Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Article in English | MEDLINE | ID: mdl-38564376

ABSTRACT

RATIONALE: The chronic lung disease bronchopulmonary dysplasia (BPD) is the most severe complication of extreme prematurity. BPD results in impaired lung alveolar and vascular development and long-term respiratory morbidity, for which only supportive therapies exist. Umbilical cord-derived mesenchymal stromal cells (UC-MSCs) improve lung structure and function in experimental BPD. Results of clinical trials with MSCs for many disorders do not yet match the promising preclinical studies. A lack of specific criteria to define functionally distinct MSCs persists. OBJECTIVES: To determine and correlate single-cell UC-MSC transcriptomic profile with therapeutic potential. METHODS: UC-MSCs from five term donors and human neonatal dermal fibroblasts (HNDFs, control cells of mesenchymal origin) transcriptomes were investigated by single-cell RNA sequencing analysis (scRNA-seq). The lung-protective effect of UC-MSCs with a distinct transcriptome and control HNDFs was tested in vivo in hyperoxia-induced neonatal lung injury in rats. MEASUREMENTS AND MAIN RESULTS: UC-MSCs showed limited transcriptomic heterogeneity, but were different from HNDFs. Gene ontology enrichment analysis revealed distinct - progenitor-like and fibroblast-like - UC-MSC subpopulations. Only the treatment with progenitor-like UC-MSCs improved lung function and structure and attenuated pulmonary hypertension in hyperoxia-exposed rat pups. Moreover, scRNA-seq identified major histocompatibility complex class I as a molecular marker of non-therapeutic cells and associated with decreased lung retention. CONCLUSIONS: UC-MSCs with a progenitor-like transcriptome, but not with a fibroblast-like transcriptome, provide lung protection in experimental BPD. High expression of major histocompatibility complex class I is associated with reduced therapeutic benefit. scRNA-seq may be useful to identify subsets of MSCs with superior repair capacity for clinical application.

2.
Heliyon ; 9(8): e18741, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37554783

ABSTRACT

Polycystic ovarian syndrome (PCOS) is one of the commonest endocrinopathies in childbearing women. The research was conducted to assess the impact of Irpex lacteus polysaccharide (ILP, 1000 mg/kg) on the letrozole (1 mg/kg)-induced PCOS model in female rats. Metformin (Met, 265 mg/kg) as the positive control. The study suggested that ILP restored the estrous cycle in rats with PCOS as well as lowered relative ovarian weight and body weight, in comparison to normal. Rats with PCOS showed improvement in ovarian structure and fibrosis when given ILP. ILP decreased the testosterone (T), low-density lipoprotein cholesterol (LDL-C), triglyceride (TG), total cholesterol (TC), luteinizing hormone (LH), homeostasis model assessment-insulin resistance (HOMA-IR), fasting blood glucose (FBG), and insulin (INS) levels and elevated the follicle-stimulating hormone (FSH) and estrogen (E2) levels in PCOS rats. In addition, ILP increased the content of superoxide dismutase (SOD) in serum and the antioxidant enzymes (Prdx3, Sod1, Gsr, Gsta4, Mgst1, Gpx3, Sod2 and Cat) expression levels in the ovaries and decreased the serum expression of malondialdehyde (MDA). In addition, ILP treatment slowed down the process of the fibrosis-associated TGF-ß1/Smad pathway and downregulated α-smooth muscle actin (α-SMA) and connective tissue growth factor (CTGF) levels in PCOS rats ovaries. According to these findings, ILP may be able to treat letrozole-induced PCOS in rats by ameliorating metabolic disturbances, sex hormone levels, oxidative stress, and ovarian fibrosis.

3.
J Clin Invest ; 132(22)2022 11 15.
Article in English | MEDLINE | ID: mdl-36136598

ABSTRACT

Preterm birth is the leading cause of death in children under 5 years of age. Premature infants who receive life-saving oxygen therapy often develop bronchopulmonary dysplasia (BPD), a chronic lung disease. Infants with BPD are at a high risk of abnormal neurodevelopment, including motor and cognitive difficulties. While neural progenitor cells (NPCs) are crucial for proper brain development, it is unclear whether they play a role in BPD-associated neurodevelopmental deficits. Here, we show that hyperoxia-induced experimental BPD in newborn mice led to lifelong impairments in cerebrovascular structure and function as well as impairments in NPC self-renewal and neurogenesis. A neurosphere assay utilizing nonhuman primate preterm baboon NPCs confirmed impairment in NPC function. Moreover, gene expression profiling revealed that genes involved in cell proliferation, angiogenesis, vascular autoregulation, neuronal formation, and neurotransmission were dysregulated following neonatal hyperoxia. These impairments were associated with motor and cognitive decline in aging hyperoxia-exposed mice, reminiscent of deficits observed in patients with BPD. Together, our findings establish a relationship between BPD and abnormal neurodevelopmental outcomes and identify molecular and cellular players of neonatal brain injury that persist throughout adulthood that may be targeted for early intervention to aid this vulnerable patient population.


Subject(s)
Bronchopulmonary Dysplasia , Cognitive Dysfunction , Hyperoxia , Premature Birth , Infant, Newborn , Female , Mice , Humans , Animals , Hyperoxia/complications , Hyperoxia/metabolism , Animals, Newborn , Bronchopulmonary Dysplasia/genetics , Neurogenesis , Cognitive Dysfunction/etiology , Cognition , Lung/metabolism
4.
Stem Cells ; 40(5): 479-492, 2022 05 27.
Article in English | MEDLINE | ID: mdl-35445270

ABSTRACT

Late lung development is a period of alveolar and microvascular formation, which is pivotal in ensuring sufficient and effective gas exchange. Defects in late lung development manifest in premature infants as a chronic lung disease named bronchopulmonary dysplasia (BPD). Numerous studies demonstrated the therapeutic properties of exogenous bone marrow and umbilical cord-derived mesenchymal stromal cells (MSCs) in experimental BPD. However, very little is known regarding the regenerative capacity of resident lung MSCs (L-MSCs) during normal development and in BPD. In this study we aimed to characterize the L-MSC population in homeostasis and upon injury. We used single-cell RNA sequencing (scRNA-seq) to profile in situ Ly6a+ L-MSCs in the lungs of normal and O2-exposed neonatal mice (a well-established model to mimic BPD) at 3 developmental timepoints (postnatal days 3, 7, and 14). Hyperoxia exposure increased the number and altered the expression profile of L-MSCs, particularly by increasing the expression of multiple pro-inflammatory, pro-fibrotic, and anti-angiogenic genes. In order to identify potential changes induced in the L-MSCs transcriptome by storage and culture, we profiled 15 000 Ly6a+ L-MSCs after in vitro culture. We observed great differences in expression profiles of in situ and cultured L-MSCs, particularly those derived from healthy lungs. Additionally, we have identified the location of Ly6a+/Col14a1+ L-MSCs in the developing lung and propose Serpinf1 as a novel, culture-stable marker of L-MSCs. Finally, cell communication analysis suggests inflammatory signals from immune and endothelial cells as main drivers of hyperoxia-induced changes in L-MSCs transcriptome.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Mesenchymal Stem Cells , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/therapy , Endothelial Cells , Humans , Hyperoxia/genetics , Hyperoxia/metabolism , Infant, Newborn , Lung/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Sequence Analysis, RNA
5.
J Food Biochem ; 46(11): e14203, 2022 11.
Article in English | MEDLINE | ID: mdl-35470867

ABSTRACT

The current study investigated the effects of sinapic acid on high-fat diet (HFD)-induced lipid metabolism and oxidative stress in male Syrian hamsters. Sinapic acid treatment significantly reduced body weight, epididymal fat, and perirenal fat mass in HFD hamsters. Sinapic acid also improved dyslipidemia levels (reducing the serum levels of total cholesterol, triglycerides, and low-density lipoprotein cholesterol, and increasing the high-density lipoprotein cholesterol) and increased T-AOC levels to mitigate oxidative stress injury. Moreover, sinapic acid intervention increased the activations of PPAR-γ, CPT-1, and CYP7A1 and decreased the activations of FAS, ACC1, SREBP1, SREBP2, and HMGCR in the livers of HFD hamsters. In addition, sinapic acid intervention also significantly inhibited the intestinal mRNA levels of Srebp2 and Npc1l1 in HFD hamsters. In conclusion, sinapic acid can significantly attenuate abnormal lipid metabolism in the development of HFD-induced obesity and reduce the level of oxidative stress to exert its anti-obesity effect. PRACTICAL APPLICATIONS: Obesity is the main cause of some chronic metabolic syndromes, such as dyslipidemia, nonalcoholic fatty liver disease, diabetes, and hyperuricemia. Searching for new, safe, and effective natural products in weight loss and fat reduction has become one of the hot research topics. As a natural source of simple phenolic acids, sinapic acid is present in fruits, vegetables, and grains and has been indicated to have anti-inflammatory, antioxidant, antihyperuricemic, lipid homeostasis regulation, and anticancer activities. However, the lipid metabolism- and oxidative stress-regulating activities of sinapic acid are not clear. Here, the current study investigated the lipid metabolism and oxidative stress regulating activities of sinapic acid in male Syrian hamsters fed a high-fat diet.


Subject(s)
Diet, High-Fat , Dyslipidemias , Cricetinae , Animals , Male , Diet, High-Fat/adverse effects , Lipid Metabolism , Mesocricetus , Cholesterol , Oxidative Stress , Obesity , Dyslipidemias/drug therapy , Dyslipidemias/etiology
6.
Am J Respir Crit Care Med ; 205(10): 1186-1201, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35286238

ABSTRACT

Rationale: Bronchopulmonary dysplasia, a chronic respiratory condition originating from preterm birth, is associated with abnormal neurodevelopment. Currently, there is an absence of effective therapies for bronchopulmonary dysplasia and its associated brain injury. In preclinical trials, mesenchymal stromal cell therapies demonstrate promise as a therapeutic alternative for bronchopulmonary dysplasia. Objectives: To investigate whether a multifactorial neonatal mouse model of lung injury perturbs neural progenitor cell function and to assess the ability of human umbilical cord-derived mesenchymal stromal cell extracellular vesicles to mitigate pulmonary and neurologic injury. Methods: Mice at Postnatal Day 7 or 8 were injected intraperitoneally with LPS and ventilated with 40% oxygen at Postnatal Day 9 or 10 for 8 hours. Treated animals received umbilical cord-mesenchymal stromal cell-derived extracellular vesicles intratracheally preceding ventilation. Lung morphology, vascularity, and inflammation were quantified. Neural progenitor cells were isolated from the subventricular zone and hippocampus and assessed for self-renewal, in vitro differentiation ability, and transcriptional profiles. Measurements and Main Results: The multifactorial lung injury model produced alveolar and vascular rarefaction mimicking bronchopulmonary dysplasia. Neural progenitor cells from lung injury mice showed reduced neurosphere and oligodendrocyte formation, as well as inflammatory transcriptional signatures. Mice treated with mesenchymal stromal cell extracellular vesicles showed significant improvement in lung architecture, vessel formation, and inflammatory modulation. In addition, we observed significantly increased in vitro neurosphere formation and altered neural progenitor cell transcriptional signatures. Conclusions: Our multifactorial lung injury model impairs neural progenitor cell function. Observed pulmonary and neurologic alterations are mitigated by intratracheal treatment with mesenchymal stromal cell-derived extracellular vesicles.


Subject(s)
Bronchopulmonary Dysplasia , Extracellular Vesicles , Lung Injury , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Premature Birth , Animals , Bronchopulmonary Dysplasia/therapy , Female , Humans , Infant, Newborn , Lung , Lung Injury/therapy , Mice , Pregnancy
8.
Physiol Rep ; 6(22): e13922, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30485704

ABSTRACT

Very preterm birth is associated with increased cardiovascular diseases and changes in myocardial structure. The current study aimed to investigate the impact of endothelial colony-forming cell (ECFC) treatment on heart morphological changes in the experimental model of neonatal high oxygen (O2 )-induced cardiomyopathy, mimicking prematurity-related conditions. Sprague-Dawley rat pups exposed to 95% O2 or room air (RA) from day 4 (P4) to day 14 (P14) were randomized to receive (jugular vein) exogenous human cord blood ECFC or vehicle at P14 (n = 5 RA-vehicle, n = 8 RA-ECFC, n = 8 O2 -vehicle and n = 7 O2 -ECFC) and the hearts collected at P28. Body and heart weights and heart to body weight ratio did not differ between groups. ECFC treatment prevented the increase in cardiomyocyte surface area in both the left (LV) and right (RV) ventricles of the O2 group (O2 -ECFC vs. O2 -vehicle LV: 121 ± 13 vs. 179 ± 21 µm2 , RV: 118 ± 12 vs. 169 ± 21 µm2 ). In O2 rats, ECFC treatment was also associated with a significant reduction in interstitial fibrosis in both ventricles (O2 -ECFC vs. O2 -vehicle LV: 1.07 ± 0.47 vs. 1.68 ± 0.41% of surface area, RV: 1.01 ± 0.74 vs. 1.77 ± 0.67%) and in perivascular fibrosis in the LV (2.29 ± 0.47 vs. 3.85 ± 1.23%) but in not the RV (1.95 ± 0.95 vs. 2.74 ± 1.14), and with increased expression of angiogenesis marker CD31. ECFC treatment had no effect on cardiomyocyte surface area or on tissue fibrosis of RA rats. Human cord blood ECFC treatment prevented cardiomyocyte hypertrophy and myocardial and perivascular fibrosis observed after neonatal high O2 exposure. ECFC could constitute a new regenerative therapy against cardiac sequelae caused by deleterious conditions of prematurity.


Subject(s)
Cardiomyopathies/therapy , Endothelial Cells/transplantation , Endothelial Progenitor Cells/transplantation , Oxygen/toxicity , Stem Cell Transplantation/methods , Animals , Animals, Newborn , Cardiomyopathies/etiology , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Progenitor Cells/metabolism , Humans , Male , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rats , Rats, Sprague-Dawley , Regeneration
9.
Stem Cells Dev ; 27(16): 1109-1124, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29957134

ABSTRACT

Bronchopulmonary dysplasia (BPD), the most common complication of extreme preterm birth, can be caused by oxygen-related lung injury and is characterized by impaired alveolar and vascular development. Mesenchymal stromal cells (MSCs) have lung protective effects. Conversely, BPD is associated with increased MSCs in tracheal aspirates. We hypothesized that endogenous lung (L-)MSCs are perturbed in a well-established oxygen-induced rat model mimicking BPD features. Rat pups were exposed to 21% or 95% oxygen from birth to postnatal day 10. On day 12, CD146+ L-MSCs were isolated and characterized according to the International Society for Cellular Therapy criteria. Epithelial and vascular repair potential were tested by scratch assay and endothelial network formation, respectively, immune function by mixed lymphocyte reaction assay. Microarray analysis was performed using the Affymetrix GeneChip and gene set enrichment analysis software. CD146+ L-MSCs isolated from rat pups exposed to hyperoxia had decreased CD73 expression and inhibited lung endothelial network formation. CD146+ L-MSCs indiscriminately promoted epithelial wound healing and limited T cell proliferation. Expression of potent antiangiogenic genes of the axonal guidance cue and CDC42 pathways was increased after in vivo hyperoxia, whereas genes of the anti-inflammatory Janus kinase (JAK)/signal transducer and activator of transcription (STAT) and lung/vascular growth-promoting fibroblast growth factor (FGF) pathways were decreased. In conclusion, in vivo hyperoxia exposure alters the proangiogenic effects and FGF expression of L-MSCs. In addition, decreased CD73 and JAK/STAT expression suggests decreased immune function. L-MSC function may be perturbed and contribute to BPD pathogenesis. These findings may lead to improvements in manufacturing exogenous MSCs with superior repair capabilities.


Subject(s)
Bronchopulmonary Dysplasia/metabolism , Lung Injury/metabolism , Mesenchymal Stem Cells/metabolism , Oxygen/adverse effects , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/physiopathology , CD146 Antigen/genetics , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gene Expression Regulation, Developmental/drug effects , Humans , Lung/metabolism , Lung/pathology , Lung Injury/chemically induced , Lung Injury/pathology , Mesenchymal Stem Cells/pathology , Oxygen/administration & dosage , Rats , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
10.
Cytotherapy ; 20(1): 108-125, 2018 01.
Article in English | MEDLINE | ID: mdl-29056548

ABSTRACT

BACKGROUND AIMS: Bronchopulmonary dysplasia (BPD), a chronic lung disease characterized by disrupted lung growth, is the most common complication in extreme premature infants. BPD leads to persistent pulmonary disease later in life. Alveolar epithelial type 2 cells (AEC2s), a subset of which represent distal lung progenitor cells (LPCs), promote normal lung growth and repair. AEC2 depletion may contribute to persistent lung injury in BPD. We hypothesized that induced pluripotent stem cell (iPSC)-derived AECs prevent lung damage in experimental oxygen-induced BPD. METHODS: Mouse AECs (mAECs), miPSCs/mouse embryonic stem sells, human umbilical cord mesenchymal stromal cells (hUCMSCs), human (h)iPSCs, hiPSC-derived LPCs and hiPSC-derived AECs were delivered intratracheally to hyperoxia-exposed newborn mice. Cells were pre-labeled with a red fluorescent dye for in vivo tracking. RESULTS: Airway delivery of primary mAECs and undifferentiated murine pluripotent cells prevented hyperoxia-induced impairment in lung function and alveolar growth in neonatal mice. Similar to hUCMSC therapy, undifferentiated hiPSCs also preserved lung function and alveolar growth in hyperoxia-exposed neonatal NOD/SCID mice. Long-term assessment of hiPSC administration revealed local teratoma formation and cellular infiltration in various organs. To develop a clinically relevant cell therapy, we used a highly efficient method to differentiate hiPSCs into a homogenous population of AEC2s. Airway delivery of hiPSC-derived AEC2s and hiPSC-derived LPCs, improved lung function and structure and resulted in long-term engraftment without evidence of tumor formation. CONCLUSIONS: hiPSC-derived AEC2 therapy appears effective and safe in this model and warrants further exploration as a therapeutic option for BPD and other lung diseases characterized by AEC injury.


Subject(s)
Alveolar Epithelial Cells/cytology , Hyperoxia/complications , Induced Pluripotent Stem Cells/cytology , Lung Injury/etiology , Lung Injury/therapy , Animals , Animals, Newborn , Cell Differentiation , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/ultrastructure , Lung Injury/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Oxygen , Teratoma/pathology
11.
Stem Cells Dev ; 26(14): 1054-1064, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28401804

ABSTRACT

Sepsis is the main cause of morbidity and mortality in neonates. Mesenchymal stromal cells (MSCs) are potent immune-modulatory cells. Their effect in neonatal sepsis has never been explored. We hypothesized that human umbilical cord-derived MSCs (hUC-MSCs) improve survival in experimental neonatal sepsis. Sepsis was induced in 3-day-old rats by intravenous injection of Escherichia coli (5 × 105/rat). One hour after infection, rats were treated intravenously with normal saline, hUC-MSCs, or with interferon-γ preconditioned hUC-MSCs (107 cells/kg). Eighteen hours after infection, survival, bacterial counts, lung neutrophil and macrophage influx, phagocytosis and apoptosis of splenocytes plasma, and LL-37 concentration were evaluated. Animals were observed for survival for 72 h after E. coli injection. Treatment with either hUC-MSCs or preconditioned hUC-MSCs significantly increased survival (hUC-MSCs, 81%; preconditioned hUC-MSCs, 89%; saline, 51%; P < 0.05). Both hUC-MSCs and preconditioned hUC-MSCs enhanced bacterial clearance. Lung neutrophil influx was decreased with preconditioned hUC-MSCs. The number of activated macrophages (CD206+) in the spleen was increased with hUC-MSCs and preconditioned hUC-MSCs; preconditioned hUC-MSCs increased the phagocytic activity of CD206+ macrophages. hUC-MSCs and preconditioned hUC-MSCs decreased splenocyte apoptosis in E. coli infected rats. Finally, LL-37 plasma levels were elevated in neonatal rats treated with hUC-MSCs or preconditioned hUC-MSCs. hUC-MSCs enhance survival and bacterial clearance in experimental neonatal sepsis. hUC-MSCs may be an effective adjunct therapy to reduce neonatal sepsis-related morbidity and mortality.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Neonatal Sepsis/microbiology , Neonatal Sepsis/therapy , Umbilical Cord/cytology , Animals , Antimicrobial Cationic Peptides , Cathelicidins/blood , Escherichia coli/physiology , Humans , Inflammation/pathology , Lung/pathology , Macrophages/metabolism , Neonatal Sepsis/blood , Neutrophils/metabolism , Phagocytosis , Rats , Spleen/pathology , Survival Analysis
12.
Stem Cells Transl Med ; 5(3): 291-300, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26819255

ABSTRACT

Alterations in the development of the placental vasculature can lead to pregnancy complications, such as preeclampsia. Currently, the cause of preeclampsia is unknown, and there are no specific prevention or treatment strategies. Further insight into the placental vasculature may aid in identifying causal factors. Endothelial colony-forming cells (ECFCs) are a subset of endothelial progenitor cells capable of self-renewal and de novo vessel formation in vitro. We hypothesized that ECFCs exist in the micro- and macrovasculature of the normal, term human placenta. Human placentas were collected from term pregnancies delivered by cesarean section (n = 16). Placental micro- and macrovasculature was collected from the maternal and fetal side of the placenta, respectively, and ECFCs were isolated and characterized. ECFCs were CD31(+), CD105(+), CD144(+), CD146(+), CD14(-), and CD45(-), took up 1,1'-dioctadecyl-3,3,3',3'-tetramethyl-indocarbocyanine perchlorate-labeled acetylated low-density lipoprotein, and bound Ulex europaeus agglutinin 1. In vitro, macrovascular ECFCs had a greater potential to generate high-proliferative colonies and formed more complex capillary-like networks on Matrigel compared with microvascular ECFCs. In contrast, in vivo assessment demonstrated that microvascular ECFCs had a greater potential to form vessels. Macrovascular ECFCs were of fetal origin, whereas microvascular ECFCs were of maternal origin. ECFCs exist in the micro- and macrovasculature of the normal, term human placenta. Although macrovascular ECFCs demonstrated greater vessel and colony-forming potency in vitro, this did not translate in vivo, where microvascular ECFCs exhibited a greater vessel-forming ability. These important findings contribute to the current understanding of normal placental vascular development and may aid in identifying factors involved in preeclampsia and other pregnancy complications.


Subject(s)
Cell Proliferation , Endothelial Cells/pathology , Placenta/cytology , Pre-Eclampsia/pathology , Antigens, CD/biosynthesis , Endothelial Progenitor Cells/pathology , Female , Fetal Blood/metabolism , Humans , Pre-Eclampsia/etiology , Pregnancy , Stem Cells/pathology
14.
Nat Protoc ; 10(11): 1697-708, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26448359

ABSTRACT

Blood vessels are crucial for the normal development, lifelong repair and homeostasis of tissues. Recently, vascular progenitor cell-driven 'postnatal vasculogenesis' has been suggested as an important mechanism that contributes to new blood vessel formation and organ repair. Among several described progenitor cell types that contribute to blood vessel formation, endothelial colony-forming cells (ECFCs) have received widespread attention as lineage-specific 'true' vascular progenitors. Here we describe a protocol for the isolation of pulmonary microvascular ECFCs from human and rat lung tissue. Our technique takes advantage of an earlier protocol for the isolation of circulating ECFCs from the mononuclear cellular fraction of peripheral blood. We adapted the earlier protocol to isolate resident ECFCs from the distal lung tissue. After enzymatic dispersion of rat or human lung samples into a cellular suspension, CD31-expressing cells are positively selected using magnetic-activated cell sorting and plated in endothelial-specific growth conditions. The colonies arising after 1-2 weeks in culture are carefully separated and expanded to yield pure ECFC cultures after a further 2-3 weeks. The resulting cells demonstrate the defining characteristics of ECFCs such as (i) 'cobblestone' morphology of cultured cell monolayers; (ii) acetylated low-density lipoprotein uptake and Ulex europaeus lectin binding; (iii) tube-like network formation in Matrigel; (iv) expression of endothelial cell-specific surface markers and the absence of hematopoietic or myeloid surface antigens; (v) self-renewal potential displayed by the most proliferative cells; and (vi) contribution to de novo vessel formation in an in vivo mouse implant model. Assuming typical initial cell adhesion and proliferation rates, the entire procedure can be completed within 4 weeks. Isolation and culture of lung vascular ECFCs will allow assessment of the functional state of these cells in experimental and human lung diseases, providing newer insights into their pathophysiological mechanisms.


Subject(s)
Cell Culture Techniques/methods , Endothelial Cells/physiology , Lung/cytology , Stem Cells/physiology , Animals , Cell Separation/methods , Cells, Cultured , Humans , Rats
15.
Circulation ; 129(21): 2144-57, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24710033

ABSTRACT

BACKGROUND: Bronchopulmonary dysplasia and emphysema are life-threatening diseases resulting from impaired alveolar development or alveolar destruction. Both conditions lack effective therapies. Angiogenic growth factors promote alveolar growth and contribute to alveolar maintenance. Endothelial colony-forming cells (ECFCs) represent a subset of circulating and resident endothelial cells capable of self-renewal and de novo vessel formation. We hypothesized that resident ECFCs exist in the developing lung, that they are impaired during arrested alveolar growth in experimental bronchopulmonary dysplasia, and that exogenous ECFCs restore disrupted alveolar growth. METHODS AND RESULTS: Human fetal and neonatal rat lungs contain ECFCs with robust proliferative potential, secondary colony formation on replating, and de novo blood vessel formation in vivo when transplanted into immunodeficient mice. In contrast, human fetal lung ECFCs exposed to hyperoxia in vitro and neonatal rat ECFCs isolated from hyperoxic alveolar growth-arrested rat lungs mimicking bronchopulmonary dysplasia proliferated less, showed decreased clonogenic capacity, and formed fewer capillary-like networks. Intrajugular administration of human cord blood-derived ECFCs after established arrested alveolar growth restored lung function, alveolar and lung vascular growth, and attenuated pulmonary hypertension. Lung ECFC colony- and capillary-like network-forming capabilities were also restored. Low ECFC engraftment and the protective effect of cell-free ECFC-derived conditioned media suggest a paracrine effect. Long-term (10 months) assessment of ECFC therapy showed no adverse effects with persistent improvement in lung structure, exercise capacity, and pulmonary hypertension. CONCLUSIONS: Impaired ECFC function may contribute to arrested alveolar growth. Cord blood-derived ECFC therapy may offer new therapeutic options for lung diseases characterized by alveolar damage.


Subject(s)
Cell Proliferation/drug effects , Endothelial Cells/physiology , Oxygen/toxicity , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/surgery , Stem Cell Transplantation/methods , Animals , Animals, Newborn , Cells, Cultured , Endothelial Cells/transplantation , Fetus , Human Umbilical Vein Endothelial Cells/physiology , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Pulmonary Alveoli/injuries , Rats , Rats, Nude , Rats, Sprague-Dawley
16.
J Cancer Prev ; 18(3): 235-48, 2013 Sep.
Article in English | MEDLINE | ID: mdl-25337551

ABSTRACT

BACKGROUND: Genome-wide association studies suggest that plasma triacylglyceride (TAG) in humans was associated with variation in the PLA2G6 locus, a gene that encodes calcium-independent phospholipase A2 (iPLA2 ß). The objective of the present study is to understand the impact of genetic inactivation of iPLA2 ß on hepatic TAG metabolism in C57BL/6 mice. METHODS: Male iPLA2 ß (+) (/-) mice were backcrossed with female iPLA2 ß (-/-) mice for up to 10 generations prior to experiments. Lipid and lipoprotein metabolism from plasma, hepatocytes, thigh subcutaneous adipose and thigh skeletal muscle tissues of the mice were determined under various experimental conditions. RESULTS: The iPLA2 ß (-/-) mice, either male or female as compared with iPLA2 ß (+) (/) (+) littermates, showed no change in fasted or postprandial plasma TAG or total cholesterol at young (12-15 weeks) or old (40-44 weeks) ages under chow diet or high-fat diet (HFD) conditions. However, fractionation of plasma lipoproteins showed that under HFD conditions, there was a significant increase (by 40%) in apoB-100 association with VLDL1 fractions in iPLA2 ß (-/-) mice as compared with iPLA2 ß (+) (/) (+) littermates. There was no significant difference in triglyceride or cholesterol contents in the liver, muscle, or adipose tissue between iPLA2 ß (-/-) and iPLA2 ß (+/+) littermates. Metabolic labeling experiments with cultured primary hepatocytes isolated from iPLA2 ß (-/-) mice also showed 2-fold increase in the secretion of [(35)S]methionine-labeled apoB-100 in VLDL1 fractions as compared with that from iPLA2 ß (+) (/) (+) hepatocytes. Likewise, secretion of [(3)H]palmitate-labeled TAG from the iPLA2 ß (-/-) hepatocytes was increased by 2-fold. CONCLUSIONS: Although iPLA2 ß may play a role in TAG-rich VLDL1 production from cultured hepatocytes, there is no evidence that inactivation of iPLA2 ß would lead to dyslipidemia in mice in vivo.

17.
Mol Cell Proteomics ; 10(10): O111.008425, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21749988

ABSTRACT

Despite their importance in many biological processes, membrane proteins are underrepresented in proteomic analysis because of their poor solubility (hydrophobicity) and often low abundance. We describe a novel approach for the identification of plasma membrane proteins and intracellular microsomal proteins that combines membrane fractionation, a centrifugal proteomic reactor for streamlined protein extraction, protein digestion and fractionation by centrifugation, and high performance liquid chromatography-electrospray ionization-tandem MS. The performance of this approach was illustrated for the study of the proteome of ER and Golgi microsomal membranes in rat hepatic cells. The centrifugal proteomic reactor identified 945 plasma membrane proteins and 955 microsomal membrane proteins, of which 63 and 47% were predicted as bona fide membrane proteins, respectively. Among these proteins, >800 proteins were undetectable by the conventional in-gel digestion approach. The majority of the membrane proteins only identified by the centrifugal proteomic reactor were proteins with ≥ 2 transmembrane segments or proteins with high molecular mass (e.g. >150 kDa) and hydrophobicity. The improved proteomic reactor allowed the detection of a group of endocytic and/or signaling receptor proteins on the plasma membrane, as well as apolipoproteins and glycerolipid synthesis enzymes that play a role in the assembly and secretion of apolipoprotein B100-containing very low density lipoproteins. Thus, the centrifugal proteomic reactor offers a new analytical tool for structure and function studies of membrane proteins involved in lipid and lipoprotein metabolism.


Subject(s)
Hepatocytes/chemistry , Membrane Proteins/analysis , Proteome/analysis , Proteomics/methods , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Centrifugation/methods , Chemical Fractionation , Chromatography, Liquid , Hepatocytes/metabolism , Lipid Metabolism , Lipoproteins/isolation & purification , Lipoproteins/metabolism , Membrane Proteins/isolation & purification , Microsomes/chemistry , Microsomes/metabolism , Proteolysis , Proteome/isolation & purification , Proteome/metabolism , Rats , Spectrometry, Mass, Electrospray Ionization
18.
J Biol Chem ; 286(31): 27769-80, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21676879

ABSTRACT

Hepatic assembly of triacylglycerol (TAG)-rich very low density lipoproteins (VLDL) is achieved through recruitment of bulk TAG (presumably in the form of lipid droplets within the microsomal lumen) into VLDL precursor containing apolipoprotein (apo) B-100. We determined protein/lipid components of lumenal lipid droplets (LLD) in cells expressing recombinant human apoC-III (C3wt) or a mutant form (K58E, C3KE) initially identified in humans that displayed hypotriglyceridemia. Although expression of C3wt markedly stimulated secretion of TAG and apoB-100 as VLDL(1), the K58E mutation (located at the C-terminal lipid binding domain) abolished the effect in transfected McA-RH7777 cells and in apoc3-null mice. Metabolic labeling studies revealed that accumulation of TAG in LLD was decreased (by 50%) in cells expressing C3KE. A Fat Western lipid protein overlay assay showed drastically reduced lipid binding of the mutant protein. Substituting Lys(58) with Arg demonstrated that the positive charge at position 58 is crucial for apoC-III binding to lipid and for promoting TAG secretion. On the other hand, substituting both Lys(58) and Lys(60) with Glu resulted in almost entire elimination of lipid binding and loss of function in promoting TAG secretion. Thus, the lipid binding domain of apoC-III plays a key role in the formation of LLD for hepatic VLDL assembly and secretion.


