Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Cell Mol Med ; 25(21): 10185-10196, 2021 11.
Article in English | MEDLINE | ID: mdl-34622573

ABSTRACT

Diabetes causes vascular injury and carries a high risk of ischaemic stroke. Human amniotic fluid stem cells (hAFSCs) can enhance cerebral vascular remodelling and have the potential to improve neurological function after stroke in diabetic rats. Five groups of female rats were examined: (1) normal control, (2) type 1 diabetic (T1DM) rats induced by streptozotocin injection (DM), (3) non-DM rats receiving 60-minute middle cerebral artery occlusion (MCAO), (4) T1DM rats receiving 60-minute MCAO (DM + MCAO) and (5) T1DM rats receiving 60-minute MCAO and injection with 5 × 106  hAFSCs at 3 h after MCAO (DM + MCAO + hAFSCs). Neurological function was examined before, and at 1, 7, 14, 21 and 28 days, and cerebral infarction volume and haemorrhage, cerebral vascular density, angiogenesis and inflammatory were examined at 7 and 28 days after MCAO. hAFSCs treatment caused a significant improvement of neurological dysfunction, infarction volume, blood-brain barrier leakage, cerebral arterial density, vascular density and angiogenesis and a reduction of brain haemorrhage and inflammation compared with non-treatment. Our results showed that the effect of hAFSCs treatment against focal cerebral ischaemia may act through the recovery of vascular remodelling and angiogenesis and the reduction of inflammation in ischaemic brain.


Subject(s)
Amniotic Fluid/cytology , Brain Ischemia/metabolism , Brain Ischemia/therapy , Stem Cell Transplantation , Stem Cells/metabolism , Vascular Remodeling , Animals , Biomarkers , Blood Glucose , Blood-Brain Barrier/metabolism , Brain Ischemia/diagnosis , Brain Ischemia/etiology , Diabetes Mellitus, Experimental , Disease Models, Animal , Disease Susceptibility , Female , Gene Expression Regulation , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Matrix Metalloproteinase 9/metabolism , Rats , Stem Cells/cytology
2.
J Cell Mol Med ; 25(16): 8074-8086, 2021 08.
Article in English | MEDLINE | ID: mdl-34288391

ABSTRACT

Second trimester foetal human amniotic fluid-derived stem cells (hAFS) have been shown to possess remarkable cardioprotective paracrine potential in different preclinical models of myocardial injury and drug-induced cardiotoxicity. The hAFS secretome, namely the total soluble factors released by cells in their conditioned medium (hAFS-CM), can also strongly sustain in vivo angiogenesis in a murine model of acute myocardial infarction (MI) and stimulates human endothelial colony-forming cells (ECFCs), the only truly recognized endothelial progenitor, to form capillary-like structures in vitro. Preliminary work demonstrated that the hypoxic hAFS secretome (hAFS-CMHypo ) triggers intracellular Ca2+ oscillations in human ECFCs, but the underlying mechanisms and the downstream Ca2+ -dependent effectors remain elusive. Herein, we found that the secretome obtained by hAFS undergoing hypoxic preconditioning induced intracellular Ca2+ oscillations by promoting extracellular Ca2+ entry through Transient Receptor Potential Vanilloid 4 (TRPV4). TRPV4-mediated Ca2+ entry, in turn, promoted the concerted interplay between inositol-1,4,5-trisphosphate- and nicotinic acid adenine dinucleotide phosphate-induced endogenous Ca2+ release and store-operated Ca2+ entry (SOCE). hAFS-CMHypo -induced intracellular Ca2+ oscillations resulted in the nuclear translocation of the Ca2+ -sensitive transcription factor p65 NF-κB. Finally, inhibition of either intracellular Ca2+ oscillations or NF-κB activity prevented hAFS-CMHypo -induced ECFC tube formation. These data shed novel light on the molecular mechanisms whereby hAFS-CMHypo induces angiogenesis, thus providing useful insights for future therapeutic strategies against ischaemic-related myocardial injury.


Subject(s)
Amniotic Fluid/metabolism , Calcium/metabolism , Culture Media, Conditioned/chemistry , Endothelial Cells/physiology , NF-kappa B/metabolism , Secretome , Stem Cells/cytology , Amniotic Fluid/chemistry , Cells, Cultured , Endothelial Cells/cytology , Humans , NF-kappa B/genetics , Protein Transport , Signal Transduction , Stem Cells/metabolism
3.
Tissue Cell ; 68: 101472, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33360545

ABSTRACT

The use of mesenchymal stem cell sheets is a promising strategy for skin regeneration. The injection of dissociated human amniotic fluid stem cells (hAFSCs) was recently found to accelerate cutaneous wound healing with reduced fibrotic scarring, similar to fetal wound healing. However, the use of hAFSCs in applications of cell sheet technology remains limited. The aim of this study was to determine the in vivo efficacy of in vitro-cultured hAFSC sheets in wound healing. The cell sheets were characterized by immunohistochemistry and RT-qPCR and grafted onto full-thickness wounds in BALB/c mice. The wound size was measured, and re-epithelialization, granulation tissue area, and collagen content of the regenerated wound were analyzed histologically. Although the hAFSC sheet contained abundant extracellular matrix molecules and expressed high levels of anti-fibrotic mediators, its grafting did not affect wound closure or the size of the granulation tissue area. In contrast, the organization of type I collagen bundles in the regenerated wound was markedly reduced, while the levels of type III collagen were increased after implantation of the hAFSC sheet. These results suggest that hAFSC sheets can exert anti-fibrotic properties without delaying wound closure.


Subject(s)
Amniotic Fluid/cytology , Skin/pathology , Stem Cells/cytology , Tissue Engineering , Wound Healing , Animals , Cell Differentiation , Cell Membrane/metabolism , Cells, Cultured , Collagen/metabolism , Epidermis/pathology , Female , Fibrosis , Granulation Tissue/pathology , Humans , Immunophenotyping , Male , Mice, Inbred BALB C
4.
J Pediatr Surg ; 56(1): 11-16, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33129508

ABSTRACT

PURPOSE: Biliary atresia (BA) is a fibro-obliterative cholangiopathy that involves both extrahepatic and intrahepatic bile ducts in infants. Cholangiocyte apoptosis has an influence on the fibrogenesis process of bile ducts and the progression of liver fibrosis in BA. Human amniotic fluid stem cells (hAFSCs) are multipotent cells that have ability to inhibit cell apoptosis. We aimed to investigate whether hAFSCs have the potential to attenuate cholangiocyte apoptosis and injury induced fibrogenic response in our ex vivo bile duct injury model of liver ductal organoids. METHODS: The anti-apoptotic effect of hAFSCs was tested in the acetaminophen-induced injury model of neonatal mouse liver ductal organoids (AUP #42681) by using direct and indirect co-culture systems. Cell apoptosis and proliferation were evaluated by immunofluorescent staining. Expression of fibrogenic cytokines was analyzed by RT-qPCR. Data were compared using one-way ANOVA with post hoc test. RESULTS: In our injury model, liver ductal organoids that were treated with hAFSCs in both direct and indirect co-culture systems had a significantly smaller number of apoptotic cholangiocytes and decreased expression of fibrogenic cytokines, transforming growth factor beta-1 (TGF-ß1) and platelet-derived growth factor-BB (PDGF-BB). Moreover, hAFSCs increased cholangiocyte proliferation in injured organoids. CONCLUSION: hAFSCs have the ability to protect the organoids from injury by decreasing cholangiocyte apoptosis and promoting cholangiocyte proliferation. This protective ability of hAFSCs leads to inhibition of the fibrogenic response in the injured organoids. hAFSCs have high therapeutic potential to attenuate liver fibrogenesis in cholangiopathic diseases such as BA.


Subject(s)
Amniotic Fluid , Organoids , Apoptosis , Bile Ducts/surgery , Humans , Liver , Stem Cells
5.
Cell Transplant ; 29: 963689720909387, 2020.
Article in English | MEDLINE | ID: mdl-32452747

ABSTRACT

The effects of human amniotic fluid stem cells (hAFSCs) transplantation on bladder dysfunction after pelvic nerve transection (PNT) remain to be clarified. Five groups of female Sprague-Dawley rats were studied including sham operation, unilateral PNT alone or plus hAFSCs transplantation, and bilateral PNT alone or plus hAFSCs transplantation. hAFSCs were injected at the site of PNT. Cystometries, neurofilament density within bladder nerves, and the expressions of bladder protein gene-product 9.5 (PGP9.5), growth-associated protein 43 (GAP-43), nerve growth factor (NGF), p75 (NGF receptor), CXCL12, CCL7, and enkephalin were studied. Compared to sham-operation group, bladder weight increased and neurofilament density decreased at 10 and 28 days after unilateral and bilateral PNT, but all improved after hAFSCs transplantation. Unilateral PNT could increase bladder capacity, residual volume, and number of nonvoiding contractions but decrease peak voiding pressure and leak point pressure. Bilateral PNT caused overflow incontinence and increased the number of nonvoiding contractions. These cystometric parameters improved after hAFSCs transplantation. After PNT, bladder PGP9.5 mRNA and immunoreactivities decreased at 10 and 28 days, GAP-43 mRNA and immunoreactivities increased at 10 days and decreased at 28 days, both NGF and p75 mRNAs and immunoreactivities increased at 10 and/or 28 days, and enkephalin immunoreactivities decreased at 10 and 28 days, but these were all improved after hAFSCs transplantation. Our results showed that bladder dysfunction induced by PNT could be improved by hAFSCs transplantation, and PGP9.5, GAP-43, and neurotrophins could be involved in the mechanisms of nerve regeneration after hAFSCs transplantation.


Subject(s)
Amniotic Fluid/metabolism , Stem Cells/cytology , Urinary Bladder/innervation , Urinary Bladder/metabolism , Animals , Disease Models, Animal , Female , Nerve Regeneration/physiology , Rats, Sprague-Dawley , Stem Cell Transplantation/methods , Urologic Diseases/therapy
6.
Int J Mol Sci ; 21(7)2020 Mar 29.
Article in English | MEDLINE | ID: mdl-32235313

ABSTRACT

Mature mammalian hearts possess very limited regenerative potential. The irreversible cardiomyocyte loss after heart injury can lead to heart failure and death. Pluripotent stem cells (PSCs) can differentiate into cardiomyocytes for cardiac repair, but there are obstacles to their clinical application. Among these obstacles is their potential for post-transplant rejection. Although human amniotic fluid-derived stem cells (hAFSCs) are immune privileged, they cannot induce cardiac differentiation. Thus, we generated hAFSC-derived induced PSCs (hAFSC-iPSCs) and used a Wnt-modulating differentiation protocol for the cardiac differentiation of hAFSC-iPSCs. In vitro studies using flow cytometry, immunofluorescence staining, and patch-clamp electrophysiological study, were performed to identify the characteristics of hAFSC-iPSC-derived cardiomyocytes (hAFSC-iPSC-CMs). We injected hAFSC-iPSC-CMs intramuscularly into rat infarcted hearts to evaluate the therapeutic potential of hAFSC-iPSC-CM transplantation. At day 21 of differentiation, the hAFSC-iPSC-CMs expressed cardiac-specific marker (cardiac troponin T), presented cardiomyocyte-specific electrophysiological properties, and contracted spontaneously. Importantly, these hAFSC-iPSC-CMs demonstrated low major histocompatibility complex (MHC) class I antigen expression and the absence of MHC class II antigens, indicating their low immunogenicity. The intramyocardial transplantation of hAFSC-iPSC-CMs restored cardiac function, partially remuscularized the injured region, and reduced fibrosis in the rat infarcted hearts. Therefore, hAFSC-iPSCs are potential candidates for the repair of infarcted myocardium.


Subject(s)
Amniotic Fluid/cytology , Cell Differentiation , Embryonic Stem Cells/cytology , Immune Privilege , Induced Pluripotent Stem Cells/cytology , Muscle Development , Myocytes, Cardiac/cytology , Animals , Biomarkers , Disease Models, Animal , Electrophysiological Phenomena , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Myocardial Infarction/diagnosis , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Phenotype , Rats , Regeneration , Stem Cell Transplantation/methods , Treatment Outcome , Ventricular Function, Left
7.
FASEB J ; 34(5): 6824-6836, 2020 05.
Article in English | MEDLINE | ID: mdl-32223023

ABSTRACT

Ischemia/reperfusion (I/R) is implicated in the pathogenesis of various acute intestinal injuries. Amniotic fluid stem cells (AFSC) are beneficial in experimental intestinal diseases. Tumor necrosis factor-induced protein 6 (TSG-6) has been shown to exert anti-inflammatory effects. We aimed to investigate if AFSC secreted TSG-6 reduces inflammation and rescues intestinal I/R injury. The superior mesenteric artery of 3-week-old rats was occluded for 90 minutes and green fluorescent protein-labeled AFSC or recombinant TSG-6 was injected intravenously upon reperfusion. AFSC distribution was evaluated at 24, 48, and 72 hours after I/R. AFSC and TSG-6 effects on the intestine were assessed 48 hours postsurgery. Intestinal organoids were used to study the effects of TSG-6 after hypoxia-induced epithelial damage. After I/R-induced intestinal injury, AFSC migrated preferentially to the ileum, the primary site of injury, through blood circulation. Engrafted AFSC reduced ileum injury, inflammation, and oxidative stress. These AFSC-mediated beneficial effects were dependent on secretion of TSG-6. Administration of TSG-6 protected against hypoxia-induced epithelial damage in intestinal organoids. Finally, TSG-6 attenuated intestinal damage during I/R by suppressing genes involved in wound and injury pathways. This study indicates that AFSC or TSG-6 have the potential of rescuing the intestine from the damage caused by I/R.


Subject(s)
Amniotic Fluid/cytology , Cell Adhesion Molecules/metabolism , Inflammation/therapy , Intestinal Diseases/therapy , Reperfusion Injury/complications , Stem Cell Transplantation/methods , Amniotic Fluid/metabolism , Animals , Cell Adhesion Molecules/genetics , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Intestinal Diseases/etiology , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Oxidative Stress , Rats , Rats, Sprague-Dawley
8.
Tissue Eng Regen Med ; 16(1): 81-92, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30815353

ABSTRACT

BACKGROUND: Despite major progress in stem cell therapy, our knowledge of the characteristics and tissue regeneration potency of long-term transported cells is insufficient. In a previous in vitro study, we established the optimal cell transport conditions for amniotic fluid stem cells (AFSCs). In the present study, the target tissue regeneration of long-term transported cells was validated in vivo. METHODS: For renal regeneration, transported AFSCs were seeded on a poly(lactide-co-glycolide) scaffold and implanted in a partially resected kidney. The target tissue regeneration of the transported cells was compared with that of freshly harvested cells in terms of morphological reconstruction, histological microstructure reformation, immune cell infiltration, presence of induced cells, migration into remote organs, expression of inflammation/fibrosis/renal differentiation-related factors, and functional recovery. RESULTS: The kidney implanted with transported cells showed recovery of total kidney volume, regeneration of glomerular/renal tubules, low CD4/CD8 infiltration, and no occurrence of cancer during 40 weeks of observation. The AFSCs gradually disappeared and did not migrate into the liver, lung, or spleen. We observed low expression levels of pro-inflammatory cytokines and fibrotic factors; enhanced expression of the genes Wnt4, Pax2, Wt1, and Emx2; and significantly reduced blood urea nitrogen and creatinine values. There were no statistical differences between the performance of freshly harvested cells and that of the transported cells. CONCLUSION: This study demonstrates that long-term transported cells under optimized conditions can be used for cell therapy without adverse effects on stem cell characteristics, in vivo safety, and tissue regeneration potency.

9.
Hum Cell ; 32(1): 51-63, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30506493

ABSTRACT

Adult wound healing can result in fibrotic scarring (FS) characterized by excess expression of myofibroblasts and increased type I/type III collagen expression. In contrast, fetal wound healing results in complete regeneration without FS, and the mechanism remains unclear. Amniotic fluid cells could contribute to scar-free wound healing, but the effects of human amniotic fluid cells are not well characterized. Here, we determined the effect of human amniotic fluid stem cells (hAFS) on FS during wound healing. Human amniotic fluid was obtained by amniocentesis at 15-17 weeks of gestation. CD117-positive cells were isolated and defined as hAFS. hAFS (1 × 106) suspended in PBS or cell-free PBS were injected around wounds created in the dorsal region of BALB/c mice. Wound size was macroscopically measured, and re-epithelialization in the epidermis, granulation tissue area in the dermis and collagen contents in the regenerated wound were histologically analyzed. The ability of hAFS to engraft in the wound was assessed by tracking hAFS labeled with PKH-26. hAFS fulfilled the minimal criteria for mesenchymal stem cells. hAFS injection into the wound accelerated wound closure via enhancement of re-epithelialization with less FS. The process was characterized by lower numbers of myofibroblasts and higher expression of type III collagen. Finally, transplanted hAFS were clearly observed in the dermis until day 7 implying that hAFS worked in a paracrine manner. hAFS can function in a paracrine manner to accelerate cutaneous wound healing, producing less FS, a process resembling fetal wound healing.


Subject(s)
Amniotic Fluid/cytology , Skin Physiological Phenomena , Stem Cells/physiology , Wound Healing/physiology , Wounds and Injuries/metabolism , Animals , Cells, Cultured , Cicatrix/metabolism , Cicatrix/pathology , Cicatrix/prevention & control , Collagen/metabolism , Escherichia coli Proteins , Humans , Membrane Transport Proteins , Mice , Mice, Inbred BALB C , Myofibroblasts/pathology , Wounds and Injuries/pathology , Wounds and Injuries/physiopathology
10.
Article in English | WPRIM (Western Pacific) | ID: wpr-742382

ABSTRACT

BACKGROUND: Despite major progress in stem cell therapy, our knowledge of the characteristics and tissue regeneration potency of long-term transported cells is insufficient. In a previous in vitro study, we established the optimal cell transport conditions for amniotic fluid stem cells (AFSCs). In the present study, the target tissue regeneration of long-term transported cells was validated in vivo. METHODS: For renal regeneration, transported AFSCs were seeded on a poly(lactide-co-glycolide) scaffold and implanted in a partially resected kidney. The target tissue regeneration of the transported cells was compared with that of freshly harvested cells in terms of morphological reconstruction, histological microstructure reformation, immune cell infiltration, presence of induced cells, migration into remote organs, expression of inflammation/fibrosis/renal differentiation-related factors, and functional recovery. RESULTS: The kidney implanted with transported cells showed recovery of total kidney volume, regeneration of glomerular/renal tubules, low CD4/CD8 infiltration, and no occurrence of cancer during 40 weeks of observation. The AFSCs gradually disappeared and did not migrate into the liver, lung, or spleen. We observed low expression levels of proinflammatory cytokines and fibrotic factors; enhanced expression of the genes Wnt4, Pax2, Wt1, and Emx2; and significantly reduced blood urea nitrogen and creatinine values. There were no statistical differences between the performance of freshly harvested cells and that of the transported cells. CONCLUSION: This study demonstrates that long-term transported cells under optimized conditions can be used for cell therapy without adverse effects on stem cell characteristics, in vivo safety, and tissue regeneration potency.


Subject(s)
Female , Amniotic Fluid , Blood Urea Nitrogen , Cell- and Tissue-Based Therapy , Creatinine , Cytokines , In Vitro Techniques , Kidney , Liver , Lung , Polyglactin 910 , Regeneration , Spleen , Stem Cells
11.
Tissue Eng Regen Med ; 15(5): 639-647, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30603585

ABSTRACT

BACKGROUND: The preservation of stem cell viability and characteristics during cell transport from the bench to patients can significantly affect the success of cell therapy. Factors such as suspending medium, time, temperature, cell density, and container type could be considered for transport conditions. METHODS: To establish optimal conditions, human amniotic fluid stem cells' (AFSCs) viabilities were analyzed under different media {DMEM(H), DMEM/F-12, K-SFM, RPMI 1640, α-MEM, DMEM(L), PBS or saline}, temperature (4, 22 or 37 °C), cell density (1 × 107 cells were suspended in 0.1, 0.5, 1.0 or 2.0 mL of medium) and container type (plastic syringe or glass bottle). After establishing the transport conditions, stem cell characteristics of AFSCs were compared to freshly prepared cells. RESULTS: Cells transported in DMEM(H) showed relatively higher viability than other media. The optimized transport temperature was 4 °C, and available transport time was within 12 h. A lower cell density was associated with a better survival rate, and a syringe was selected as a transport container because of its clinical convenience. In compare of stem cell characteristics, the transported cells with established conditions showed similar potency as the freshly prepared cells. CONCLUSION: Our findings can provide a foundation to optimization of conditions for stem cell transport.

12.
Neurourol Urodyn ; 37(1): 123-131, 2018 01.
Article in English | MEDLINE | ID: mdl-28605059

ABSTRACT

AIMS: This study investigated the protective effect of human amniotic fluid-derived stem cells (hAFSCs) against bladder overactivity in rat model of atherosclerosis-induced chronic bladder ischemia. METHODS: Adult female Sprague-Dawley rats were divided into six groups: (1) Normal control with a regular diet for 8 weeks, (2) Sham-operation, (3) arterial balloon endothelial injury (AEI) of common iliac artery (AEI only), and post-AEI consecutive hAFSCs treatment for (4) 1 day, (5) 3 days, and (6) 7 days. Groups 2-6 were given 2% cholesterol diet for 8 weeks after operation (sham or AEI). Bladder functions were analyzed by cystometry at 8 weeks in controls and after operation in groups 2-6. Wall morphology of common iliac artery was examined by hematoxylin and eosin stain. Bladder oxidative stress and inflammatory markers were studied by immunohistochemistry of 8-hydroxy-2'-deoxyguanosine (8OHdG), malondialdehyde (MDA), and tumor necrosis factor-alpha (TNF-alpha). RESULTS: Bladder overactivity with decreased voided volumes and intercontraction intervals and increased residual volumes was seen in AEI only group, but improved after hAFSCs treatment for 1, 3, and 7 days. Compared with controls and shams, the wall thickness of iliac artery was increased in AEI only group, but improved after hAFSCs treatment for 3 and 7 days. The expressions of 8OHdG, MDA, and TNF-alpha were increased in AEI only group, but improved after hAFSCs treatment for 1, 3, and 7 days. CONCLUSIONS: Bladder overactivity caused by chronic bladder ischemia can be improved by hAFSCs treatment, probably by acting through down-regulation of oxidative stress and TNF-alpha expressions.


Subject(s)
Amniotic Fluid/cytology , Ischemia/therapy , Stem Cells/cytology , Urinary Bladder, Overactive/therapy , Urinary Bladder/physiopathology , 8-Hydroxy-2'-Deoxyguanosine , Animals , Deoxyguanosine/analogs & derivatives , Disease Models, Animal , Female , Ischemia/physiopathology , Malondialdehyde/metabolism , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Treatment Outcome , Urinary Bladder/blood supply , Urinary Bladder, Overactive/physiopathology
13.
Article in English | WPRIM (Western Pacific) | ID: wpr-717539

ABSTRACT

BACKGROUND: The preservation of stem cell viability and characteristics during cell transport from the bench to patients can significantly affect the success of cell therapy. Factors such as suspending medium, time, temperature, cell density, and container type could be considered for transport conditions. METHODS: To establish optimal conditions, human amniotic fluid stem cells' (AFSCs) viabilities were analyzed under different media {DMEM(H), DMEM/F-12, K-SFM, RPMI 1640, α-MEM, DMEM(L), PBS or saline}, temperature (4, 22 or 37 ℃), cell density (1 × 10⁷ cells were suspended in 0.1, 0.5, 1.0 or 2.0 mL of medium) and container type (plastic syringe or glass bottle). After establishing the transport conditions, stem cell characteristics of AFSCs were compared to freshly prepared cells. RESULTS: Cells transported in DMEM(H) showed relatively higher viability than other media. The optimized transport temperature was 4 ℃, and available transport time was within 12 h. A lower cell density was associated with a better survival rate, and a syringe was selected as a transport container because of its clinical convenience. In compare of stem cell characteristics, the transported cells with established conditions showed similar potency as the freshly prepared cells. CONCLUSION: Our findings can provide a foundation to optimization of conditions for stem cell transport.


Subject(s)
Female , Humans , Amniotic Fluid , Cell Count , Cell Survival , Cell- and Tissue-Based Therapy , Glass , Stem Cells , Survival Rate , Syringes
14.
Stem Cells Transl Med ; 6(4): 1227-1236, 2017 04.
Article in English | MEDLINE | ID: mdl-28186672

ABSTRACT

The objective is to investigate whether human amniotic fluid stem cells (hAFSCs) grafting into the bladder may influence bladder functional and molecular changes in an animal stroke model. Female rats were divided into three groups: sham, middle cerebral artery occlusion (MCAO) alone, and MCAO plus 1 × 106 hAFSCs transplanting into bladder wall. Bladder function was analyzed by cystometry at days 3 and 10 after MCAO. The expressions of bladder nerve growth factor (NGF), M2-muscarinic, M3-muscarinic, and P2X1 receptors were measured by immunohistochemistry and real-time polymerase chain reaction. When compared with sham-operated group, MCAO alone rats had significant increase in residual volume and decrease in voided volume and intercontraction interval; however, these bladder dysfunctions were improved following hAFSCs transplantation. The immunoreactivities of NGF, M3, and P2X1 significantly decreased at days 3 and 10, but M2 increased at day 3 after MCAO. Following hAFSCs transplantation, the immunoreactivities of NGF and P2X1 significantly increased at day 3, and M2 increased at day 10 after MCAO. The mRNAs of NGF, M2, and M3 significantly increased at day 3, but NGF and M2 decreased at day 10 after MCAO. Following hAFSCs transplantation, there was significant decrease in M2 mRNA at day 3 and increase in P2X1 mRNA at days 3 and 10 after MCAO. Bladder dysfunction caused by MCAO can be improved by hAFSCs transplanting into bladder which may be related to the expressions of bladder NGF, and muscarinic and P2X1 receptors. Stem Cells Translational Medicine 2017;6:1227-1236.


