-D conditions, and the co-culture condition continued to stimulate greater adipogenesis. Alterations in 3-D scaffold physical properties had only a minimal effect on the function of mature adipocytes, but significantly impacted the ability of preadipocytes to undergo adipogenic differentiation in vitro. Specifically, two scaffolds (3.4 and 8.0 kDa polyethylene glycol-based hydrogels) emerged as most supportive of adipogenic differentiation. These alterations in scaffold environment and in media conditions, particularly the application of adipocyte/preadipocyte co-culture methods in lieu of traditional differentiation medium, may provide further means for optimizing adipogenic outcomes in vitro and in vivo.

PMID: 19402786 [PubMed - as supplied by publisher]


Molecular switching of osteoblastogenesis versus adipogenesis: implications for targeted therapies.
August 22, 2009 at 12:51 pm

Related Articles

Molecular switching of osteoblastogenesis versus adipogenesis: implications for targeted therapies.

Expert Opin Ther Targets. 2009 May;13(5):593-603

Authors: Takada I, Kouzmenko AP, Kato S

Osteoblasts and adipocytes differentiate from a common precursor, the pluripotent mesenchymal stem cell (MSC) found in bone marrow (BMSC) and adipose tissue (AD-MSC). Numerous transcription factors and multiple extracellular and intracellular signals regulating adipogenesis and osteoblastogenesis have been identified and analyzed. Significantly, inducers of differentiation towards one lineage may inhibit cell differentiation into an alternative lineage. For example, the canonical Wnt/beta-catenin pathway induces osteoblastogenesis and inhibits adipogenesis, whereas the peroxisome proliferator activated receptor-gamma (PPAR-gamma) is a prime inducer of adipogenesis and, as shown in recent studies, inhibits osteoblastogenesis. We have identified two signaling pathways that switch the cell fate decision from adipocytes to osteoblasts by suppressing the transactivation function of PPAR-gamma. In the first pathway, the TNF-alpha- or IL-1-induced TAK1/TAB1/NIK signaling cascade attenuates PPAR-gamma-mediated adipogenesis by inhibiting the binding of PPAR-gamma to the DNA response element. The second is the noncanonical Wnt pathway through the CaMKII-TAK1/TAB2-NLK (nemo-like kinase) signaling cascade. Specifically, Wnt-5a-induced phosphorylation of NLK triggers formation of a complex with the histone methyltransferase SETDB1 (SET domain, bifurcated 1) that represses PPAR-gamma transactivation through histone H3-K9 methylation at the target genes. Thus, two signaling cascades promote osteoblastic differentiation from MSC through two distinct modes of PPAR-gamma transrepression.

PMID: 19397478 [PubMed - indexed for MEDLINE]


Non-expanded adipose stromal vascular fraction cell therapy for multiple sclerosis.
August 22, 2009 at 12:51 pm

Related Articles

Non-expanded adipose stromal vascular fraction cell therapy for multiple sclerosis.

J Transl Med. 2009;7:29

Authors: Riordan NH, Ichim TE, Min WP, Wang H, Solano F, Lara F, Alfaro M, Rodriguez JP, Harman RJ, Patel AN, Murphy MP, Lee RR, Minev B

The stromal vascular fraction (SVF) of adipose tissue is known to contain mesenchymal stem cells (MSC), T regulatory cells, endothelial precursor cells, preadipocytes, as well as anti-inflammatory M2 macrophages. Safety of autologous adipose tissue implantation is supported by extensive use of this procedure in cosmetic surgery, as well as by ongoing studies using in vitro expanded adipose derived MSC. Equine and canine studies demonstrating anti-inflammatory and regenerative effects of non-expanded SVF cells have yielded promising results. Although non-expanded SVF cells have been used successfully in accelerating healing of Crohn's fistulas, to our knowledge clinical use of these cells for systemic immune modulation has not been reported. In this communication we discuss the rationale for use of autologous SVF in treatment of multiple sclerosis and describe our experiences with three patients. Based on this rationale and initial experiences, we propose controlled trials of autologous SVF in various inflammatory conditions.

PMID: 19393041 [PubMed - indexed for MEDLINE]


The 4th dimension and adult stem cells: Can timing be everything?
August 22, 2009 at 12:51 pm

Related Articles

The 4th dimension and adult stem cells: Can timing be everything?

J Cell Biochem. 2009 Jul 1;107(4):569-78

Authors: Gimble JM, Floyd ZE, Bunnell BA

The rotation of the earth on its axis influences the physiology of all organisms. A highly conserved set of genes encoding the core circadian regulatory proteins (CCRP) has evolved across species. The CCRP acts through transcriptional and post-transcriptional mechanisms to direct the oscillatory expression of genes essential for key metabolic events. In addition to the light:dark cycle, the CCRP expression can be entrained by changes in feeding and physical activity patterns. While mammalian CCRP were originally associated with the central clock located within the suprachiasmatic nucleus of the brain, there is a growing body of evidence documenting the presence of the CCRP in peripheral tissues. It is now evident that the CCRP play a role in regulating the proliferation, differentiation, and function of adult stem cells in multiple organs. This concise review highlights findings concerning the role of the CCRP in modulating the adult stem cell activities. Although the manuscript focuses on hematopoietic stem cells (HSCs), bone marrow-derived mesenchymal stem cells (BMSCs), adipose-derived stem cells (ASCs) and cancer stem cells, it is likely that the contribution of the CCRP merits consideration and evaluation in all stem cell pathways.

PMID: 19384905 [PubMed - in process]


Comparative chondrogenesis of human cell sources in 3D scaffolds.
August 22, 2009 at 12:51 pm

Related Articles

Comparative chondrogenesis of human cell sources in 3D scaffolds.

J Tissue Eng Regen Med. 2009 Jul;3(5):348-360

Authors: Seda Tıglı R, Ghosh S, Laha MM, Shevde NK, Daheron L, Gimble J, Gümüşderelioglu M, Kaplan DL

Cartilage tissue can be engineered by starting from a diversity of cell sources, including stem-cell based and primary cell-based platforms. Selecting an appropriate cell source for the process of cartilage tissue engineering or repair is critical and challenging, due to the variety of cell options available. In this study, cellular responses of isolated human chondrocytes, human embryonic stem cells and mesenchymal stem cells (MSCs) derived from three sources, human embryonic stem cells, bone marrow and adipose tissue, were assessed for chondrogenic potential in 3D culture. All cell sources were characterized by FACS analysis to compare expression of some surface markers. The cells were differentiated in two different biomaterial matrices, silk and chitosan scaffolds, in the presence and absence of bone morphogenetic protein 6 (BMP6), along with the standard chondrogenic differentiating factors. Embryonic stem cells-derived MSCs showed unique characteristics, with preserved chondrogenic phenotype in both scaffolds with regard to chondrogenesis, as determined by real time RT-PCR, histological and microscopical analyses. After 4 weeks of cultivation, embryonic stem cells-derived MSCs were promising for chondrogenesis, particularly in the silk scaffolds with BMP6. The results suggest that cell source differences are important to consider with regard to chondrogenic outcomes, and among the variables addressed here the human embryonic stem cells-derived MSCs were the preferred cell source. Copyright (c) 2009 John Wiley & Sons, Ltd.

PMID: 19382119 [PubMed - as supplied by publisher]

 

This email was sent to agupta1213+termsc@gmail.comManage Your Account
Don't want to receive this feed any longer? Unsubscribe here.

No comments: