Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.561
Filter
1.
Sci Rep ; 14(1): 12750, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830952

ABSTRACT

The current practice of restoring the anatomical structure in the treatment of pelvic floor dysfunction includes implantation of synthetic sling, which carries potential complications. This study aimed to develop biological substitutes to improve tissue function using scaffolds as a support to the host cells, through formation of new tissue. Human amniotic fluid stem cells (hAFSCs) were seeded on synthetic mesh-scaffold of AlloDerm Regenerative Tissue Matrix (RTM), Poly-DL-lactico-glycolic acid (PLGA) mesh (VICRYL) and Polydioxanone (PDS) meshes. In vitro study evaluates the metabolic activity of hAFSCs seeded mesh-scaffolds. In vivo study involving Sprague-Dawley rats was performed by assigning into 7 groups of sham control with fascia operation, AlloDerm implant, PDS implant, PLGA implant, AlloDerm harvest with hAFSC (AlloDerm-SC), PDS harvest with hAFSC(PDS-SC) and PLGS harvest with hAFSC (PGLA-SC). In vitro study reveals cell viability and proliferation of hAFSC on mesh scaffolds varies between meshes, with AlloDerm growing the fastest. The biomechanical properties of tissue-mesh-complex tension strength declined over time, showing highest tension strength on week-1, deteriorated similar to control group on week-12. All hAFSC-seeded mesh provides higher tension strength, compared to without. This study shed the potential of synthetic mesh as a scaffold for hAFSC for the surgical treatment of pelvic floor dysfunction.


Subject(s)
Amniotic Fluid , Rats, Sprague-Dawley , Stem Cells , Tissue Scaffolds , Animals , Tissue Scaffolds/chemistry , Humans , Amniotic Fluid/cytology , Rats , Stem Cells/cytology , Female , Plastic Surgery Procedures/methods , Tissue Engineering/methods , Surgical Mesh , Cell Proliferation , Pelvic Floor/surgery , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
2.
Clin Chem ; 70(6): 820-829, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38517460

ABSTRACT

BACKGROUND: Optical genome mapping (OGM) is a novel assay for detecting structural variants (SVs) and has been retrospectively evaluated for its performance. However, its prospective evaluation in prenatal diagnosis remains unreported. This study aimed to prospectively assess the technical concordance of OGM with standard of care (SOC) testing in prenatal diagnosis. METHODS: A prospective cohort of 204 pregnant women was enrolled in this study. Amniotic fluid samples from these women were subjected to OGM and SOC testing, which included chromosomal microarray analysis (CMA) and karyotyping (KT) in parallel. The diagnostic yield of OGM was evaluated, and the technical concordance between OGM and SOC testing was assessed. RESULTS: OGM successfully analyzed 204 cultured amniocyte samples, even with a cell count as low as 0.24 million. In total, 60 reportable SVs were identified through combined OGM and SOC testing, with 22 SVs detected by all 3 techniques. The diagnostic yield for OGM, CMA, and KT was 25% (51/204), 22.06% (45/204), and 18.14% (37/204), respectively. The highest diagnostic yield (29.41%, 60/204) was achieved when OGM and KT were used together. OGM demonstrated a concordance of 95.56% with CMA and 75.68% with KT in this cohort study. CONCLUSIONS: Our findings suggest that OGM can be effectively applied in prenatal diagnosis using cultured amniocytes and exhibits high concordance with SOC testing. The combined use of OGM and KT appears to yield the most promising diagnostic outcomes.


Subject(s)
Prenatal Diagnosis , Humans , Female , Pregnancy , Prospective Studies , Prenatal Diagnosis/methods , Adult , Karyotyping , Chromosome Mapping , Amniotic Fluid/chemistry , Amniotic Fluid/cytology
3.
J Reprod Immunol ; 152: 103656, 2022 08.
Article in English | MEDLINE | ID: mdl-35752068

ABSTRACT

Inflammatory cytokines contribute to the pathophysiology of preeclampsia. However, whether the imbalance of Th1/Th2 cytokines in amniotic fluid is associated with preeclampsia is not well defined. In the present study, we collected peripheral blood and amniotic fluid from normal pregnancy (n = 25) and preeclampsia (n = 22) at last trimester during cesarean section. The Th1/Th2 cytokine levels in amniotic fluid supernatant were detected by a bead-based immunoassay. The percentage of IFN-γ+CD4+ T cells, TNF-α+CD4+ T cells, IL-4+CD4+ T cells and IL-10+CD4+ T cells in peripheral blood was detected by flow cytometry. We found that in normal pregnancy, the IFN-γ/IL-4 and IFN-γ/IL-5 ratios were decreased in amniotic fluid supernatant compared to that in plasma, indicating a Th2 bias. However, IFN-γ/IL-4 (P = 0.014), IFN-γ/IL-5 (P = 0.005) and IFN-γ/IL-13 (P = 0.047) ratios in amniotic fluid supernatant was significantly increased in preeclampsia patients. The percentage of IFN-γ+CD4+ T cells (20.70 ± 7.61% vs 16.55 ± 4.96%, P = 0.041) and TNF-α+CD4+ T cells (31.78 ± 10.66% vs 19.47 ± 13.54%, P = 0.048) was significantly elevated in preeclampsia compared to normal pregnancy. Our finding demonstrates that a shift away from Th2 bias in amniotic fluid and circulating CD4+ T cells is involved in the pathogenesis of preeclampsia. This study suggests restoring the Th2 bias in amniotic fluid might be a therapeutic target of preeclampsia.


Subject(s)
Amniotic Fluid , Pre-Eclampsia , Th2 Cells , Amniotic Fluid/cytology , Amniotic Fluid/metabolism , Cesarean Section , Cytokines , Female , Humans , Interleukin-4 , Interleukin-5 , Pre-Eclampsia/diagnosis , Pre-Eclampsia/etiology , Pregnancy , Th1 Cells , Tumor Necrosis Factor-alpha
4.
FASEB J ; 36(4): e22218, 2022 04.
Article in English | MEDLINE | ID: mdl-35218567

ABSTRACT

An immunoregulatory role of stem cells, often mediated by their secretome, has been claimed by several studies. Stem cell-derived extracellular vesicles (EVs) are crucial components of the secretome. EVs, a heterogeneous group of membranous vesicles released by many cell types into the extracellular space, are now considered as an additional mechanism for intercellular communication. In this study, we aimed at investigating whether human amniotic stem cell-derived extracellular vesicles (HASC-EVs) were able to interfere with inflammasome activation in the THP-1 cell line. Two subsets of HASC-EVs were collected by sequential centrifugation, namely HASC-P10 and HASC-P100. We demonstrated that HASC-EVs were neither internalized into nor undertake a direct interaction with THP-1 cells. We showed that HASC-P10 and P100 were able to intrinsically produce ATP, which was further converted to adenosine by 5'-nucleotidase (CD73) and ectonucleoside triphosphate diphosphohydrolase-1 (CD39). We found that THP-1 cells conditioned with both types of HASC-EVs failed to activate the NLRP3/caspase-1/inflammasome platform in response to LPS and ATP treatment by a mechanism involving A2a adenosine receptor activation. These results support a role for HASC-EVs as independent metabolic units capable of modifying the cellular functions, leading to anti-inflammatory effects in monocytic cells.


Subject(s)
Amniotic Fluid/cytology , Anti-Inflammatory Agents/pharmacology , Extracellular Vesicles/metabolism , Inflammasomes/antagonists & inhibitors , Inflammation/prevention & control , Monocytes/cytology , Stem Cells/cytology , Adenosine/metabolism , Amniotic Fluid/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Monocytes/metabolism , Purinergic P1 Receptor Antagonists/pharmacology , Receptors, Purinergic P1/chemistry , Receptors, Purinergic P1/metabolism , Stem Cells/metabolism , THP-1 Cells
5.
Prenat Diagn ; 42(2): 157-163, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35001398

ABSTRACT

OBJECTIVE: Spina bifida is a debilitating neutral tube defect affecting many infants. The impact and severity of spina bifida depends on whether the patient exhibits a closed defect, spina bifida occulta, or an open defect, spina bifida aperta. Patients with spina bifida have physical and mental disabilities which merit further research into less invasive, more successful treatments. In addition to serving as protection for the growing fetus and facilitating nutrient exchange, amniotic fluid (AF) is a rich source of a mixed population of stem cells. As such, in vitro culture of AF-derived stem cells has shown promise among therapeutic and surgical applications. We present a critical evaluation of the current preclinical efforts, amniotic fluid-derived stem cell (AFSC) culture process, and the subsequent therapeutic application, with a focus on improvements for spina bifida outcomes in the pediatric patient population. METHOD: An evidence - based literature review to investigate the current literature surrounding AFSC culture and use, with an emphasis on the benefits for spina bifida treatment. RESULTS: 47 literature sources from PubMed and three studies from ClinicalTrials.gov. CONCLUSION: This review synthesizes the current literature, which shows promising data on AFSC pluripotency, as well as successful in utero coverage from AFSC - supported environments in a multitude of animal models.


Subject(s)
Amniotic Fluid/cytology , Spinal Dysraphism/therapy , Stem Cell Transplantation/methods , Animals , Cell Culture Techniques , Female , Humans , Pregnancy , Treatment Outcome
6.
Pediatr Res ; 91(1): 101-106, 2022 01.
Article in English | MEDLINE | ID: mdl-34561550

ABSTRACT

BACKGROUND: Stem cell therapy has been proven to rescue intestinal injury and stimulate intestinal regeneration in necrotizing enterocolitis (NEC). Specifically, stem cells derived from amniotic fluid (AFSCs) and mesenchymal stem cells (MSCs) derived from bone marrow have shown promising results in the treatment of experimental NEC. This study aims to examine the effects of AFSCs and MSCs on the prevention of intestinal injury during experimental NEC. METHODS: Supernatants from AFSC and MSC cultures were collected to perform proteomic analysis. Prior to NEC induction, mice received intraperitoneal injections of phosphate-buffered saline (PBS), 2 × 106 AFSCs, or 2 × 106 MSCs. RESULTS: We found that AFSCs grew faster than MSCs. Proteomic analysis indicated that AFSCs are primarily involved in cell development and growth, while MSCs are involved in immune regulation. Administering AFSCs before NEC induction decreased NEC severity and mucosal inflammation. Intestinal proliferation and endogenous stem cell activation were increased after AFSC administration. However, administering MSCs before NEC induction had no beneficial effects. CONCLUSIONS: This study demonstrated that AFSCs and MSCs have different protein release profiles. AFSCs can potentially be used as a preventative strategy for neonates at risk of NEC, while MSCs cannot be used. IMPACT: AFSCs and MSCs have distinct protein secretory profiles, and AFSCs are primarily involved in cell development and growth, while MSCs are involved in immune regulation. AFSCs are unique in transiently enhancing healthy intestinal epithelial cell growth, which offers protection against the development of experimental NEC. The prevention of NEC via the administration of AFSCs should be evaluated in infants at great risk of developing NEC or in infants with early signs of NEC.


Subject(s)
Amniotic Fluid/cytology , Stem Cell Transplantation , Animals , Enterocolitis, Necrotizing , Humans , Infant, Newborn , Mice
7.
Cells ; 10(12)2021 11 23.
Article in English | MEDLINE | ID: mdl-34943786

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) are currently one of the most extensively researched fields due to their promising opportunity for use in regenerative medicine. There are many sources of MSCs, of which cells of perinatal origin appear to be an invaluable pool. Compared to embryonic stem cells, they are devoid of ethical conflicts because they are derived from tissues surrounding the fetus and can be safely recovered from medical waste after delivery. Additionally, perinatal MSCs exhibit better self-renewal and differentiation properties than those derived from adult tissues. It is important to consider the anatomy of perinatal tissues and the general description of MSCs, including their isolation, differentiation, and characterization of different types of perinatal MSCs from both animals and humans (placenta, umbilical cord, amniotic fluid). Ultimately, signaling pathways are essential to consider regarding the clinical applications of MSCs. It is important to consider the origin of these cells, referring to the anatomical structure of the organs of origin, when describing the general and specific characteristics of the different types of MSCs as well as the pathways involved in differentiation.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Mesenchymal Stem Cells/cytology , Regenerative Medicine , Amniotic Fluid/cytology , Cell Self Renewal/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Female , Humans , Mesenchymal Stem Cell Transplantation , Placenta/cytology , Placenta/transplantation , Pregnancy , Umbilical Cord/cytology , Umbilical Cord/transplantation
8.
Eur Rev Med Pharmacol Sci ; 25(1 Suppl): 49-55, 2021 12.
Article in English | MEDLINE | ID: mdl-34890034

ABSTRACT

OBJECTIVE: The amniotic fluid contains a large population of stem keratinocytes demonstrating minimal immunological rejection. Recent evidence suggests that stem cells from the amniotic fluid can be employed in the field of tissue engineering. In this work we identified precursors of the epithelial cells and expanded them in vitro. MATERIALS AND METHODS: After collecting samples of amniotic fluid and separating the cells via centrifugation, we seeded a portion of these cells in selection media to analyze the proliferation of epithelial cells. The stem cells precursors of keratinocytes were identified through specific markers. The expression of these markers was evaluated by immunofluorescence and reverse transcription polymerase chain reaction (PCR). RESULTS: The stem cells demonstrated 90% confluence, after undergoing proliferation in the selection medium for 15 days. Most of these cells tested positive for the keratinocyte-specific markers, but negative for stem cell specific markers. Of note, the identity of the keratinocytes was well established even after several subcultures. CONCLUSIONS: These results suggested that it is feasible to isolate and expand differentiated cell populations in the amniotic fluid from precursor cells. Furthermore, amniotic membranes can be utilized as scaffolds to grow keratinocytes, which can be potentially exploited in areas of skin ulcer transplantation and tissue engineering interventions.


Subject(s)
Amnion/cytology , Amnion/physiology , Amniotic Fluid/cytology , Amniotic Fluid/physiology , Keratinocytes/physiology , Skin Ulcer/therapy , Adult , Amnion/transplantation , Cell Proliferation/physiology , Cells, Cultured , Embryonic Stem Cells/physiology , Embryonic Stem Cells/transplantation , Female , Humans , Keratinocytes/transplantation , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
9.
Mutat Res Rev Mutat Res ; 788: 108399, 2021.
Article in English | MEDLINE | ID: mdl-34893150

ABSTRACT

During pregnancy several types of fetal cells and fetal stem cells, including pregnancy-associated progenitor cells (PAPCs), traffic into the maternal circulation. Whereas they also migrate to various maternal organs and adopt the phenotype of the target tissues to contribute to regenerative processes, fetal cells also play a role in the pathogenesis of maternal diseases. In addition, cell-free fetal DNA (cffDNA) is detectable in the plasma of pregnant women. Together they constitute the well-known phenomenon of fetomaternal microchimerism, which inspired the concept of non-invasive prenatal testing (NIPT) using maternal blood. An in-depth knowledge concerning the origins of these fetal cells and cffDNA allows a more comprehensive understanding of the biological relevance of fetomaternal microchimerism and has implications for the ongoing expansion of resultant clinical applications.


Subject(s)
Chimerism , Noninvasive Prenatal Testing/methods , Pregnancy Complications/genetics , Amniotic Fluid/cytology , Cell Movement , Cell-Free Nucleic Acids/genetics , Female , Humans , Pregnancy , Pregnancy Complications/diagnosis , Stem Cells/metabolism , Stem Cells/physiology
10.
Gac. méd. espirit ; 23(3): [12], dic. 2021.
Article in Spanish | LILACS | ID: biblio-1404882

ABSTRACT

RESUMEN Fundamento: El cultivo celular permite el análisis directo de las células vivas mediante un microscopio. El estudio de las células contenidas en el líquido amniótico, mediante técnicas de cultivo, detecta anomalías en número y morfología de los cromosomas, que pueden relacionarse con enfermedades genéticas. Objetivo: Caracterizar las variedades de cultivo de líquido amniótico para el diagnóstico in vitro de poliploidías. Metodología: Se realizó un estudio descriptivo transversal, en el Centro Provincial de Genética Médica de Camagüey, en el periodo de noviembre de 2016 a abril de 2018.La población de estudio estuvo constituida por 1571 muestras útiles de líquido amniótico obtenidas por amniocentesis, en gestantes en el segundo trimestre, evaluadas en consulta multidisciplinaria con criterios clínicos de estudios cromosómicos según lo establecido en el diagnóstico prenatal citogenético, previo consentimiento informado. Se utilizaron 20 mL de líquido amniótico para la siembra de células fetales, y se aplicaron tres variantes de cultivo abierto (directo, centrifugado y expandido). Se determinó el complemento cromosómico en cada variedad. Resultados: Predominó el complemento cromosómico normal. Las tetraploidías prevalecieron en el cultivo expandido. El índice mitótico fue similar en las tres variedades de cultivo y el cultivo directo tuvo el más bajo índice de poliploidías. Conclusiones: El cariotipo normal fue predominante. Las tetrapolidías fueron las alteraciones más frecuentes y prevalecieron en el cultivo expandido. En el cultivo directo se presentó el más bajo índice de errores inducidos in vitro.


ABSTRACT Background: Cell culture allows direct analysis of live cells under a microscope. The cell study contained in amniotic fluid, by culture techniques, detects abnormalities in chromosome number and morphology, which can be related to genetic diseases. Objective: To describe amniotic fluid culture strains for the in vitro diagnosis of polyploidy. Methodology: A cross-sectional descriptive study was conducted at the Camagüey Provincial Center of Medical Genetics, from November 2016 to April 2018.The study population consisted of 1571 useful amniotic fluid samples obtained by amniocentesis, in pregnant women in the second trimester, evaluated by multidisciplinary discussion with clinical criteria for chromosomal studies as established in the cytogenetic prenatal diagnosis, prior informed consent. 20 mL of amniotic fluid were used for fetal cell seeding, and three open culture strains (direct, centrifuged and expanded) were applied. Chromosomal complement was determined in each variety. Results: Normal chromosome complement was predominant. Tetraploidy prevailed in the expanded culture. The mitotic index was similar in the three culture strains and the direct culture had the lowest polyploidy index. Conclusions: Normal karyotype was predominant. Tetraploidy were the most frequent modifications and prevailed in the expanded culture. Direct culture had the lowest rate of the in vitro induced errors.


Subject(s)
Polyploidy , Tetraploidy , Blood Culture , Amniotic Fluid/cytology
11.
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
12.
Medicine (Baltimore) ; 100(25): e26331, 2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34160397

ABSTRACT

ABSTRACT: Mosaicism can be observed in karyotype analyses of amniotic fluid cells. Differentiating between true mosaicism and pseudomosaicism and determining mosaic proportions can help avoid misjudgments by doctors and effectively reduce mental and physical harm to pregnant women. However, the detection of mosaicism and mosaic proportions via karyotype analysis and fluorescence in situ hybridization (FISH) is extremely complex. We have developed a novel approach, "segmental duplication quantitative fluorescent PCR" (SD-QF-PCR), to detect mosaicism and mosaic proportions.In this study, twenty control samples and fourteen mosaic samples were tested by first-line karyotype analysis; by second-line karyotype analysis, SD-QF-PCR and FISH were used to reassess fetal sex chromosome mosaicism and mosaic proportions.Detection of the 20 control samples by second-line karyotype analysis via FISH and SD-QF-PCR showed normal and consistent results. Among the 14 mosaic samples, the numbers of samples showing true mosaicism and pseudomosaicism detected by the three methods were 6 and 8, respectively.Our study demonstrates that SD-QF-PCR can be used as a complementary method to traditional cytogenetic analysis of amniotic fluid mosaics and has clinical application value.


Subject(s)
Karyotyping/methods , Mosaicism , Polymerase Chain Reaction/methods , Prenatal Diagnosis/methods , Sex Chromosome Aberrations , Amniocentesis , Amniotic Fluid/cytology , Cells, Cultured , Feasibility Studies , Female , Humans , In Situ Hybridization, Fluorescence , Pregnancy , Primary Cell Culture
13.
Mol Biol Rep ; 48(5): 3913-3923, 2021 May.
Article in English | MEDLINE | ID: mdl-34050503

ABSTRACT

The caprine mesenchymal stem cells (MSCs) derived from fetal adnexa are highly proliferative. These cells possess tri-lineage differentiation potential and express MSC surface antigens and pluripotency markers with a wound-healing potential. This present study was conducted to compare the immunomodulatory potential of caprine MSCs derived from the fetal adnexa. Mid-gestation caprine uteri (2-3 months) were collected from the abattoir to isolate MSCs from amniotic fluid (cAF), amniotic sac (cAS), Wharton's jelly (cWJ) and cord blood (cCB), which were expanded and characterized at the 3rd passage. These MSCs were then stimulated with inflammatory cytokines (IFN-γ and TNF-α) to assess the percentage of inhibition produced on peripheral blood mononuclear cells (PBMCs) proliferation. The percentage of inhibition on activated PBMCs proliferation produced by cWJ MSCs and cAS MSCs was significantly higher than cCB and cAF MSCs. The relative mRNA expression profile and immunofluorescent localization of different immunomodulatory cytokines and growth factors were conducted upon stimulation. The mRNA expression profile of a set of different cytokines and growth factors in each caprine fetal adnexa MSCs were modulated. Indoleamine 2, 3 dioxygenase appeared to be the major immunomodulator in cWJ, cAF, and cCB MSCs whereas inducible nitric oxide synthase in cAS MSCs. This study suggests that caprine MSCs derived from fetal adnexa display variable immunomodulatory potential, which appears to be modulated by different molecules among sources.


Subject(s)
Adnexa Uteri/metabolism , Immunomodulation/immunology , Mesenchymal Stem Cells/immunology , Adnexa Uteri/immunology , Adnexa Uteri/physiology , Amniotic Fluid/cytology , Animals , Cell Differentiation/immunology , Cell Proliferation/physiology , Cells, Cultured , Female , Fetal Blood/immunology , Gene Expression/genetics , Goats , Transcriptome/genetics , Transcriptome/immunology , Umbilical Cord/cytology , Wharton Jelly/cytology
14.
Int J Mol Sci ; 22(7)2021 Apr 02.
Article in English | MEDLINE | ID: mdl-33918297

ABSTRACT

We previously reported that c-KIT+ human amniotic-fluid derived stem cells obtained from leftover samples of routine II trimester prenatal diagnosis (fetal hAFS) are endowed with regenerative paracrine potential driving pro-survival, anti-fibrotic and proliferative effects. hAFS may also be isolated from III trimester clinical waste samples during scheduled C-sections (perinatal hAFS), thus offering a more easily accessible alternative when compared to fetal hAFS. Nonetheless, little is known about the paracrine profile of perinatal hAFS. Here we provide a detailed characterization of the hAFS total secretome (i.e., the entirety of soluble paracrine factors released by cells in the conditioned medium, hAFS-CM) and the extracellular vesicles (hAFS-EVs) within it, from II trimester fetal- versus III trimester perinatal cells. Fetal- and perinatal hAFS were characterized and subject to hypoxic preconditioning to enhance their paracrine potential. hAFS-CM and hAFS-EV formulations were analyzed for protein and chemokine/cytokine content, and the EV cargo was further investigated by RNA sequencing. The phenotype of fetal- and perinatal hAFS, along with their corresponding secretome formulations, overlapped; yet, fetal hAFS showed immature oxidative phosphorylation activity when compared to perinatal ones. The profiling of their paracrine cargo revealed some differences according to gestational stage and hypoxic preconditioning. Both cell sources provided formulations enriched with neurotrophic, immunomodulatory, anti-fibrotic and endothelial stimulating factors, and the immature fetal hAFS secretome was defined by a more pronounced pro-vasculogenic, regenerative, pro-resolving and anti-aging profile. Small RNA profiling showed microRNA enrichment in both fetal- and perinatal hAFS-EV cargo, with a stably- expressed pro-resolving core as a reference molecular signature. Here we confirm that hAFS represents an appealing source of regenerative paracrine factors; the selection of either fetal or perinatal hAFS secretome formulations for future paracrine therapy should be evaluated considering the specific clinical scenario.


Subject(s)
Fetal Stem Cells/metabolism , Pregnancy Trimester, Second/metabolism , Pregnancy Trimester, Third/metabolism , Proteome , Adult , Amniotic Fluid/cytology , Bodily Secretions , Extracellular Vesicles/ultrastructure , Female , Humans , Hypoxia/metabolism , Pregnancy
15.
J Reprod Immunol ; 145: 103319, 2021 06.
Article in English | MEDLINE | ID: mdl-33848896

ABSTRACT

Preterm birth (PTB) is one of the most frequent pregnancy complications. It affects millions of babies each year worldwide and is associated with increased morbidity and mortality. PTB-associated alterations in the maternal immune response may have a direct effect on the developing fetal immune system. Having recently shown that B regulatory (Breg) cells are decreased in number and functionally impaired in maternal blood from women delivering preterm, we now addressed the question whether the adaptive immune system is also altered in cord blood (CB) after the onset of PTB. PTB was associated with increased concentrations of IL-6, TNF-α and IL-21 in CB and enhanced IL-6, but decreased IFN-γ and IL-4 in amniotic fluid (AF) samples compared to term delivery (TD). We found no differences in the frequency of CD19 + B cells, CD4 + T cells or CD4+Foxp3+CD25+ T regulatory (Treg) cells in CB cells in PTB vs TD. The frequency of CD86 + B cells was increased, while the percentage of CD24hiCD38hiCD19 + Breg and CD1dhiCD5+ Breg cells and the ability of B cells to convert into Breg cells was diminished in PTB compared to TD. CB B cells from PTB secreted more IL-6, TNF-α, IL-9 and IL-2 compared to B cells obtained from term samples. We conclude that, after PTB onset, a shift from immunoregulation towards inflammation takes place in CB cells that are reportedly representative of the fetal compartment. B cells have a substantial contribution herein. This phenomenon might account for the observed enhanced mortality and morbidity in prematurely born infants. Further studies will clarify how to employ this easy-to-obtain information for closely monitoring newborns at risk.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Fetal Blood/immunology , Premature Birth/immunology , Adult , Amniotic Fluid/cytology , Amniotic Fluid/immunology , B-Lymphocytes, Regulatory/metabolism , Case-Control Studies , Female , Fetal Blood/cytology , Humans , Infant, Newborn , Male , Pregnancy , Premature Birth/blood , Term Birth/blood , Term Birth/immunology
16.
Sci Rep ; 11(1): 9158, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33911155

ABSTRACT

Spinal muscular atrophy (SMA) is a single gene disorder affecting motor function in uterus. Amniotic fluid is an alternative source of stem cell to ameliorate SMA. Therefore, this study aims to examine the therapeutic potential of Human amniotic fluid stem cell (hAFSC) for SMA. Our SMA model mice were generated by deletion of exon 7 of Smn gene and knock-in of human SMN2. A total of 16 SMA model mice were injected with 1 × 105 hAFSC in uterus, and the other 16 mice served as the negative control. Motor function was analyzed by three behavioral tests. Engraftment of hAFSC in organs were assessed by flow cytometry and RNA scope. Frequency of myocytes, neurons and innervated receptors were estimated by staining. With hAFSC transplantation, 15 fetuses survived (93.75% survival) and showed better performance in all motor function tests. Higher engraftment frequency were observed in muscle and liver. Besides, the muscle with hAFSC transplantation expressed much laminin α and PAX-7. Significantly higher frequency of myocytes, neurons and innervated receptors were observed. In our study, hAFSC engrafted on neuromuscular organs and improved cellular and behavioral outcomes of SMA model mice. This fetal therapy could preserve the time window and treat in the uterus.


Subject(s)
Amniotic Fluid/cytology , Spinal Muscular Atrophies of Childhood/therapy , Stem Cell Transplantation/methods , Adult , Animals , Disease Models, Animal , Female , Humans , Mice, Transgenic , Neurons/physiology , Pregnancy , Spinal Muscular Atrophies of Childhood/etiology , Stem Cells/cytology , Stem Cells/physiology
17.
In Vitro Cell Dev Biol Anim ; 57(4): 457-467, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33721206

ABSTRACT

The purpose of this study is to observe the effect of icariside II (ICS II) on the differentiation of human amniotic mesenchymal stem cells (hAMSCs) into dopaminergic neuron-like cells, the involvement of PI3K signaling pathway inhibitors. After identifying hAMSCs by flow cytometry, hAMSCs were induced and treated with ICS II at 10 µmol/L, 3 µmol/L, 1 µmol/L, and 0 µmol/L. hAMSCs in the LY294002+3µM ICS II group were pretreated with 20 µmol/L LY294002, a PI3K-specific inhibitor, for 1 h, and then hAMSCs were induced with 3 µmol/L ICS II. On the 21st day of induction, immunofluorescence was used to detect expression of the neuronal nuclei (NeuN), neuron-specific enolase (NSE), microtubule-associated protein-2 (MAP-2), glial fibrillary acidic protein (GFAP), and tyrosine hydroxylase (TH) antigens in each induced cell group. Western blotting was used to detect the relative protein expression of NSE, MAP-2, GFAP, and TH. ELISA was used to detect the dopamine concentration in the induction medium supernatant of each group. After 21 d of ICS II induction, immunofluorescence showed that GFAP expression was not obvious in any hAMSC group. The NeuN, NSE, MAP-2, and TH fluorescent proteins were expressed in each group. NeuN was expressed in the nucleus and cytoplasm, while NSE, MAP-2, and TH were mainly expressed in the cytoplasm. The positive cell rates of NeuN, NSE, MAP-2, and TH in the 10 µmol/L, 3 µmol/L, and 1 µmol/L ICS II groups were higher than those in the LY294002+3µM ICS II and control groups. After 21 d of induction, the Western blot results showed that the protein expression levels of NSE, MAP-2, and TH in the 10 µmol/L, 3 µmol/L, and 1 µmol/L ICS II groups were significantly higher than those in the LY294002+3µM ICS II and control groups. The MAP-2 protein expression levels in the 10 µmol/L and 3 µmol/L groups were higher than that in the 1 µmol/L group. After 21 d of induction, the dopamine concentrations in the culture supernatants of the 10 µmol/L, 3 µmol/L, and 1 µmol/L ICS II groups were higher than those in the LY294002+3µM ICS II and control groups. In our experiment, ICS II induced hAMSCs to differentiate into dopaminergic neuron-like cells, and the optimal concentration range of ICS II was 3-10 µmol/L. Moreover, the PI3K signaling pathway is involved in the above differentiation process.


Subject(s)
Cell Differentiation/drug effects , Dopaminergic Neurons/cytology , Flavonoids/pharmacology , Mesenchymal Stem Cells/cytology , Amnion/cytology , Amniotic Fluid/cytology , Carrier Proteins/genetics , Gene Expression Regulation, Developmental/drug effects , Glial Fibrillary Acidic Protein/genetics , Humans , Mesenchymal Stem Cells/drug effects , Microtubule-Associated Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/drug effects , Tyrosine 3-Monooxygenase/genetics
18.
Pediatr Surg Int ; 37(3): 301-309, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33566163

ABSTRACT

PURPOSE: Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease. Amniotic fluid stem cells (AFSC) improve NEC injury but human translation remains difficult. We aimed to evaluate the use of extracellular vesicles (EV) derived from human AFSC. METHODS: Human AFSC (hAFSC) were cultured according to the protocol (Celprogen Inc., California, U.S.A.). Conditioned medium was obtained, ultra-centrifuged, and EV were suspended in phosphate-buffered saline (PBS). C57BL/6 pups were grouped into: (1) breast-fed (Control, n = 11); (2) NEC + placebo (NEC + PBS; n = 10); and (3) NEC + treatment (NEC + EV; n = 11). NEC was induced post-natal days P5-9 by (A) gavage feeding hyperosmolar formula; (B) hypoxia for 10 min; and (C) lipopolysaccharide. Intra-peritoneal injections of PBS or hAFSC-EV were given on P6-7. All animals were sacrificed on P9 and terminal ileum harvested. RESULTS: hAFSC-EV administration reduced intestinal injury (p = 0.0048), NEC incidence (score ≥ 2), and intestinal inflammation (IL-6 p < 0.0001; TNF-α p < 0.0001). Intestinal stem cell expression (Lgr5 +) and cellular proliferation (Ki67) were enhanced above control levels following hAFSC-EV administration (Lgr5 p = 0.0003; Ki67 p < 0.0001). CONCLUSION: hAFSC-EV administration reduced intestinal NEC injury and inflammation while increasing stem cell expression and cellular proliferation. hAFSC-EV administration may induce similar beneficial effects to exogenous stem cells.


Subject(s)
Amniotic Fluid/cytology , Enterocolitis, Necrotizing/metabolism , Extracellular Vesicles/metabolism , Animals , Animals, Newborn , Cell Proliferation , Disease Models, Animal , Enterocolitis, Necrotizing/therapy , Female , Humans , Ileum/metabolism , Infant, Newborn , Infant, Newborn, Diseases , Inflammation/metabolism , Intestines , Mice , Mice, Inbred C57BL , Stem Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Sci Rep ; 11(1): 3063, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542256

ABSTRACT

Articular chondral lesions, caused either by trauma or chronic cartilage diseases such as osteoarthritis, present very low ability to self-regenerate. Thus, their current management is basically symptomatic, progressing very often to invasive procedures or even arthroplasties. The use of amniotic fluid stem cells (AFSCs), due to their multipotentiality and plasticity, associated with scaffolds, is a promising alternative for the reconstruction of articular cartilage. Therefore, this study aimed to investigate the chondrogenic potential of AFSCs in a micromass system (high-density cell culture) under insulin-like growth factor 1 (IGF-1) stimuli, as well as to look at their potential to differentiate directly when cultured in a porous chitosan-xanthan (CX) scaffold. The experiments were performed with a CD117 positive cell population, with expression of markers (CD117, SSEA-4, Oct-4 and NANOG), selected from AFSCs, after immunomagnetic separation. The cells were cultured in both a micromass system and directly in the scaffold, in the presence of IGF-1. Differentiation to chondrocytes was confirmed by histology and by using immunohistochemistry. The construct cell-scaffold was also analyzed by scanning electron microscopy (SEM). The results demonstrated the chondrogenic potential of AFSCs cultivated directly in CX scaffolds and also in the micromass system. Such findings support and stimulate future studies using these constructs in osteoarthritic animal models.


Subject(s)
Adult Stem Cells/cytology , Cartilage, Articular/drug effects , Chondrogenesis/genetics , Osteoarthritis/genetics , Tissue Scaffolds/chemistry , Adult Stem Cells/transplantation , Amniotic Fluid/cytology , Cartilage, Articular/growth & development , Cartilage, Articular/ultrastructure , Cell Culture Techniques , Cell Differentiation/drug effects , Chitosan/pharmacology , Gene Expression Regulation, Developmental/drug effects , Humans , Insulin-Like Growth Factor I/pharmacology , Microscopy, Electron, Scanning , Osteoarthritis/pathology , Osteoarthritis/therapy , Polysaccharides, Bacterial/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Tissue Engineering/methods
20.
Cells ; 10(1)2021 01 15.
Article in English | MEDLINE | ID: mdl-33467440

ABSTRACT

Human amniotic fluid stem cells (hAFSCs) are broadly multipotent immature progenitor cells with high self-renewal and no tumorigenic properties. These cells, even amplified, present very variable morphology, density, intracellular composition and stemness potential, and this heterogeneity can hinder their characterization and potential use in regenerative medicine. Celector® (Stem Sel ltd.) is a new technology that exploits the Non-Equilibrium Earth Gravity Assisted Field Flow Fractionation principles to characterize and label-free sort stem cells based on their solely physical characteristics without any manipulation. Viable cells are collected and used for further studies or direct applications. In order to understand the intrapopulation heterogeneity, various fractions of hAFSCs were isolated using the Celector® profile and live imaging feature. The gene expression profile of each fraction was analysed using whole-transcriptome sequencing (RNAseq). Gene Set Enrichment Analysis identified significant differential expression in pathways related to Stemness, DNA repair, E2F targets, G2M checkpoint, hypoxia, EM transition, mTORC1 signalling, Unfold Protein Response and p53 signalling. These differences were validated by RT-PCR, immunofluorescence and differentiation assays. Interestingly, the different fractions showed distinct and unique stemness properties. These results suggest the existence of deep intra-population differences that can influence the stemness profile of hAFSCs. This study represents a proof-of-concept of the importance of selecting certain cellular fractions with the highest potential to use in regenerative medicine.


Subject(s)
Amniotic Fluid/cytology , Stem Cells/cytology , Cell Differentiation , Cell Proliferation , Cells, Cultured , DNA Repair , Gene Expression Profiling , Humans , Leukocytes, Mononuclear/cytology , Multipotent Stem Cells/cytology , RNA-Seq , Regenerative Medicine , Signal Transduction , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...