Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Am J Physiol Renal Physiol ; 318(4): F861-F869, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32003597

ABSTRACT

Serum soluble Fas (sFas) levels are associated with erythropoietin (Epo) hyporesponsiveness in patients with chronic kidney disease (CKD). Whether sFas could predict the need for erythropoiesis-stimulating agent (ESA) usage and its influence in erythropoiesis remain unclear. We evaluated the relation between sFas and ESA therapy in patients with CKD with anemia and its effect on erythropoiesis in vitro. First, we performed a retrospective cohort study with 77 anemic patients with nondialysis CKD. We performed in vitro experiments to investigate whether sFas could interfere with the behavior of hematopoietic stem cells (HSCs). HSCs were isolated from umbilical cord blood and incubated with recombinant sFas protein in a dose-dependent manner. Serum sFas positively correlated with Epo levels (r = 0.30, P = 0.001) but negatively with hemoglobin (r = -0.55, P < 0.001) and glomerular filtration rate (r = -0.58, P < 0.001) in patients with CKD at baseline. Elevated sFas serum levels (4,316 ± 897 vs. 2,776 ± 749, P < 0.001) with lower estimated glomerular filtration rate (26.2 ± 10.1 vs. 33.5 ± 14.3, P = 0.01) and reduced hemoglobin concentration (11.1 ± 0.9 vs. 12.5 ± 1.2, P < 0.001) were identified in patients who required ESA therapy compared with patients with non-ESA. Afterward, we detected that the sFas level was slight correlated with a necessity of ESA therapy in patients with nondialysis CKD and anemia. In vitro assays demonstrated that the erythroid progenitor cell frequency negatively correlated with sFas concentration (r = -0.72, P < 0.001). There was decreased erythroid colony formation in vitro when CD34+ HSCs were incubated with a higher concentration of sFas protein (1.56 ± 0.29, 4.33 ± 0.53, P < 0.001). Our findings suggest that sFas is a potential predictor for ESA therapy in patients with nondialysis CKD and that elevated sFas could affect erythropoiesis in vitro.


Subject(s)
Anemia/blood , Erythropoiesis , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Renal Insufficiency, Chronic/complications , fas Receptor/blood , Adult , Aged , Anemia/diagnosis , Anemia/drug therapy , Anemia/etiology , Biomarkers/blood , Brazil , Cells, Cultured , Clinical Decision-Making , Databases, Factual , Erythropoiesis/drug effects , Erythropoietin/blood , Female , Hematinics/therapeutic use , Hematopoietic Stem Cells/drug effects , Hemoglobins/metabolism , Humans , Male , Middle Aged , Multipotent Stem Cells/drug effects , North Carolina , Patient Selection , Predictive Value of Tests , Recombinant Proteins/pharmacology , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diagnosis , Retrospective Studies
2.
Cell Tissue Res ; 373(2): 421-438, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29651556

ABSTRACT

Fetal onset hydrocephalus and abnormal neurogenesis are two inseparable phenomena turned on by a cell junction pathology first affecting neural stem/progenitor cells (NSPCs) and later the multiciliated ependyma. The neurological impairment of children born with hydrocephalus is not reverted by derivative surgery. NSPCs and neurosphere (NE) grafting into the cerebrospinal fluid (CSF) of hydrocephalic fetuses thus appears as a promising therapeutic procedure. There is little information about the cell lineages actually forming the NE as they grow throughout their days in vitro (DIV). Furthermore, there is no information on how good a host the CSF is for grafted NE. Here, we use the HTx rat, a model with hereditary hydrocephalus, with the mutation expressed in about 30% of the litter (hyHTx), while the littermates develop normally (nHTx). The investigation was designed (i) to establish the nature of the cells forming 4 and 6-DIV NE grown from NSPCs collected from PN1/nHTx rats and (ii) to study the effects on these NEs of CSF collected from nHTx and hyHTx. Immunofluorescence analyses showed that 90% of cells forming 4-DIV NEs were non-committed multipotential NSPCs, while in 6-DIV NE, 40% of the NSPCs were already committed into neuronal, glial and ependymal lineages. Six-DIV NE further cultured for 3 weeks in the presence of fetal bovine serum, CSF from nHTx or CSF from hyHTx, differentiated into neurons, astrocytes and ßIV-tubulin+ multiciliated ependymal cells that were joined together by adherent junctions and displayed synchronized cilia beating. This supports the possibility that ependymal cells are born from subpopulations of NSC with their own time table of differentiation. As a whole, the findings indicate that the CSF is a supportive medium to host NE and that NE grafted into the CSF have the potential to produce neurons, glia and ependyma.


Subject(s)
Astrocytes/cytology , Cerebrospinal Fluid/physiology , Ependyma/cytology , Ependymoglial Cells/cytology , Hydrocephalus/pathology , Neural Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cilia/metabolism , Disease Models, Animal , Humans , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Neural Stem Cells/cytology , Neurons/cytology , Rats
3.
Stem Cell Res Ther ; 8(1): 27, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28173846

ABSTRACT

BACKGROUND: Stem cells are capable of unlimited self-renewal and are able to remain undifferentiated for extended periods of time prior to their differentiation into specific cell lineages. Because of the issues (ethical and religious) involved in the use of embryonic stem cells and the limited plasticity of adult stem cells, an alternative cell source could be foetal stem cells derived from extra-embryonic tissue, which are highly proliferative, grow in vitro and possess interesting immunogenic characteristics. As a result, the amniotic membrane of several species has been studied as an important new source of stem cells. METHODS: Here, we cultured and characterized mesenchymal progenitor cells derived from the rabbit amniotic membrane, and investigated their differentiation potential. In total, amniotic membranes were collected from eight rabbit foetuses and were isolated by the explant technique. The obtained cells were cultured in DMEM-HIGH glucose and incubated at 37 °C in a humidified atmosphere with 5% CO2. RESULTS: The cells adhered to the culture plates and showed a high proliferative capacity with fibroblast-like morphologies. The cells showed a positive response for markers for the cytoskeleton, mesenchymal stem cells and proliferation, pluripotency and haematopoietic precursor stem cells. However, the cells were negative for CD45, a marker of haematopoietic cells. Furthermore, the cells had the capacity to be induced to differentiate into osteogenic, adipogenic and chondrogenic lineages. In addition, when the cells were injected into nude mice, we did not observe the formation of tumours. CONCLUSIONS: In summary, our results demonstrate that multipotent mesenchymal stem cells can be obtained from the rabbit amniotic membrane for possible use in future cell therapy applications.


Subject(s)
Adipocytes/cytology , Amnion/cytology , Chondrocytes/cytology , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Osteoblasts/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Amnion/drug effects , Amnion/metabolism , Animals , Cell Differentiation , Cell Proliferation , Chondrocytes/drug effects , Chondrocytes/metabolism , Culture Media/pharmacology , Glucose/metabolism , Glucose/pharmacology , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Phenotype , Primary Cell Culture , Rabbits
4.
Stem Cell Res Ther ; 8(1): 20, 2017 01 28.
Article in English | MEDLINE | ID: mdl-28129776

ABSTRACT

BACKGROUND: The liver has the remarkable capacity to regenerate in order to compensate for lost or damaged hepatic tissue. However, pre-existing pathological abnormalities, such as hepatic steatosis (HS), inhibits the endogenous regenerative process, becoming an obstacle for liver surgery and living donor transplantation. Recent evidence indicates that multipotent mesenchymal stromal cells (MSCs) administration can improve hepatic function and increase the potential for liver regeneration in patients with liver damage. Since HS is the most common form of chronic hepatic illness, in this study we evaluated the role of MSCs in liver regeneration in an animal model of severe HS with impaired liver regeneration. METHODS: C57BL/6 mice were fed with a regular diet (normal mice) or with a high-fat diet (obese mice) to induce HS. After 30 weeks of diet exposure, 70% hepatectomy (Hpx) was performed and normal and obese mice were divided into two groups that received 5 × 105 MSCs or vehicle via the tail vein immediately after Hpx. RESULTS: We confirmed a significant inhibition of hepatic regeneration when liver steatosis was present, while the hepatic regenerative response was promoted by infusion of MSCs. Specifically, MSC administration improved the hepatocyte proliferative response, PCNA-labeling index, DNA synthesis, liver function, and also reduced the number of apoptotic hepatocytes. These effects may be associated to the paracrine secretion of trophic factors by MSCs and the hepatic upregulation of key cytokines and growth factors relevant for cell proliferation, which ultimately improves the survival rate of the mice. CONCLUSIONS: MSCs represent a promising therapeutic strategy to improve liver regeneration in patients with HS as well as for increasing the number of donor organs available for transplantation.


Subject(s)
Fatty Liver/therapy , Liver Regeneration/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Obesity/therapy , Animals , Apoptosis , Biomarkers/metabolism , Cell Proliferation , Cytokines/genetics , Cytokines/metabolism , DNA/biosynthesis , Diet, High-Fat , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/pathology , Gene Expression , Hepatectomy , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Liver/pathology , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Multipotent Stem Cells/metabolism , Obesity/etiology , Obesity/genetics , Obesity/pathology , Paracrine Communication , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Transplantation, Homologous
5.
Cell Tissue Res ; 367(2): 243-256, 2017 02.
Article in English | MEDLINE | ID: mdl-27677269

ABSTRACT

The possibility of isolating bovine mesenchymal multipotent stromal cells (MSCs) from fetal adnexa is an interesting prospect due to the potential use of these cells in biotechnological applications. However, little is known about the properties of these progenitor cells in bovine species. Wharton's jelly (WJ) MSC cells were obtained from the umbilical cord of bovine fetuses at three different stages of pregnancy and divided into groups 1, 2 and 3 according to gestational trimester. Cell morphology, from the three stages of pregnancy, typically appeared fibroblast-like spindle-shaped, presenting the same viability and number. Moreover, the proliferative ability of T-cells in response to a mitogenic stimulus was suppressed when WJMSC cells were added to the culture. Multilineage properties were confirmed by their ability to undergo adipogenic, osteogenic/chondrogenic and neurogenic differentiation. Mesenchymal phenotyping, CD105+, CD29+, CD73+ and CD90+ cell markers were detected in all three cell groups, yet these markers were considered more expressed in MSCs of group 2 (p < 0.005). Expression of cytokines IL2, IL6RR, INFAC, INFB1, IFNG, TNF and LTBR were downregulated, whereas IL1F10 expression was upregulated in all tested WJMSCs. The present study demonstrated that WJMSCs harvested from the bovine umbilical cord at different gestational stages showed proliferative capacity, immune privilege and stemness potential.


Subject(s)
Cell Separation/methods , Immunomodulation/genetics , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Pregnancy Trimesters/genetics , Transcription, Genetic , Wharton Jelly/cytology , Animals , Biomarkers/metabolism , Cattle , Cell Differentiation , Cell Lineage , Cell Proliferation , Cell Shape , Cell Survival , Female , Flow Cytometry , Gene Expression Profiling , Mesenchymal Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Phenotype , Pregnancy , Telomerase/metabolism , Umbilical Cord/cytology
6.
Mol Neurobiol ; 53(10): 7184-7199, 2016 12.
Article in English | MEDLINE | ID: mdl-26687184