Subject(s)
Amniotic Fluid/cytology , Brain Ischemia/therapy , Stem Cell Transplantation/methods , Stem Cells/cytology , Urinary Bladder/cytology , Animals , Brain Ischemia/metabolism , Female , Immunohistochemistry , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
15.
Stem Cells ; 35(7): 1663-1673, 2017 07.
Article in English | MEDLINE | ID: mdl-28009066

ABSTRACT

The amniotic fluid has been identified as an untapped source of cells with broad potential, which possess immunomodulatory properties and do not have the ethical and legal limitations of embryonic stem cells. CD117(c-Kit)+ cells selected from amniotic fluid have been shown to differentiate into cell lineages representing all three embryonic germ layers without generating tumors, making them ideal candidates for regenerative medicine applications. Moreover, their ability to engraft in injured organs and modulate immune and repair responses of host tissues, suggest that transplantation of such cells may be useful for the treatment of various degenerative and inflammatory diseases. Although significant questions remain regarding the origin, heterogeneous phenotype, and expansion potential of amniotic fluid stem cells, evidence to date supports their potential role as a valuable stem cell source for the field of regenerative medicine. Stem Cells 2017;35:1663-1673.


Subject(s)
Amniotic Fluid/cytology , Cell Lineage/physiology , Regenerative Medicine/methods , Stem Cell Transplantation , Stem Cells/cytology , Amniocentesis , Amniotic Fluid/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Fetus , Gene Expression , Gestational Age , Humans , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Stem Cells/physiology
16.
Article in Korean | WPRIM (Western Pacific) | ID: wpr-211730

ABSTRACT

During the past decades, there has been a great evolution in the field of fetal therapy for congenital defects. Prenatal screening or diagnostic methods including non-invasive and invasive methods and fetal ultrasound have led to earlier and more accurate diagnosis of congenital anomalies. Recent advances in several therapeutic techniques including ultrasound-guided needle therapy, laser therapy or fetal endoscopy, have allowed some fetuses at risk with anatomical defects, to be corrected in utero but still, its clinical indications remain limited. Over the last 30 years, many researchers found usefulness of pluripotent stem cells from amniotic fluid and placenta because they are sources of diverse progenitor cell populations called mesenchymal stem cells. In some human conditions like severe combined immunodeficiency syndrome and chronic granulomatous disease, fetal therapy using stem cell replacement showed some promising results in researches but more studies are required to apply in clinical settings. The aim of this article is to summarize a current status and future perspective of stem cell therapy for treatment of congenital fetal anomalies.


Subject(s)
Female , Humans , Amniotic Fluid , Congenital Abnormalities , Diagnosis , Endoscopy , Fetal Therapies , Fetus , Granulomatous Disease, Chronic , Laser Therapy , Mesenchymal Stem Cells , Needles , Placenta , Pluripotent Stem Cells , Prenatal Diagnosis , Severe Combined Immunodeficiency , Stem Cells , Ultrasonography
17.
J Physiol Sci ; 66(6): 463-476, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27053101

ABSTRACT

We characterized human amniotic fluid stem cells (AFSC) in senescent cultures (6 weeks) versus cryopreserved cells using whole-cell patch-clamp, immunophenotyping, and differential gene expression profiling for senescence genes. We evidenced five ion current components (outward rectifier, A-type, inward rectifier, and big conductance Ca2+-dependent K+ currents, fast voltage-dependent Na+ currents). Senescent AFSC showed reduced expression of CD90, CD44, CD133, over 500-fold increase of interferon gamma and telomerase reverse transcriptase genes, increased cycle-dependent kinase 4 inhibitors, p53-binding protein 1, and decreased calreticulin and CD44. HLA-ABC immune expression was similar, and HLA-DR expression very low in both cell types. A subset of cryopreserved AFSC featured large inward rectifier K+ currents, voltage-dependent Na+ currents, and neural progenitor markers evidenced by immunophenotyping and RT-PCR. In all AFSC, in both culture conditions, at patch rupture the outward currents were very low, and they increased progressively over several minutes upon cytoplasm dialysis with pipette solution.


Subject(s)
Amniotic Fluid/cytology , Cryopreservation , Gene Expression Regulation/physiology , Stem Cells/physiology , Cell Culture Techniques , Cells, Cultured , Electrophysiological Phenomena , Humans , Immunophenotyping
18.
Int J Biol Sci ; 12(5): 558-68, 2016.
Article in English | MEDLINE | ID: mdl-27019637

ABSTRACT

Amniotic fluid stem cells (AFSCs) are a type of fetal stem cell whose stemness encompasses both embryonic and adult stem cells, suggesting that they may be easily and efficiently reprogrammed into induced pluripotent stem cells (iPSCs). To further simplify the reprogramming process, the creation of AFSC-derived iPSCs using a single factor is desirable. Here we report the generation of one-factor human AFSC-iPSCs (AiPSCs) from human AFSCs by ectopic expression of the transcription factor OCT4. Just like human embryonic stem cells, AiPSCs exhibited similar epigenetic status, global gene expression profiles, teratoma formation and in vitro & in vivo pluripotency. Our results indicate that the OCT4 is necessary and sufficient to directly reprogram human AFSCs into pluripotent AiPSCs. Moreover, reflecting the similar memory characteristics of AFSCs and neural stem cells, we show that AiPSC membrane-derived vesicles (MVs) repair cerebral ischemia damage. We anticipate that the successful generation of one-factor AiPSCs will facilitate the creation of patient-specific pluripotent stem cells without the need for transgenic expression of oncogenes. Moreover, MVs from tissue-specific AiPSCs have potential in tissue repair, representing a novel application of iPSCs.


Subject(s)
Amniotic Fluid/cytology , Brain Ischemia/metabolism , Brain Ischemia/therapy , Cellular Reprogramming/physiology , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred BALB C , Octamer Transcription Factor-3/genetics , Rats , Rats, Wistar
19.
Article in English | MEDLINE | ID: mdl-26542929

ABSTRACT

Regenerative medicine has recently been established as an emerging field focussing on repair, replacement or regeneration of cells, tissues and whole organs. The significant recent advances in the field have intensified the search for novel sources of stem cells with potential for therapy. Recently, researchers have identified the amniotic fluid as an untapped source of stem cells that are multipotent, possess immunomodulatory properties and do not have the ethical and legal limitations of embryonic stem cells. Stem cells from the amniotic fluid have been shown to differentiate into cell lineages representing all three embryonic germ layers without generating tumours, which make them an ideal candidate for tissue engineering applications. In addition, their ability to engraft in injured organs and modulate immune and repair responses of host tissues suggest that transplantation of such cells may be useful for the treatment of various degenerative and inflammatory diseases affecting major tissues/organs. This review summarises the evidence on amniotic fluid cells over the past 15 years and explores the potential therapeutic applications of amniotic fluid stem cells and amniotic fluid mesenchymal stem cells.


Subject(s)
Amniotic Fluid/cytology , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Humans , Mesenchymal Stem Cell Transplantation , Multipotent Stem Cells/transplantation , Regenerative Medicine , Stem Cell Transplantation , Stem Cells/cytology
20.
Cell Transplant ; 25(3): 615, 2016 03.
Article in English | MEDLINE | ID: mdl-28836829

ABSTRACT

Long-term engraftment and phenotype correction has been difficult to achieve in humans after in utero stem cell transplantation mainly because of allogeneic rejection. Autologous cells could be obtained during gestation from the amniotic fluid with minimal risk for the fetus and the mother. Using a sheep model, we explored the possibility of using amniotic fluid mesenchymal stem cells (AFMSCs) for autologous in utero stem cell/gene therapy. We collected amniotic fluid (AF) under ultrasound-guided amniocentesis in early gestation pregnant sheep ( n = 9, 58 days of gestation, term = 145 days). AFMSCs were isolated and expanded in all sampled fetal sheep. Those cells were transduced using an HIV vector encoding enhanced green fluorescent protein (GFP) with 63.2% (range 38.3-96.2%) transduction efficiency rate. After expansion, transduced AFMSCs were injected into the peritoneal cavity of each donor fetal sheep at 76 days under ultrasound guidance. One ewe miscarried twin fetuses after amniocentesis. Intraperitoneal injection was successful in the remaining 7 fetal sheep giving a 78% survival for the full procedure. Tissues were sampled at postmortem examination 2 weeks later. PCR analysis detected GFP-positive cells in fetal tissues including liver, heart, placenta, membrane, umbilical cord, adrenal gland, and muscle. GFP protein was detected in these tissues by Western blotting and further confirmed by cytofluorimetric and immunofluorescence analyses. This is the first demonstration of autologous stem cell transplantation in the fetus using AFMSCs. Autologous cells derived from AF showed widespread organ migration and could offer an alternative way to ameliorate prenatal congenital disease.

SELECTION OF CITATIONS
SEARCH DETAIL
...