Subject(s)
Apolipoprotein C-III/metabolism , Lipoproteins, LDL/metabolism , Mutation, Missense , Triglycerides/metabolism , Apolipoprotein C-III/chemistry , Apolipoprotein C-III/genetics , Chromatography, Gel , Humans , Microsomes, Liver/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions/metabolism , Tandem Mass Spectrometry
19.
J Lipid Res ; 52(3): 540-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21189265

ABSTRACT

Hepatic lipase (HL) plays a role in the catabolism of apolipoprotein (apo)B-containing lipoproteins through its lipolytic and ligand-binding properties. We describe a potential intracellular role of HL in the assembly and secretion of VLDL. Transient or stable expression of HL in McA-RH7777 cells resulted in decreased (by 40%) incorporation of [(3)H]glycerol into cell-associated and secreted triacylglycerol (TAG) relative to control cells. However, incorporation of [(35)S]methionine/cysteine into cell and medium apoB-100 was not decreased by HL expression. The decreased (3)H-TAG synthesis/secretion in HL expressing cells was not attributable to decreased expression of genes involved in lipogenesis. Fractionation of medium revealed that the decreased [(3)H]TAG from HL expressing cells was mainly attributable to decreased VLDL. Expression of catalytically-inactive HL (HL(SG)) (Ser-145 at the catalytic site was substituted with Gly) in the cells also resulted in decreased secretion of VLDL-[(3)H]TAG. Examination of lumenal contents of microsomes showed a 40% decrease in [(3)H]TAG associated with lumenal lipid droplets in HL or HL(SG) expressing cells as compared with control. The microsomal membrane-associated [(3)H]TAG was decreased by 50% in HL expressing cells but not in HL(SG) expressing cells. Thus, expression of HL, irrespective of its lipolytic function, impairs formation of VLDL precursor [(3)H]TAG in the form of lumenal lipid droplets. These results suggest that HL expression in McA-RH7777 cells result in secretion of [(3)H]TAG-poor VLDL.


Subject(s)
Lipase/genetics , Lipoproteins, VLDL/chemistry , Lipoproteins, VLDL/metabolism , Triglycerides , Amino Acid Substitution , Biocatalysis , Cell Line , Gene Expression , Humans , Lipase/metabolism , Lipogenesis/genetics , Lipoproteins, VLDL/biosynthesis , Transfection
20.
J Lipid Res ; 51(6): 1524-34, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20097930

ABSTRACT

We have shown that expression of apolipoprotein (apo) C-III promotes VLDL secretion from transfected McA-RH7777 cells under lipid-rich conditions. To determine structural elements within apoC-III that confer to this function, we contrasted wild-type apoC-III with a mutant Ala23Thr originally identified in hypotriglyceridemia subjects. Although synthesis of [(3)H]glycerol-labeled TAG was comparable between cells expressing wild-type apoC-III (C3wt cells) or Ala23Thr mutant (C3AT cells), secretion of [(3)H]TAG from C3AT cells was markedly decreased. The lowered [(3)H]TAG secretion was associated with an inability of C3AT cells to assemble VLDL(1). Moreover, [(3)H]TAG within the microsomal lumen in C3AT cells was 60% higher than that in C3wt cells, yet the activity of microsomal triglyceride-transfer protein in C3AT cells was not elevated. The accumulated [(3)H]TAG in C3AT microsomal lumen was mainly associated with lumenal IDL/LDL-like lipoproteins. Phenotypically, this [(3)H]TAG fractionation profiling resembled what was observed in cells treated with brefeldin A, which at low dose specifically blocked the second-step VLDL(1) maturation. Furthermore, lumenal [(35)S]Ala23Thr protein accumulated in IDL/LDL fractions and was absent in VLDL fractions in C3AT cells. These results suggest that the presence of Ala23Thr protein in lumenal IDL/LDL particles might prevent effective fusion between lipid droplets and VLDL precursors. Thus, the current study reveals an important structural element residing within the N-terminal region of apoC-III that governs the second step VLDL(1) maturation.


Subject(s)
Apolipoproteins C/genetics , Apolipoproteins C/metabolism , Lipid Metabolism Disorders/genetics , Mutation, Missense , Triglycerides/metabolism , Alanine/genetics , Apolipoproteins C/chemistry , Brefeldin A/pharmacology , Carrier Proteins/metabolism , Cell Line , Gene Expression Regulation , Humans , Lipoproteins, IDL/metabolism , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/metabolism , Microsomes/metabolism , Models, Molecular , Protein Binding/drug effects , Protein Structure, Secondary , Threonine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...