ABSTRACT

Mesenchymal stem cells (MSCs) have recently been described to home to brain tumors and to integrate into the tumor-associated stroma. Understanding the communication between cancer cells and MSCs has become fundamental to determine whether MSC-tumor interactions should be exploited as a vehicle for therapeutic agents or considered a target for intervention. Therefore, we investigated whether conditioned medium from adipose-derived stem cells (ADSCs-CM) modulate glioma tumor cells by analyzing several cell biology processes in vitro. C6 rat glioma cells were treated with ADSCs-CM, and cell proliferation, cell cycle, cell viability, cell morphology, adhesion, migration, and expression of epithelial-mesenchymal transition (EMT)-related surface markers were analyzed. ADSCs-CM did not alter cell viability, cell cycle, and growth rate of C6 glioma cells but increased their migratory capacity. Moreover, C6 cells treated with ADSC-CM showed reduced adhesion and underwent changes in cell morphology. Up-regulation of EMT-associated markers (vimentin, MMP2, and NRAS) was also observed following treatment with ADSC-CM. Our findings demonstrate that the paracrine factors released by ADSCs are able to modulate glioma cell biology. Therefore, ADSC-tumor cell interactions in a tumor microenvironment must be considered in the design of clinical application of stem cell therapy. Graphical Abstract Factors released by adipose-derived stem cells (ADSCs) may modulate the biology of C6 glioma cells. When C6 cells are exposed to a conditioned medium from adipose-derived stem cells (ADSCs-CM), some of these cells can undergo an EMT-like process and trans-differentiate into cells with a more mesenchymal phenotype, characterized by enhanced expression of EMT-related surface markers, reduced cell adhesion capacity, increased migratory capacity, as well as changes in cell and nuclei morphology.


Subject(s)
Adipose Tissue/cytology , Brain Neoplasms/pathology , Culture Media, Conditioned/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Glioma/pathology , Stem Cells/cytology , Actins/metabolism , Animals , Biomarkers/metabolism , Biomarkers, Tumor/metabolism , Cell Adhesion/drug effects , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Disease Progression , L-Lactate Dehydrogenase/metabolism , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Rats
8.
Exp Cell Res ; 327(1): 37-47, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-24907656

ABSTRACT

Epidermal neural crest stem cells (EPI-NCSCs), which reside in the bulge of hair follicles, are attractive candidates for several applications in cell therapy, drug screening and tissue engineering. As suggested remnants of the embryonic neural crest (NC) in an adult location, EPI-NCSCs are able to generate a wide variety of cell types and are readily accessible by a minimally invasive procedure. Since the combination of epidermal growth factor (EGF) and fibroblast growth factor type 2 (FGF2) is mitogenic and promotes the neuronal commitment of various stem cell populations, we examined its effects in the proliferation and neuronal potential of mouse EPI-NCSCs. By using a recognized culture protocol of bulge whiskers follicles, we were able to isolate a population of EPI-NCSCs, characterized by the migratory potential, cell morphology and expression of phenotypic markers of NC cells. EPI-NCSCs expressed neuronal, glial and smooth muscle markers and exhibited the NC-like fibroblastic morphology. The treatment with the combination EGF and FGF2, however, increased their proliferation rate and promoted the acquisition of a neuronal-like morphology accompanied by reorganization of neural cytoskeletal proteins ßIII-tubulin and nestin, as well as upregulation of the pan neuronal marker ßIII-tubulin and down regulation of the undifferentiated NC, glial and smooth muscle cell markers. Moreover, the treatment enhanced the response of EPI-NCSCs to neurogenic stimulation, as evidenced by induction of GAP43, and increased expression of Mash-1 in neuron-like cell, both neuronal-specific proteins. Together, the results suggest that the combination of EGF-FGF2 stimulates the proliferation and improves the neuronal potential of EPI-NCSCs similarly to embryonic NC cells, ES cells and neural progenitor/stem cells of the central nervous system and highlights the advantage of using EGF-FGF2 in neuronal differentiation protocols.


Subject(s)
Epidermal Growth Factor/metabolism , Epidermis/metabolism , Fibroblast Growth Factor 2/metabolism , Neural Crest/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Differentiation/physiology , Cell Proliferation , Down-Regulation/physiology , Epithelial Cells/metabolism , GAP-43 Protein/metabolism , Hair Follicle/metabolism , Mice , Multipotent Stem Cells/metabolism , Myocytes, Smooth Muscle/metabolism , Up-Regulation/physiology
9.
PLoS One ; 9(2): e89542, 2014.
Article in English | MEDLINE | ID: mdl-24586857

ABSTRACT

New strategies for skin regeneration are needed in order to provide effective treatment for cutaneous wounds and disease. Mesenchymal stem cells (MSCs) are an attractive source of cells for tissue engineering because of their prolonged self-renewal capacity, multipotentiality, and ability to release active molecules important for tissue repair. In this paper, we show that human skin-derived mesenchymal stromal cells (SD-MSCs) display similar characteristics to the multipotent MSCs. We also evaluate their growth in a three-dimensional (3D) culture system with dermal substitutes (Integra and Pelnac). When cultured in monolayers, SD-MSCs expressed mesenchymal markers, such as CD105, Fibronectin, and α-SMA; and neural markers, such as Nestin and ßIII-Tubulin; at transcriptional and/or protein level. Integra and Pelnac equally supported the adhesion, spread and growth of human SD-MSCs in 3D culture, maintaining the MSC characteristics and the expression of multilineage markers. Therefore, dermal substitutes support the growth of mesenchymal stromal cells from human skin, promising an effective tool for tissue engineering and regenerative technology.


Subject(s)
Cell Differentiation , Dermis/cytology , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Regeneration/physiology , Skin, Artificial , Skin/cytology , Biomarkers , Blotting, Western , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Dermis/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mesenchymal Stem Cells/metabolism , Multipotent Stem Cells/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin/metabolism , Tissue Engineering
10.
Dev Neurobiol ; 72(12): 1482-97, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22234946

ABSTRACT

In this study, we have analyzed the specific contribution of the cortical radial glia (RG) for gap junctional communication (GJC) within the postnatal subventricular zone (SVZ). To specifically target RG as source of dye-coupling in situ, we have developed a new technique that involves direct cell loading through the processes that reach the pial surface, with a mix of gap junction permeant (Lucifer yellow, LY) and nonpermeant (rhodamine-conjugated dextran 3 KDa, RD) fluorochromes, the latter used as a marker for direct loaded cells. Tissue sections were analyzed for identification of directly loaded (LY+RD+) and coupled cells (LY+RD-) in the SVZ. Directly loaded cells were restricted to the region underlying the pial loading surface area. Coupled cells were distributed in a bistratified manner, along the outer dorsal surface of the SVZ and aligning the ventricle, leaving the SVZ core relatively free. Blocking GJC prior to pial loading greatly reduced dye coupling. Phenotypic analysis indicated that coupling by RG excludes neuroblasts and is mostly restricted to cells of glial lineage. Notwithstanding, no corresponding restriction to specific cell phenotype was found for two connexin isotypes, Cx43 and Cx45, in the postnatal SVZ. The extensive homocellular cell coupling by RG suggests an important role in the regulation of neurogenesis and functional compartmentalization of the postnatal SVZ.


Subject(s)
Cerebral Cortex/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , Neuroglia/cytology , Animals , Cell Communication/physiology , Cerebral Cortex/metabolism , Connexins/analysis , Connexins/metabolism , Gap Junctions/metabolism , Immunohistochemistry , Isoquinolines , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Neuroglia/metabolism , Rats , Rats, Wistar
11.
Stem Cells Dev ; 19(3): 395-402, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19290805

ABSTRACT

The canine model provides a large animal system to evaluate many treatment modalities using stem cells (SCs). However, only bone marrow (BM) protocols have been widely used in dogs for preclinical approaches. BM donation consists of an invasive procedure and the number and differentiation potential of its mesenchymal stem cells (MSCs) decline with age. More recently, umbilical cord was introduced as an alternative source to BM since it is obtained from a sample that is routinely discarded. Here, we describe the isolation of MSCs from canine umbilical cord vein (cUCV). These cells can be obtained from every cord received and grow successfully in culture. Their multipotent plasticity was demonstrated by their capacity to differentiate in adipocytic, chondrocytic, and osteocytic lineages. Furthermore, our results open possibilities to use cUCV cells in preclinical trials for many well-characterized canine model conditions homologs to human diseases.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Umbilical Veins/cytology , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Cell Separation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Dogs , Flow Cytometry , Gene Expression Profiling , Immunohistochemistry , Immunophenotyping , Mesenchymal Stem Cells/metabolism , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Osteocytes/cytology , Osteocytes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
12.
Biocell ; Biocell;33(1): 33-38, Apr. 2009. ilus
Article in English | BINACIS | ID: bin-127216

