Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.371
Filter
1.
Elife ; 122024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829685

ABSTRACT

Precision gene editing in primary hematopoietic stem and progenitor cells (HSPCs) would facilitate both curative treatments for monogenic disorders as well as disease modelling. Precise efficiencies even with the CRISPR/Cas system, however, remain limited. Through an optimization of guide RNA delivery, donor design, and additives, we have now obtained mean precise editing efficiencies >90% on primary cord blood HSCPs with minimal toxicity and without observed off-target editing. The main protocol modifications needed to achieve such high efficiencies were the addition of the DNA-PK inhibitor AZD7648, and the inclusion of spacer-breaking silent mutations in the donor in addition to mutations disrupting the PAM sequence. Critically, editing was even across the progenitor hierarchy, did not substantially distort the hierarchy or affect lineage outputs in colony-forming cell assays or the frequency of high self-renewal potential long-term culture initiating cells. As modelling of many diseases requires heterozygosity, we also demonstrated that the overall editing and zygosity can be tuned by adding in defined mixtures of mutant and wild-type donors. With these optimizations, editing at near-perfect efficiency can now be accomplished directly in human HSPCs. This will open new avenues in both therapeutic strategies and disease modelling.


Subject(s)
Gene Editing , Hematopoietic Stem Cells , Humans , Gene Editing/methods , CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems/genetics , Fetal Blood/cytology , Cells, Cultured
2.
J Neuroinflammation ; 21(1): 121, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720368

ABSTRACT

BACKGROUND: Umbilical cord blood (UCB) cells are a promising treatment for preterm brain injury. Access to allogeneic sources of UCB cells offer the potential for early administration to optimise their therapeutic capacities. As preterm infants often require ventilatory support, which can contribute to preterm brain injury, we investigated the efficacy of early UCB cell administration following ventilation to reduce white matter inflammation and injury. METHODS: Preterm fetal sheep (0.85 gestation) were randomly allocated to no ventilation (SHAM; n = 5) or 15 min ex utero high tidal volume ventilation. One hour following ventilation, fetuses were randomly allocated to i.v. administration of saline (VENT; n = 7) or allogeneic term-derived UCB cells (24.5 ± 5.0 million cells/kg; VENT + UCB; n = 7). Twenty-four hours after ventilation, lambs were delivered for magnetic resonance imaging and post-mortem brain tissue collected. Arterial plasma was collected throughout the experiment for cytokine analyses. To further investigate the results from the in vivo study, mononuclear cells (MNCs) isolated from human UCB were subjected to in vitro cytokine-spiked culture medium (TNFα and/or IFNγ; 10 ng/mL; n = 3/group) for 16 h then supernatant and cells collected for protein and mRNA assessments respectively. RESULTS: In VENT + UCB lambs, systemic IFNγ levels increased and by 24 h, there was white matter neuroglial activation, vascular damage, reduced oligodendrocytes, and increased average, radial and mean diffusivity compared to VENT and SHAM. No evidence of white matter inflammation or injury was present in VENT lambs, except for mRNA downregulation of OCLN and CLDN1 compared to SHAM. In vitro, MNCs subjected to TNFα and/or IFNγ displayed both pro- and anti-inflammatory characteristics indicated by changes in cytokine (IL-18 & IL-10) and growth factor (BDNF & VEGF) gene and protein expression compared to controls. CONCLUSIONS: UCB cells administered early after brief high tidal volume ventilation in preterm fetal sheep causes white matter injury, and the mechanisms underlying these changes are likely dysregulated responses of the UCB cells to the degree of injury/inflammation already present. If immunomodulatory therapies such as UCB cells are to become a therapeutic strategy for preterm brain injury, especially after ventilation, our study suggests that the inflammatory state of the preterm infant should be considered when timing UCB cells administration.


Subject(s)
Tidal Volume , Animals , Sheep , Female , Humans , Tidal Volume/physiology , Fetal Blood/cytology , Pregnancy , Cytokines/metabolism , Cord Blood Stem Cell Transplantation/methods , Respiration, Artificial/methods , Respiration, Artificial/adverse effects , Animals, Newborn
3.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(2): 577-582, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38660869

ABSTRACT

OBJECTIVE: To explore the optimal storage condition and time of umbilical cord blood from collection to preparation. METHODS: Collect cord blood samples from 30 healthy newborns, with each new born's umbilical cord blood was divided into two parts on average. One part was stored in cold storage (4 ℃) and the other was stored at room temperature (20-24 ℃). Samples were taken at 24, 36, 48, 60 and 72 h, respectively, total nucleated cells (TNC) count and TNC viability was analyzed. Flow cytometry was used to detect the ratio of viable CD34+ cells to viable CD45+ cells and viability of CD34+ cells, and colony-forming unit-granulocyte-macrophage (CFU-GM) count was performed by hematopoietic progenitor cell colony culture. The change trend of each index over time was observed, and the differences in each index was compared between cold storage and room temperature storage under the same storage time. RESULTS: The TNC count (r 4 ℃=-0.9588, r 20-24 ℃=-0.9790), TNC viability (r 4 ℃=-0.9941, r 20-24 ℃=-0.9970), CD34+ cells viability (r 4 ℃=-0.9932, r 20-24 ℃=-0.9828) of cord blood stored in cold storage (4 ℃) and room temperature storage (20-24 ℃) showed a consistent downward trend with the prolongation of storage time. The percentage of viable CD34+ cells (r 4 ℃=0.9169, r 20-24 ℃=0.7470) and CFU-GM count (r 4 ℃=-0.2537, r 20-24 ℃=-0.8098) did not show consistent trends. When the storage time was the same, the TNC count, TNC viability, CD34+ cells viability and CFU-GM count of cord blood stored in cold storage were higher than those stored at room temperature. Under the same storage time (24, 36, 48, 60 or 72 h), TNC viability in room temperature storage was significantly lower than that in cold storage (P <0.001), but TNC count, percentage of viable CD34+ cells and CFU-GM count were not significantly different between room temperature storage and cold storage. When stored at room temperature for 24 h and 36 h, the viability of CD34+ cells was significantly lower than that in cold storage (P <0.001, P <0.01), when the storage time for 48, 60 and 72 h, there was no significant difference in the CD34+ cells viability between room temperature storage and cold storage. CONCLUSION: It is recommended that cord blood be stored in cold storage (4 ℃) from collection to preparation, and processed as soon as possible.


Subject(s)
Antigens, CD34 , Blood Preservation , Fetal Blood , Humans , Fetal Blood/cytology , Infant, Newborn , Time Factors , Flow Cytometry , Hematopoietic Stem Cells/cytology , Cell Survival , Temperature , Blood Specimen Collection
4.
Int J Mol Sci ; 25(8)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38674031

ABSTRACT

Hemangioblasts give rise to endothelial progenitor cells (EPCs), which also express the cell surface markers CD133 and c-kit. They may differentiate into the outgrowth endothelial cells (OECs) that control neovascularization in the developing embryo. According to numerous studies, reduced levels of EPCs in circulation have been linked to human cardiovascular disorders. Furthermore, preeclampsia and senescence have been linked to levels of EPCs produced from cord blood. Uncertainties surround how preeclampsia affects the way EPCs function. It is reasonable to speculate that preeclampsia may have an impact on the function of fetal EPCs during the in utero period; however, the present literature suggests that maternal vasculopathies, including preeclampsia, damage fetal circulation. Additionally, the differentiation potential and general activity of EPCs may serve as an indicator of the health of the fetal vascular system as they promote neovascularization and repair during pregnancy. Thus, the purpose of this review is to compare-through the assessment of their quantity, differentiation potency, angiogenic activity, and senescence-the angiogenic function of fetal EPCs obtained from cord blood for normal and pregnancy problems (preeclampsia, gestational diabetes mellitus, and fetal growth restriction). This will shed light on the relationship between the angiogenic function of fetal EPCs and pregnancy complications, which could have an effect on the management of long-term health issues like metabolic and cardiovascular disorders in offspring with abnormal vasculature development.


Subject(s)
Diabetes, Gestational , Endothelial Progenitor Cells , Fetal Blood , Fetal Growth Retardation , Pre-Eclampsia , Humans , Pregnancy , Female , Diabetes, Gestational/metabolism , Diabetes, Gestational/blood , Pre-Eclampsia/blood , Endothelial Progenitor Cells/metabolism , Fetal Blood/cytology , Fetal Blood/metabolism , Fetal Growth Retardation/pathology , Cell Differentiation
5.
Curr Opin Hematol ; 31(4): 168-174, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38602152

ABSTRACT

PURPOSE OF REVIEW: Here, we review classic and emerging uses of umbilical cord blood and highlight strategies to improve its utility, focusing on selection of the appropriate units and cell types for the intended applications. RECENT LITERATURE: Recent studies have shown advancements in cord blood cell utility in a variety of cellular therapies and have made strides in elucidating manners to select the best units for therapy and target new ways to improve the various cell subpopulations for their respective applications. SUMMARY: Umbilical cord blood is a proven source of cells for hematopoietic cell transplantation and research and is an important potential source for additional cellular therapies. However, cord blood utility is limited by low "doses" of potent cells that can be obtained from individual units, a limitation that is specific to cord blood as a donor source. In addition to traditional CD34 + progenitor cells, cord blood lymphocytes are being pursued as therapeutic entities with their own unique properties and characteristics. Thus, selection of ideal units depends on the intended therapeutic entity and target, and identification of differential potency parameters is critical to drive effective banking strategies accommodating successful clinical use of cord blood in broader cell therapy settings.


Subject(s)
Cord Blood Stem Cell Transplantation , Fetal Blood , Humans , Fetal Blood/cytology , Cord Blood Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism
6.
Eur J Haematol ; 113(1): 127-129, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38644613

ABSTRACT

While there is intense interest in the production of allogeneic CAR-T cells from umbilical cord units, little is known about the reactivity and persistence of CAR-T cells of umbilical origin. We report the case of a patient at our hematological center with multiple relapsing Ph+ B-ALL, notably a Blinatunomab non-responder, who underwent therapy with Brexucabtagene Autoleucel following relapse on Ponatinib post-allogeneic hematopoietic stem cell transplantation. The patient achieved a rapid CAR-T expansion and durable remission presenting in good clinical conditions 6 months post-CAR-T infusion, without manifestations of graft-versus-host disease. The case report provides insight into the reactivity and persistence of CAR-T cells of umbilical origin, confirming the potential promise of allogeneic umbilical cord-derived CAR-T cells.


Subject(s)
Fetal Blood , Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Humans , Hematopoietic Stem Cell Transplantation/methods , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Fetal Blood/cytology , Fetal Blood/transplantation , Recurrence , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Treatment Outcome , Receptors, Chimeric Antigen , Male , Philadelphia Chromosome , Transplantation, Homologous
7.
Article in English | MEDLINE | ID: mdl-38642731

ABSTRACT

Current treatments for schizophrenia (SCZ) remain largely ineffective in one-third of patients. Recent studies using stem cell therapy show a close relationship between stem cell immunomodulatory function and neuroinflammation in SCZ. To better investigate the efficacy of stem cell therapy for SCZ, human umbilical cord blood mesenchymal stem cells (hUC-MSC) with powerful immunomodulatory effects were administered to rats via the tail vein (once a week for 5 consecutive weeks starting from the weaning period) using a maternal immune activation (MIA) rodent model. Open field, PPI, Western blotting, Q-PCR, and immunofluorescence were used to assess the biological effects of repeated tail vein injections of hUC-MSC in offspring rats following the MIA model of SCZ. The results indicated that offspring of MIA rats exhibited schizophrenia-like (SCZ-like) anxiety behavior, with observed microglial activation triggering neuroinflammation. Furthermore, levels of IBA1, HMGB1, and PSD95 were significantly up-regulated, while SYP was significantly down-regulated. It is suggested that hUCB-MSCs may act through HMGB1, Iba1, PSD95, and related pathway molecules to alleviate neuroinflammation and repair synaptic damage by regulating the activity state of microglia. Consequently, this could improve the abnormal behavior observed in MIA offspring rats.


Subject(s)
Anxiety , Disease Models, Animal , HMGB1 Protein , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Microglia , Rats, Sprague-Dawley , Schizophrenia , Animals , Rats , Schizophrenia/therapy , Schizophrenia/chemically induced , Mesenchymal Stem Cell Transplantation/methods , Humans , Female , Anxiety/therapy , HMGB1 Protein/metabolism , Pregnancy , Disks Large Homolog 4 Protein/metabolism , Calcium-Binding Proteins/metabolism , Microfilament Proteins/metabolism , Male , Fetal Blood/cytology , Neuroinflammatory Diseases , Synaptophysin/metabolism , Cord Blood Stem Cell Transplantation/methods , Prenatal Exposure Delayed Effects
8.
Stem Cells Dev ; 33(9-10): 214-227, 2024 May.
Article in English | MEDLINE | ID: mdl-38445374

ABSTRACT

Cellular therapies provide promising options for inducing tolerance in transplantation of solid organs, bone marrow, and vascularized composite allografts. However, novel tolerance-inducing protocols remain limited, despite extensive research. We previously introduced and characterized a human multi-chimeric cell (HMCC) line, created through ex vivo fusion of human umbilical cord blood (UCB) cells derived from three unrelated donors. In this study, we assessed in vivo biodistribution and safety of HMCCs in the NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ NOD scid gamma (NSG) mouse model. Twenty-four NSG mice were randomly assigned to four groups (n = 6/group) and received intraosseous (IO.) or intravenous (IV.) injections of 0.6 × 106 donor UCB cells or fused HMCC: Group 1-UCB (IO.), Group 2-UCB (IV.), Group 3-HMCC (IO.), and Group 4-HMCC (IV.). Hematopoietic phenotype maintenance and presence of human leukocyte antigens (HLA), class I antigens, in the selected lymphoid and nonlymphoid organs were assessed by flow cytometry. Weekly evaluation and magnetic resonance imaging (MRI) assessed HMCC safety. Comparative analysis of delivery routes revealed significant differences in HLA class I percentages for IO.: 1.83% ± 0.79%, versus IV. delivery: 0.04% ± 0.01%, P < 0.01, and hematopoietic stem cell marker percentages of CD3 (IO.: 1.41% ± 0.04%, vs. IV.: 0.07% ± 0.01%, P < 0.05) and CD4 (IO.: 2.74% ± 0.31%, vs. IV.: 0.59% ± 0.11%, P < 0.01). Biodistribution analysis after IO. delivery confirmed HMCC presence in lymphoid organs and negligible presence in nonlymphoid organs, except for lung (IO.: 0.19% ± 0.06%, vs. IV.: 6.33% ± 0.56%, P < 0.0001). No evidence of tumorigenesis was observed by MRI at 90 days following IO. and IV. administration of HMCC. This study confirmed biodistribution and safety of HMCC therapy in the NSG mouse model, both following IO. and IV. administration. However, IO. delivery route confirmed higher efficacy of engraftment and safety profile, introducing HMCCs as a novel cell-based therapeutic approach with promising clinical applications in solid organ, bone marrow, and vascularized composite allotransplantation transplantation.


Subject(s)
Mice, Inbred NOD , Mice, SCID , Animals , Humans , Mice , Tissue Distribution , Administration, Intravenous , Fetal Blood/cytology , Infusions, Intraosseous/methods
9.
Hum Immunol ; 85(3): 110769, 2024 May.
Article in English | MEDLINE | ID: mdl-38429146

ABSTRACT

Fortunately, ample efforts are being made to find the best strategy to improve the anti-leukemia capacity of NK cells for treating different types of cancer. Despite the favorable ADCC capacity of functional CD16 + NK cells for immunotherapy, when NK cells face leukemia cells, the CD16 receptor is cleaved during the process mediated by a disintegrin and metalloproteinase-17(ADAM17). Reduced CD16 expression on NK cells weakens their cytotoxicity against leukemia cells. In addition, the expression of the CD47 receptor is high in acute lymphoblastic leukemia (ALL) compared to normal cells and can be correlated with poor prognosis. In the present study, ADAM17 was inhibited in cord blood-derived CD16 + NK cells, and their activity against ALL cell lines was evaluated following blockage with anti-CD47 antibody. As the results showed, the CD16 expression was reduced in the NK cells co-cultured with ALL cell lines. However, the ADAM17 inhibition increased the CD16 expression on the NK cells. This enhanced the cytotoxicity of those cells as well as cytokine production was evaluated by measuring expression of CD107-a expression, and IFN-γ production. Moreover, the presence of the ADAM17 inhibitor increased the apoptosis effect of the generated NK cells in response to ALL cells. Therefore, the inhibition of ADAM17 is useful for the activity of CD16 + NK cells against cancer cells.


Subject(s)
ADAM17 Protein , Fetal Blood , Killer Cells, Natural , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Receptors, IgG , Humans , Killer Cells, Natural/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , ADAM17 Protein/metabolism , ADAM17 Protein/antagonists & inhibitors , Receptors, IgG/metabolism , Fetal Blood/cytology , Cell Line, Tumor , Cytotoxicity, Immunologic , GPI-Linked Proteins/metabolism , Coculture Techniques , Apoptosis , Antibody-Dependent Cell Cytotoxicity , Interferon-gamma/metabolism , CD47 Antigen
10.
Cell Tissue Bank ; 25(2): 605-611, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38168846

ABSTRACT

Umbilical cord blood is a rich source of hematopoietic stem cells that has been used for transplantation for over 30 years, especially when there is no compatible hematopoietic stem cell donor available. Its use has decreased more recently, since the development of methods to improve haploidentical transplants has allowed the use of mobilized peripheral blood as a source of hematopoietic stem cells. Public cord blood banks collect, process and store cord blood samples from voluntary donations. In addition, many public banks are involved in research to enhance hematopoietic stem cell therapies and develop new treatments for haematological and genetic diseases. The COVID-19 pandemic, which emerged in 2019, has had a profound and wide-ranging impact on human health and treatment. The area of hematopoietic stem cell transplantation was deeply affected by reductions in bone marrow, peripheral blood and cord blood donations; logistical challenges; exposure of healthcare workers and other challenges. The present study reviews the impact of the COVID-19 pandemic on cord blood banking and transportation around the world with a special focus on Brazil.


Subject(s)
Blood Banks , COVID-19 , Cord Blood Stem Cell Transplantation , Fetal Blood , Pandemics , SARS-CoV-2 , Humans , COVID-19/epidemiology , Fetal Blood/cytology , SARS-CoV-2/isolation & purification , Brazil/epidemiology , Blood Donors
11.
J Thromb Haemost ; 22(5): 1447-1462, 2024 May.
Article in English | MEDLINE | ID: mdl-38160730

ABSTRACT

BACKGROUND: Recent clinical studies have shown that transfusions of adult platelets increase morbidity and mortality in preterm infants. Neonatal platelets are hyporesponsive to agonist stimulation, and emerging evidence suggests developmental differences in platelet immune functions. OBJECTIVES: This study was designed to compare the proteome and phosphoproteome of resting adult and neonatal platelets. METHODS: We isolated resting umbilical cord blood-derived platelets from healthy full-term neonates (n = 8) and resting blood platelets from healthy adults (n = 6) and compared protein and phosphoprotein contents using data-independent acquisition mass spectrometry. RESULTS: We identified 4770 platelet proteins with high confidence across all samples. Adult and neonatal platelets were clustered separately by principal component analysis. Adult platelets were significantly enriched in immunomodulatory proteins, including ß2 microglobulin and CXCL12, whereas neonatal platelets were enriched in ribosomal components and proteins involved in metabolic activities. Adult platelets were enriched in phosphorylated GTPase regulatory enzymes and proteins participating in trafficking, which may help prime them for activation and degranulation. Neonatal platelets were enriched in phosphorylated proteins involved in insulin growth factor signaling. CONCLUSION: Using label-free data-independent acquisition mass spectrometry, our findings expanded the known neonatal platelet proteome and identified important differences in protein content and phosphorylation between neonatal and adult platelets. These developmental differences suggested enhanced immune functions for adult platelets and presence of molecular machinery related to platelet activation. These findings are important to understanding mechanisms underlying key platelet functions as well as the harmful effects of adult platelet transfusions given to preterm infants.


Subject(s)
Blood Platelets , Fetal Blood , Phosphoproteins , Proteomics , Signal Transduction , Humans , Blood Platelets/metabolism , Infant, Newborn , Adult , Fetal Blood/metabolism , Fetal Blood/cytology , Phosphorylation , Proteomics/methods , Phosphoproteins/blood , Proteome , Female , Age Factors , Male , Principal Component Analysis , Mass Spectrometry , Tandem Mass Spectrometry
12.
Neurosci Res ; 203: 42-50, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38154662

ABSTRACT

Numerous evidences showed that human umbilical cord blood (UCB) mononuclear cells were a promising approach for the therapy of ischemic stroke(IS). The effect of stage-specific embryonic antigen 3 (SSEA3)positive subpopulation in UCB was not investigated in IS. In this study, we isolated SSEA3 positive cells from healthy UCB mononuclear cells, which comprised about 7.01% of the total UCB-mononuclear cells. Flow cytometry analysis revealed that SSEA3(+)UCB cells were almost positive for CD44 and CD45, and negative for CD73, CD90 and CD105. The expression of Oct3/4 in SSEA3(+)UCB cells were higher than that in UCB. SSEA3(+)UCB cells sorted by magnetic cell sorting were intravenously injected into the middle cerebral arterial occlusion(MCAO) rat model. Neurological score showed that SSEA3(+)UCB transplantation group exhibited significant improvements in the functional outcome of MCAO rats than UCB transplantation group. Nissl staining and microtubule association protein-2(MAP2) immunofluorescence staining showed that the SSEA3(+)UCB transplantation group decreased neuronal loss. SSEA3(+)UCB transplantation group reduced neuronal apoptosis, inhibited caspase3 expression, and decreased tumor necrosis factor α(TNF-α). These results indicate that SSEA3 positive cells are a novel subpopulation of UCB cells, which exhibit great potential for the treatment of ischemic stroke.


Subject(s)
Disease Models, Animal , Fetal Blood , Ischemic Stroke , Animals , Humans , Ischemic Stroke/therapy , Ischemic Stroke/metabolism , Fetal Blood/cytology , Leukocytes, Mononuclear/transplantation , Leukocytes, Mononuclear/metabolism , Male , Rats, Sprague-Dawley , Rats , Infarction, Middle Cerebral Artery/therapy , Cord Blood Stem Cell Transplantation/methods , Stage-Specific Embryonic Antigens/metabolism , Brain Ischemia/therapy , Apoptosis/physiology
13.
Nature ; 615(7950): 127-133, 2023 03.
Article in English | MEDLINE | ID: mdl-36813966

ABSTRACT

Haematopoietic stem cells (HSCs) are a rare cell type that reconstitute the entire blood and immune systems after transplantation and can be used as a curative cell therapy for a variety of haematological diseases1,2. However, the low number of HSCs in the body makes both biological analyses and clinical application difficult, and the limited extent to which human HSCs can be expanded ex vivo remains a substantial barrier to the wider and safer therapeutic use of HSC transplantation3. Although various reagents have been tested in attempts to stimulate the expansion of human HSCs, cytokines have long been thought to be essential for supporting HSCs ex vivo4. Here we report the establishment of a culture system that allows the long-term ex vivo expansion of human HSCs, achieved through the complete replacement of exogenous cytokines and albumin with chemical agonists and a caprolactam-based polymer. A phosphoinositide 3-kinase activator, in combination with a thrombopoietin-receptor agonist and the pyrimidoindole derivative UM171, were sufficient to stimulate the expansion of umbilical cord blood HSCs that are capable of serial engraftment in xenotransplantation assays. Ex vivo HSC expansion was further supported by split-clone transplantation assays and single-cell RNA-sequencing analysis. Our chemically defined expansion culture system will help to advance clinical HSC therapies.


Subject(s)
Cell Culture Techniques , Cell Proliferation , Cytokines , Hematopoietic Stem Cells , Humans , Cell Proliferation/drug effects , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Fetal Blood/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cell Culture Techniques/methods , Albumins , Caprolactam , Polymers , Receptors, Thrombopoietin , Transplantation, Heterologous , Single-Cell Gene Expression Analysis
14.
Clin Transl Med ; 13(1): e1175, 2023 01.
Article in English | MEDLINE | ID: mdl-36683248

ABSTRACT

BACKGROUND: Hematopoietic stem cells (HSCs) from different sources show varied repopulating capacity, and HSCs lose their stemness after long-time ex vivo culture. A deep understanding of these phenomena may provide helpful insights for HSCs. METHODS: Here, we applied single-cell RNA-seq (scRNA-seq) to analyse the naïve and stimulated human CD34+ cells from cord blood (CB) and mobilised peripheral blood (mPB). RESULTS: We collected over 16 000 high-quality single-cell data to construct a comprehensive inference map and characterised the HSCs under a quiescent state on the hierarchy top. Then, we compared HSCs in CB with those in mPB and HSCs of naïve samples to those of cultured samples, and identified stemness-related genes (SRGs) associated with cell source (CS-SRGs) and culture time (CT-SRGs), respectively. Interestingly, CS-SRGs and CT-SRGs share genes enriched in the signalling pathways such as mRNA catabolic process, translational initiation, ribonucleoprotein complex biogenesis and cotranslational protein targeting to membrane, suggesting dynamic protein translation and processing may be a common requirement for stemness maintenance. Meanwhile, CT-SRGs are enriched in pathways involved in glucocorticoid and corticosteroid response that affect HSCs homing and engraftment. In contrast, CS-SRGs specifically contain genes related to purine and ATP metabolic process, which is crucial for HSC homeostasis in the stress settings. Particularly, when CT-SRGs are used as reference genes for the construction of the development trajectory of CD34+ cells, lymphoid and myeloid lineages are clearly separated after HSCs/MPPs. Finally, we presented an application through a small-scale drug screening using Connectivity Map (CMap) against CT-SRGs. A small molecule, cucurbitacin I, was found to efficiently expand HSCs ex vivo while maintaining its stemness. CONCLUSIONS: Our findings provide new perspectives for understanding HSCs, and the strategy to identify candidate molecules through SRGs may be applicable to study other stem cells.


Subject(s)
Cell Differentiation , Fetal Blood , Hematopoietic Stem Cells , Humans , Antigens, CD34/analysis , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Single-Cell Analysis , Gene Expression Profiling , Cell Differentiation/genetics
15.
Am J Reprod Immunol ; 88(1): e13555, 2022 07.
Article in English | MEDLINE | ID: mdl-35452164

ABSTRACT

PROBLEM: Although pregnant women with gestational diabetes (GD), morbidly adherent placenta (MAP), and pregnancy hypertension (pHT) diseases lead to intrauterine growth restriction (IUGR), little is known about their effect on mucosal-associated invariant T (MAIT) and innate lymphoid cells (ILC) in the umbilical cord. This study aimed to quantify and characterize MAIT cells and ILCs in the cord blood of pregnant women with GD, MAP, and pHT diseases. METHOD OF STUDY: Cord blood mononuclear cells (CBMCs) were isolated by Ficoll-Paque gradient. CD3+ TCRVα7.2+ CD161high cells and ILC subsets were quantified by flow cytometry. CBMCs were stimulated with PMA/Ionomycin and Golgi Plug for 4 h and stained for IFN-γ, TNF-α, and granzyme B. The stained cells were analyzed on FACS ARIA III. RESULTS: Compared with healthy pregnancies, in the cord blood of the pHT group, elevated number of lymphocytes was observed. Moreover, the absolute number of IFN-γ producing CD4+ or CD4- subsets of CD3+ TCRVα7.2+ CD161high cells as well as those producing granzyme B were significantly elevated in the pHT group compared to healthy controls suggesting increased MAIT cell activity in the pHT cord blood. Similarly, in the MAP group, the absolute number of total CD3+ TCRVα7.2+ CD161high cells, but not individual CD4+ or negative subsets, were significantly increased compared with healthy controls' cord blood. Absolute numbers of total CD3+ TCRVα7.2+ CD161high cells and their subsets were comparable in the cord blood of the GD group compared with healthy controls. Finally, the absolute number of total ILCs and ILC3 subset were significantly elevated in only pHT cord blood compared with healthy controls. Our data also reveal that IFN-γ+ or granzyme B+ cell numbers negatively correlated with fetal birth weight. CONCLUSIONS: CD3+ TCRVα7.2+ CD161high cells and ILCs show unique expansion and activity in the cord blood of pregnant women with distinct diseases causing IUGR and may play roles in fetal growth restriction.


Subject(s)
Diabetes, Gestational , Hypertension, Pregnancy-Induced , Placenta Accreta , T-Lymphocyte Subsets , Diabetes, Gestational/immunology , Female , Fetal Blood/cytology , Fetal Blood/immunology , Granzymes , Humans , Hypertension, Pregnancy-Induced/immunology , Immunity, Innate , Lymphocytes , Placenta/pathology , Placenta Accreta/immunology , Pregnancy , T-Lymphocyte Subsets/cytology
16.
Hematology ; 27(1): 263-273, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35192776

ABSTRACT

BACKGROUND: The reactivation of fetal γ-globin expression is an effective strategy for ameliorating the clinical symptoms of ß-hemoglobinopathies. However, the mechanism of globin switching, especially the roles of long non-coding RNAs (lncRNAs) in this process, remains elusive. METHODS: We compared the in vivo transcriptome profiles of nucleated red blood cells (NRBCs) isolated from the umbilical cord blood of preterm and full-term newborns. We collected 75 umbilical cord blood samples and performed qPCR of the candidate genes. RESULTS: In this study, we identified 7,166 differentially expressed protein-coding genes, 3,243 differentially expressed lncRNAs, and 79 differentially expressed microRNAs. Our data show that the Fanconi anemia pathway and the H19/let-7/LIN28B axis may be involved in γ- to ß-globin gene switching. Moreover, we constructed the hub gene network of the differentially expressed transcription factors. Based on qPCR, we found that BCL11A was differentially expressed based on biological sex. We also confirmed that H19 is differentially expressed and established the H19-related network to reveal the potential regulatory mechanisms. CONCLUSION: We present the profiles of the in vivo transcriptome differences of NRBCs between preterm and full-term neonates for the first time, and provide novel research targets for ß-hemoglobinopathies.


Subject(s)
Erythrocytes/metabolism , Fetal Blood/metabolism , Transcriptome/immunology , Female , Fetal Blood/cytology , Humans , Infant, Newborn , Infant, Premature , Pregnancy
17.
PLoS One ; 17(1): e0262854, 2022.
Article in English | MEDLINE | ID: mdl-35077481

ABSTRACT

Chondroitin sulfate (CS) and its isomeric variant, dermatan sulfate (DS), are complex glycosaminoglycans (GAGs) which are ubiquitous components of the extracellular matrix in various tissues including the brain. CS and/or DS are known to bind to a variety of growth factors and regulate many cellular events such as proliferation and differentiation. Although the biological activities of CS and/or DS towards neural stem/progenitor cells (NSPCs) have been well investigated, the CS and/or DS of hematopoietic stem cells (HSCs) have not been fully characterized. Here, we analyzed GAGs on mononuclear cells of rat umbilical cord blood cells (UCB-MNCs). CS was detected in vascular intima and media of rat umbilical cord at embryonic day 19 (E19) by immunohistochemistry. The stem-cell-enriched-UCBCs (SCE-UCBCs), which were expanded from rat UCB-MNCs, expressed CS. CS chains are composed of repeating disaccharide units, which are classified into several types such as O-, A-, B-, C-, D-, and E-unit according to the number and positions of sulfation. A disaccharide composition analysis revealed that CS and/or DS were abundant in rat UCB-MNCs as well as in their expanded SCE-UCBCs, while the amount of heparan sulfate (HS) was less. The degree of sulfation of CS/DS was relatively low and the major component in UCB-MNCs and SCE-UCBCs was the A-unit. A colony-forming cell assay revealed that the percentage of colony-forming cells decreased in culture with CS degradation enzyme. The CS and/or DS of UCBCs may be involved in biological activities such as stem cell proliferation and/or differentiation.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondroitin Sulfates/pharmacology , Fetal Blood/metabolism , Stem Cells/metabolism , Animals , Cell Culture Techniques , Cells, Cultured , Chondroitin Sulfates/chemistry , Disaccharides/chemistry , Disaccharides/pharmacology , Female , Fetal Blood/cytology , Rats , Stem Cells/cytology
18.
Sci Rep ; 12(1): 412, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013490

ABSTRACT

The endometrium plays a critical role in embryo implantation and pregnancy, and a thin uterus is recognized as a key factor in embryo implantation failure. Umbilical cord mesenchymal stem cells (UC-MSCs) have attracted interest for the repair of intrauterine adhesions. The current study investigated the repair of thin endometrium in rats using the UC-MSCs and the mechanisms involved. Rats were injected with 95% ethanol to establish a model of thin endometrium. The rats were randomly divided into normal, sham, model, and UC-MSCs groups. Endometrial morphological alterations were observed by hematoxylin-eosin staining and Masson staining, and functional restoration was assessed by testing embryo implantation. The interaction between UC-MSCs and rat endometrial stromal cells (ESCs) was evaluated using a transwell 3D model and immunocytochemistry. Microarray mRNA and miRNA platforms were used for miRNA-mRNA expression profiling. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) analyses were performed to identify the biological processes, molecular functions, cellular components, and pathways of endometrial injury and UC-MSCs transplantation repair and real-time quantitative reverse transcription PCR (qRT-PCR) was performed to further identify the expression changes of key molecules in the pathways. Endometrium thickness, number of glands, and the embryo implantation numbers were improved, and the degree of fibrosis was significantly alleviated by UC-MSCs treatment in the rat model of thin endometrium. In vitro cell experiments showed that UC-MSCs migrated to injured ESCs and enhanced their proliferation. miRNA microarray chip results showed that expression of 45 miRNAs was downregulated in the injured endometrium and upregulated after UC-MSCs transplantation. Likewise, expression of 39 miRNAs was upregulated in the injured endometrium and downregulated after UC-MSCs transplantation. The miRNA-mRNA interactions showed the changes in the miRNA and mRNA network during the processes of endometrial injury and repair. GO and KEGG analyses showed that the process of endometrial injury was mainly attributed to the decomposition of the extracellular matrix (ECM), protein degradation and absorption, and accompanying inflammation. The process of UC-MSCs transplantation and repair were accompanied by the reconstruction of the ECM, regulation of chemokines and inflammation, and cell proliferation and apoptosis. The key molecules involved in ECM-receptor interaction pathways were further verified by qRT-PCR. Itga1 and Thbs expression decreased in the model group and increased by UC-MSCs transplantation, while Laminin and Collagen expression increased in both the model group and MSCs group, with greater expression observed in the latter. This study showed that UC-MSCs transplantation could promote recovery of thin endometrial morphology and function. Furthermore, it revealed the expression changes of miRNA and mRNA after endometrial injury and UC-MSCs transplantation repair processed, and signaling pathways that may be involved in endometrial injury and repair.


Subject(s)
Cell Proliferation , Cord Blood Stem Cell Transplantation , Endometrium/pathology , Extracellular Matrix/pathology , Regeneration , Uterine Diseases/surgery , Animals , Cell Communication , Cell Culture Techniques, Three Dimensional , Cells, Cultured , Disease Models, Animal , Endometrium/metabolism , Endometrium/physiopathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Fetal Blood/cytology , Gene Expression Regulation , Gene Regulatory Networks , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Rats, Sprague-Dawley , Signal Transduction , Transcriptome , Uterine Diseases/metabolism , Uterine Diseases/pathology , Uterine Diseases/physiopathology
19.
Mol Biol Rep ; 49(2): 931-941, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34741711

ABSTRACT

BACKGROUND: Hematopoietic stem cell (HSC) transplantation is considered a possible treatment option capable of curing various diseases. The aim of this study was the co-culturing of mesenchymal stem cell (MSC) spheres with HSCs under hypoxic condition to enhance the proliferation, self-renewal, stemness, and homing capacities of HSCs. METHODS AND RESULTS: HSCs were expanded after being subjected to different conditions including cytokines without feeder (Cyto), co-culturing with adherent MSCs (MSC), co-culturing with adherent MSCs + hypoxia (MSC + Hyp), co-culturing with MSCs spheres (Sph-MSC), co-culturing with MSCs spheres + hypoxia (Sph-MSC + Hyp), co-culturing with MSC spheres + cytokines (Sph-MSC + Cyto). After 10 days, total nucleated cell (TNC) and CD34+/CD38- cell counts, colony-forming unit assay (CFU), long-term culture initiating cell (LTC-IC), the expression of endothelial protein C receptor (EPCR), nucleostemin (NS), nuclear factor I/X (Nfix) CXCR4, and VLA-4 were evaluated. The TNC, CD34+/CD38- cell count, CFU, and LTC-IC were higher in the Sph-MSC + Hyp and Sph-MSC + Cyto groups as compared with those of the MSC + Hyp group (P < 0.001). The expanded HSCs co-cultured with MSC spheres in combination with hypoxia expressed more EPCR, CXCR4, VLA-4, NS, and Nfix mRNA. The protein expression was also more up-regulated in the Sph-MSC + Cyto and Sph-MSC + Hyp groups. CONCLUSION: Co-culturing HSCs with MSC spheres under hypoxic condition not only leads to higher cellular yield but also increases the expression of self-renewal and homing genes. Therefore, we suggest this approach as a simple and non-expensive strategy that might improve the transplantation efficiency of HSCs.


Subject(s)
Coculture Techniques/methods , Hematopoietic Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Antigens, CD34/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Hypoxia/physiology , Cell Proliferation , Cells, Cultured , Coculture Techniques/economics , Cost-Benefit Analysis , Cytokines/metabolism , Fetal Blood/cytology , Humans , Receptors, CXCR4
20.
Gene ; 809: 146005, 2022 Jan 30.
Article in English | MEDLINE | ID: mdl-34673210

ABSTRACT

Stem cells from umbilical cord blood (UCB) are able to proliferate and differentiate into various somatic cell types. Thereby, they are considered as one of the attractive stem cell sources in tissue engineering and regenerative medicine. However, the limited number of hematopoietic CD 133+ stem cells in UCB restricted the clinical application of such stem cells. This study was aimed to expand CD 133+ stem cells derived from UCB on a 3D silk scaffold. UCB133+ stem cells were extracted using Magnetic cell sorting (MACS) and characterized by flow cytometry. Isolated cells were seeded on a fabricated electrospun silk scaffold and cultured for 7 days. The real-time PCR, cell counting, colony-forming assay, and MTT assay were performed to evaluate the expansion and homing of stem cells. The results showed a higher expression of CXCR4 gene, the number of cultured stem cells, and colony-forming units in the 3D silk scaffold group after 7 days when compared to the tissue culture plate. Moreover, higher viability and proliferation of stem cells were seen in cells cultured on silk scaffold. It seems electrospun silk scaffold could be used as a suitable substrate for UCB CD 133+ stem cell expansion.


Subject(s)
AC133 Antigen/metabolism , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Nanofibers , Cell Culture Techniques/methods , Cell Proliferation , Cell Separation/methods , Cells, Cultured , Colony-Forming Units Assay , Hematopoietic Stem Cells/metabolism , Humans , Microscopy, Electron, Scanning , Nanofibers/chemistry , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Silk/chemistry , Tissue Scaffolds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...