ABSTRACT

The kidney has an inherent ability for recovery and regeneration following acute damage. However, there has been much contention as to the source of regenerating renal cells. The aim of this study was to isolate and characterize these cells. Normal rat kidneys were minced and cells were isolated with collagenase I and were cultured in an expansion medium. Adherent cells were isolated and expanded for more than 120 days in vitro. These cells had the potential of trans-lineage differentiation into neural cells, adipocytes and osteocytes. These cells also expressed Nucleostemin, Cyclin D1, Notch1 and Survivin which are commonly expressed in stem cells. The results of the current work show that the adult kidney contains a population of multipotent stem cells.(AU)


Subject(s)
Animals , Female , Rats , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Carrier Proteins/metabolism , Cyclin D1/metabolism , Kidney/cytology , Kidney/physiology , Receptor, Notch1/metabolism , /metabolism , Rats, Wistar , Cell Differentiation/physiology , Cell Separation/methods , Nuclear Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Regeneration
13.
Biocell ; Biocell;33(1): 33-38, Apr. 2009. ilus
Article in English | LILACS | ID: lil-595027

ABSTRACT

The kidney has an inherent ability for recovery and regeneration following acute damage. However, there has been much contention as to the source of regenerating renal cells. The aim of this study was to isolate and characterize these cells. Normal rat kidneys were minced and cells were isolated with collagenase I and were cultured in an expansion medium. Adherent cells were isolated and expanded for more than 120 days in vitro. These cells had the potential of trans-lineage differentiation into neural cells, adipocytes and osteocytes. These cells also expressed Nucleostemin, Cyclin D1, Notch1 and Survivin which are commonly expressed in stem cells. The results of the current work show that the adult kidney contains a population of multipotent stem cells.


Subject(s)
Animals , Female , Rats , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Cyclin D1/metabolism , /metabolism , Carrier Proteins/metabolism , Receptor, Notch1/metabolism , Kidney/cytology , Kidney/physiology , Cell Differentiation/physiology , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Rats, Wistar , Regeneration , Cell Separation/methods
14.
Biochem Biophys Res Commun ; 378(3): 456-61, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19032948

ABSTRACT

Mesenchymal stem cells (MSCs) have received special attention for cardiomyoplasty because several studies have shown that they differentiate into cardiomyocytes both in vitro and in vivo. Nitric oxide (NO) is a free radical signaling molecule that regulates several differentiation processes including cardiomyogenesis. Here, we report an investigation of the effects of two NO agents (SNAP and DEA/NO), able to activate both cGMP-dependent and -independent pathways, on the cardiomyogenic potential of bone marrow-derived mesenchymal stem cells (BM-MSCs) and adipose tissue-derived stem cells (ADSCs). The cells were isolated, cultured and treated with NO agents. Cardiac- and muscle-specific gene expression was analyzed by indirect immunofluorescence, flow cytometry, RT-PCR and real-time PCR. We found that untreated (control) ADSCs and BM-MSCs expressed some muscle markers and NO-derived intermediates induce an increased expression of some cardiac function genes in BM-MSCs and ADSCs. Moreover, NO agents considerably increased the pro-angiogenic potential mostly of BM-MSCs as determined by VEGF mRNA levels.


Subject(s)
Adult Stem Cells/drug effects , Cell Differentiation/drug effects , Hydrazines/pharmacology , Mesenchymal Stem Cells/drug effects , Myocytes, Cardiac/cytology , Nitric Oxide Donors/pharmacology , Penicillamine/analogs & derivatives , Adult , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Aged , Antigens, CD/genetics , Cardiomyoplasty , Cell Differentiation/genetics , Cells, Cultured , Connexin 43/genetics , Gene Expression , Genetic Markers , Heart/physiology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Middle Aged , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Muscle Proteins/genetics , Nitric Oxide/metabolism , Penicillamine/pharmacology , Vascular Endothelial Growth Factor A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL