Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Article in English | MEDLINE | ID: mdl-38670297

ABSTRACT

BACKGROUND: Cardiac allograft vasculopathy (CAV) remains the leading cause of long-term graft failure and mortality after heart transplantation. Effective preventive and treatment options are not available to date, largely because underlying mechanisms remain poorly understood. We studied the potential role of leukotriene B4 (LTB4), an inflammatory lipid mediator, in the development of CAV. METHODS: We used an established preclinical rat CAV model to study the role of LTB4 in CAV. We performed syngeneic and allogeneic orthotopic aortic transplantation, after which neointimal proliferation was quantified. Animals were then treated with Bestatin, an inhibitor of LTB4 synthesis, or vehicle control for 30 days post-transplant, and evidence of graft CAV was determined by histology. We also measured serial LTB4 levels in a cohort of 28 human heart transplant recipients with CAV, 17 matched transplant controls without CAV, and 20 healthy nontransplant controls. RESULTS: We showed that infiltration of the arterial wall with macrophages leads to neointimal thickening and a rise in serum LTB4 levels in our rat model of CAV. Inhibition of LTB4 production with the drug Bestatin prevents development of neointimal hyperplasia, suggesting that Bestatin may be effective therapy for CAV prevention. In a parallel study of heart transplant recipients, we found nonsignificantly elevated plasma LTB4 levels in patients with CAV, compared to patients without CAV and healthy, nontransplant controls. CONCLUSIONS: This study provides key evidence supporting the role of the inflammatory cytokine LTB4 as an important mediator of CAV development and provides preliminary data suggesting the clinical benefit of Bestatin for CAV prevention.

2.
Cell Stem Cell ; 31(3): 334-340.e5, 2024 03 07.
Article in English | MEDLINE | ID: mdl-38335966

ABSTRACT

Allogeneic transplantation of pancreatic islets for patients with difficult-to-control diabetes mellitus is severely hampered by the requirement for continuous immunosuppression and its associated morbidity. We report that allogeneic transplantation of genetically engineered (B2M-/-, CIITA-/-, CD47+), primary, hypoimmune, pseudo-islets (p-islets) results in their engraftment into a fully immunocompetent, diabetic non-human primate wherein they provide stable endocrine function and enable insulin independence without inducing any detectable immune response in the absence of immunosuppression. Hypoimmune primary p-islets may provide a curative cell therapy for type 1 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Islets of Langerhans , Animals , Humans , Insulin/metabolism , Islets of Langerhans Transplantation/methods , Islets of Langerhans/metabolism , Primates , Diabetes Mellitus, Type 1/therapy , Transplantation, Homologous
3.
Nat Biotechnol ; 42(3): 413-423, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37156915

ABSTRACT

Genetic engineering of allogeneic cell therapeutics that fully prevents rejection by a recipient's immune system would abolish the requirement for immunosuppressive drugs or encapsulation and support large-scale manufacturing of off-the-shelf cell products. Previously, we generated mouse and human hypoimmune pluripotent (HIP) stem cells by depleting HLA class I and II molecules and overexpressing CD47 (B2M-/-CIITA-/-CD47+). To determine whether this strategy is successful in non-human primates, we engineered rhesus macaque HIP cells and transplanted them intramuscularly into four allogeneic rhesus macaques. The HIP cells survived unrestricted for 16 weeks in fully immunocompetent allogeneic recipients and differentiated into several lineages, whereas allogeneic wild-type cells were vigorously rejected. We also differentiated human HIP cells into endocrinologically active pancreatic islet cells and showed that they survived in immunocompetent, allogeneic diabetic humanized mice for 4 weeks and ameliorated diabetes. HIP-edited primary rhesus macaque islets survived for 40 weeks in an allogeneic rhesus macaque recipient without immunosuppression, whereas unedited islets were quickly rejected.


Subject(s)
Hematopoietic Stem Cell Transplantation , Induced Pluripotent Stem Cells , Islets of Langerhans Transplantation , Mice , Animals , Macaca mulatta , CD47 Antigen , Graft Rejection
4.
Cell Stem Cell ; 30(11): 1538-1548.e4, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37922880

ABSTRACT

Immune rejection of allogeneic cell therapeutics remains a major problem for immuno-oncology and regenerative medicine. Allogeneic cell products so far have inferior persistence and efficacy when compared with autologous alternatives. Engineering of hypoimmune cells may greatly improve their therapeutic benefit. We present a new class of agonistic immune checkpoint engagers that protect human leukocyte antigen (HLA)-depleted induced pluripotent stem cell-derived endothelial cells (iECs) from innate immune cells. Engagers with agonistic functionality to their inhibitory receptors TIM3 and SIRPα effectively protect engineered iECs from natural killer (NK) cell and macrophage killing. The SIRPα engager can be combined with truncated CD64 to generate fully immune evasive iECs capable of escaping allogeneic cellular and immunoglobulin G (IgG) antibody-mediated rejection. Synthetic immune checkpoint engagers have high target specificity and lack retrograde signaling in the engineered cells. This modular design allows for the exploitation of more inhibitory immune pathways for immune evasion and could contribute to the advancement of allogeneic cell therapeutics.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Endothelial Cells/metabolism , HLA Antigens , Killer Cells, Natural , Immunity, Innate
5.
Sci Transl Med ; 15(691): eadg5794, 2023 04 12.
Article in English | MEDLINE | ID: mdl-37043559

ABSTRACT

Transplantation of allogeneic pancreatic donor islets has successfully been performed in selected patients with difficult-to-control insulin-dependent diabetes and impaired awareness of hypoglycemia (IAH). However, the required systemic immunosuppression associated with this procedure prevents this cell replacement therapy from more widespread adoption in larger patient populations. We report the editing of primary human islet cells to the hypoimmune HLA class I- and class II-negative and CD47-overexpressing phenotype and their reaggregation into human HIP pseudoislets (p-islets). Human HIP p-islets were shown to survive, engraft, and ameliorate diabetes in immunocompetent, allogeneic, diabetic humanized mice. HIP p-islet cells were further shown to avoid autoimmune killing in autologous, diabetic humanized autoimmune mice. The survival and endocrine function of HIP p-islet cells were not impaired by contamination of unedited or partially edited cells within the p-islets. HIP p-islet cells were eliminated quickly and reliably in this model using a CD47-targeting antibody, thus providing a safety strategy in case HIP cells exert toxicity in a future clinical setting. Transplantation of human HIP p-islets for which no immunosuppression is required has the potential to lead to wider adoption of this therapy and help more diabetes patients with IAH and history of severe hypoglycemic events to achieve insulin independence.


Subject(s)
Diabetes Mellitus, Type 1 , Hematopoietic Stem Cell Transplantation , Islets of Langerhans Transplantation , Islets of Langerhans , Humans , Animals , Mice , CD47 Antigen , Islets of Langerhans Transplantation/methods , Autoimmunity , Diabetes Mellitus, Type 1/therapy , Insulin
6.
Nat Commun ; 14(1): 2020, 2023 04 10.
Article in English | MEDLINE | ID: mdl-37037829

ABSTRACT

Manufacturing autologous chimeric antigen receptor (CAR) T cell therapeutics is complex, and many patients experience treatment delays or cannot be treated at all. Although current allogeneic CAR products have the potential to overcome manufacturing bottlenecks, they are subject to immune rejection and failure to persist in the host, and thus do not provide the same level of efficacy as their autologous counterparts. Here, we aimed to develop universal allogeneic CAR T cells that evade the immune system and produce a durable response. We generated human hypoimmune (HIP) T cells with disrupted B2M, CIITA, and TRAC genes using CRISPR-Cas9 editing. In addition, CD47 and anti-CD19 CAR were expressed using lentiviral transduction. These allogeneic HIP CD19 CAR T cells were compared to allogeneic CD19 CAR T cells that only expressed the anti-CD19 CAR (allo CAR T). In vitro assays for cancer killing and exhaustion revealed no differences between allo CAR T and HIP CAR T cells, confirming that the HIP edits did not negatively affect T cell performance. Clearance of CD19+ tumors by HIP CAR T cells in immunodeficient NSG mice was comparable to that of allo CAR T cells. In fully immunocompetent humanized mice, HIP CAR T cells significantly outperformed allo CAR T cells, showed improved persistence and expansion, and provided lasting cancer clearance. Furthermore, CD47-targeting safety strategies reliably and specifically eliminated HIP CAR T cells. These findings suggest that universal allogeneic HIP CAR T cell-based therapeutics might overcome the limitations associated with poor persistence of allogeneic CAR T cells and exert durable anti-tumor responses.


Subject(s)
Hematopoietic Stem Cell Transplantation , Neoplasms , Receptors, Chimeric Antigen , Humans , Mice , Animals , Receptors, Chimeric Antigen/genetics , CD47 Antigen , T-Lymphocytes , Receptors, Antigen, T-Cell/genetics
7.
Nat Biotechnol ; 41(5): 717-727, 2023 05.
Article in English | MEDLINE | ID: mdl-36593395

ABSTRACT

Allogeneic cell therapeutics for cancer therapy or regenerative medicine are susceptible to antibody-mediated killing, which diminishes their efficacy. Here we report a strategy to protect cells from antibody-mediated killing that relies on engineered overexpression of the IgG receptor CD64. We show that human and mouse iPSC-derived endothelial cells (iECs) overexpressing CD64 escape antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity from IgG antibodies in vitro and in ADCC-enabled mice. When CD64 expression was combined with hypoimmune genetic modifications known to protect against cellular immunity, B2M-/-CIITA-/- CD47/CD64-transgenic iECs were resistant to both IgG antibody-mediated and cellular immune killing in vitro and in humanized mice. Mechanistic studies demonstrated that CD64 or its intracellularly truncated analog CD64t effectively capture monomeric IgG and occupy their Fc, and the IgG bind and occupy their target antigens. In three applications of the approach, human CD64t-engineered thyroid epithelial cells, pancreatic beta cells and CAR T cells withstood clinically relevant levels of graft-directed antibodies and fully evaded antibody-mediated killing.


Subject(s)
Endothelial Cells , Receptors, IgG , Humans , Animals , Mice , Endothelial Cells/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Immunoglobulin G/genetics , Antibody-Dependent Cell Cytotoxicity , Immunity, Cellular
9.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34244428

ABSTRACT

The emerging field of regenerative cell therapy is still limited by the few cell types that can reliably be differentiated from pluripotent stem cells and by the immune hurdle of commercially scalable allogeneic cell therapeutics. Here, we show that gene-edited, immune-evasive cell grafts can survive and successfully treat diseases in immunocompetent, fully allogeneic recipients. Transplanted endothelial cells improved perfusion and increased the likelihood of limb preservation in mice with critical limb ischemia. Endothelial cell grafts transduced to express a transgene for alpha1-antitrypsin (A1AT) successfully restored physiologic A1AT serum levels in mice with genetic A1AT deficiency. This cell therapy prevented both structural and functional changes of emphysematous lung disease. A mixture of endothelial cells and cardiomyocytes was injected into infarcted mouse hearts, and both cell types orthotopically engrafted in the ischemic areas. Cell therapy led to an improvement in invasive hemodynamic heart failure parameters. Our study supports the development of hypoimmune, universal regenerative cell therapeutics for cost-effective treatments of major diseases.


Subject(s)
Cardiovascular Diseases/immunology , Cardiovascular Diseases/therapy , Immunocompetence , Induced Pluripotent Stem Cells/immunology , Lung Diseases/immunology , Lung Diseases/therapy , Stem Cell Transplantation , Animals , Endothelial Cells/transplantation , Heart Failure/therapy , Hindlimb/blood supply , Hindlimb/pathology , Ischemia/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocytes, Cardiac/transplantation , Transplantation, Homologous , alpha 1-Antitrypsin/metabolism
10.
Stem Cells Dev ; 30(10): 515-525, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33726521

ABSTRACT

Inflammation plays a major role in the pathogenesis of pulmonary hypertension (PH). We sought to investigate the effects of a cell-based immunomodulation in a dysimmune model of PH. PH was induced in athymic nude rats using semaxinib (Su group, n = 6). Tolerogenic macrophages (toM) were generated from monocyte isolation and then injected either the day before semaxinib injection (Prevention group, n = 6) or 3 weeks after (Reversion group, n = 6). Six athymic nude rats were used as controls. In vivo trafficking of toM was investigated with bioluminescence imaging showing that toM were mainly located into the lungs until 48 h after injection. Right ventricular (RV) end-systolic pressure and RV systolic function were assessed at 4 weeks using echocardiography. Morphometric analysis and RNA sequencing of the lungs were realized at 4 weeks. Rats treated with toM (Prevention and Reversion groups) had a significantly lower RV end-systolic pressure at 4 weeks (respectively, 25 ± 8 and 30 ± 6 mmHg vs. 67 ± 9 mmHg, P < 0.001), while RV systolic dysfunction was observed in Su and Reversion groups. Mean medial wall thickness of small arterioles was lower in Prevention and Reversion groups compared with the Su group (respectively, 10.9% ± 0.8% and 16.4% ± 1.3% vs. 28.2% ± 2.1%, P < 0.001). Similarly, cardiomyocyte area was decreased in rats treated with toM (150 ± 18 and 160 ± 86 µm2 vs. 279 ± 50 µm2, P < 0.001). A trend toward upregulation of genes involved in pulmonary arterial hypertension pathobiology was found in Su rats, while KCNK3 was significantly downregulated (fold-change = 9.8, P < 0.001). Injection of toM was associated with a less severe phenotype of PH in rats exposed to angioproliferative stress. Preserved expression of KCNK3 may explain the protective effect of toM.


Subject(s)
Disease Models, Animal , Hypertension, Pulmonary/therapy , Immunomodulation/immunology , Immunotherapy/methods , Macrophages/immunology , Animals , Gene Expression Profiling/methods , Humans , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/physiopathology , Immune Tolerance/immunology , Indoles/pharmacology , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Rats, Nude , Rodentia , Stroke Volume/drug effects , Stroke Volume/immunology , Stroke Volume/physiology , Tomography, Emission-Computed, Single-Photon
11.
J Exp Med ; 218(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33416832

ABSTRACT

Here we report on the existence and functionality of the immune checkpoint signal regulatory protein α (SIRPα) in NK cells and describe how it can be modulated for cell therapy. NK cell SIRPα is up-regulated upon IL-2 stimulation, interacts with target cell CD47 in a threshold-dependent manner, and counters other stimulatory signals, including IL-2, CD16, or NKG2D. Elevated expression of CD47 protected K562 tumor cells and mouse and human MHC class I-deficient target cells against SIRPα+ primary NK cells, but not against SIRPα- NKL or NK92 cells. SIRPα deficiency or antibody blockade increased the killing capacity of NK cells. Overexpression of rhesus monkey CD47 in human MHC-deficient cells prevented cytotoxicity by rhesus NK cells in a xenogeneic setting. The SIRPα-CD47 axis was found to be highly species specific. Together, the results demonstrate that disruption of the SIRPα-CD47 immune checkpoint may augment NK cell antitumor responses and that elevated expression of CD47 may prevent NK cell-mediated killing of allogeneic and xenogeneic tissues.


Subject(s)
CD47 Antigen/metabolism , Killer Cells, Natural/metabolism , Receptors, Immunologic/metabolism , Animals , Cells, Cultured , Cytokines/pharmacology , Cytotoxicity, Immunologic/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Immune Checkpoint Inhibitors , Killer Cells, Natural/drug effects , Macaca mulatta , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Binding/drug effects , Species Specificity
12.
Cell ; 183(5): 1185-1201.e20, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33242417

ABSTRACT

Spaceflight is known to impose changes on human physiology with unknown molecular etiologies. To reveal these causes, we used a multi-omics, systems biology analytical approach using biomedical profiles from fifty-nine astronauts and data from NASA's GeneLab derived from hundreds of samples flown in space to determine transcriptomic, proteomic, metabolomic, and epigenetic responses to spaceflight. Overall pathway analyses on the multi-omics datasets showed significant enrichment for mitochondrial processes, as well as innate immunity, chronic inflammation, cell cycle, circadian rhythm, and olfactory functions. Importantly, NASA's Twin Study provided a platform to confirm several of our principal findings. Evidence of altered mitochondrial function and DNA damage was also found in the urine and blood metabolic data compiled from the astronaut cohort and NASA Twin Study data, indicating mitochondrial stress as a consistent phenotype of spaceflight.


Subject(s)
Genomics , Mitochondria/pathology , Space Flight , Stress, Physiological , Animals , Circadian Rhythm , Extracellular Matrix/metabolism , Humans , Immunity, Innate , Lipid Metabolism , Metabolic Flux Analysis , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscles/immunology , Organ Specificity , Smell/physiology
13.
Stem Cells Dev ; 29(18): 1179-1189, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32723003

ABSTRACT

Pluripotent stem cells are promising candidates for cell-based regenerative therapies. To avoid rejection of transplanted cells, several approaches are being pursued to reduce immunogenicity of the cells or modulate the recipient's immune response. These include gene editing to reduce the antigenicity of cell products, immunosuppression of the host, or using major histocompatibility complex-matched cells from cell banks. In this context, we have investigated the antigenicity of H-Y antigens, a class of minor histocompatibility antigens encoded by the Y chromosome, to assess whether the gender of the donor affects the cell's antigenicity. In a murine transplant model, we show that the H-Y antigen in undifferentiated embryonic stem cells (ESCs), as well as ESC-derived endothelial cells, provokes T- and B cell responses in female recipients.


Subject(s)
Embryonic Stem Cells/metabolism , Graft Rejection/immunology , H-Y Antigen/metabolism , Animals , Animals, Newborn , B-Lymphocytes/immunology , Female , Immune Tolerance , Immunity , Male , Mice , Mice, Inbred BALB C , Stem Cell Transplantation , Survival Analysis , T-Lymphocytes/immunology
14.
J Vis Exp ; (151)2019 09 19.
Article in English | MEDLINE | ID: mdl-31609308

ABSTRACT

The use of animal models is essential for developing new therapeutic strategies for acute coronary syndrome and its complications. In this article, we demonstrate a murine cryoinjury infarct model that generates precise infarct sizes with high reproducibility and replicability. In brief, after intubation and sternotomy of the animal, the heart is lifted from the thorax. The probe of a handheld liquid nitrogen delivery system is applied onto the myocardial wall to induce cryoinjury. Impaired ventricular function and electrical conduction can be monitored with echocardiography or optical mapping. Transmural myocardial remodeling of the infarcted area is characterized by collagen deposition and loss of cardiomyocytes. Compared to other models (e.g., LAD-ligation), this model utilizes a handheld liquid nitrogen delivery system to generate more uniform infarct sizes.


Subject(s)
Cryosurgery/adverse effects , Disease Models, Animal , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/physiopathology , Animals , Echocardiography/methods , Mice , Mice, Inbred BALB C , Myocardial Infarction/etiology , Myocardium/pathology , Myocytes, Cardiac/pathology , Reproducibility of Results
15.
Nat Biotechnol ; 37(10): 1137-1144, 2019 10.
Article in English | MEDLINE | ID: mdl-31427818

ABSTRACT

The utility of autologous induced pluripotent stem cell (iPSC) therapies for tissue regeneration depends on reliable production of immunologically silent functional iPSC derivatives. However, rejection of autologous iPSC-derived cells has been reported, although the mechanism underlying rejection is largely unknown. We hypothesized that de novo mutations in mitochondrial DNA (mtDNA), which has far less reliable repair mechanisms than chromosomal DNA, might produce neoantigens capable of eliciting immune recognition and rejection. Here we present evidence in mice and humans that nonsynonymous mtDNA mutations can arise and become enriched during reprogramming to the iPSC stage, long-term culture and differentiation into target cells. These mtDNA mutations encode neoantigens that provoke an immune response that is highly specific and dependent on the host major histocompatibility complex genotype. Our results reveal that autologous iPSCs and their derivatives are not inherently immunologically inert for autologous transplantation and suggest that iPSC-derived products should be screened for mtDNA mutations.


Subject(s)
DNA, Mitochondrial/genetics , Epitopes/genetics , Epitopes/immunology , Graft vs Host Disease/immunology , Induced Pluripotent Stem Cells , Animals , Antigens , Cell Transplantation/methods , Embryonic Stem Cells , Graft Rejection/immunology , Humans , Kidney Transplantation/adverse effects , Liver Transplantation/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Transplantation, Autologous
16.
Nat Biotechnol ; 37(3): 252-258, 2019 03.
Article in English | MEDLINE | ID: mdl-30778232

ABSTRACT

Autologous induced pluripotent stem cells (iPSCs) constitute an unlimited cell source for patient-specific cell-based organ repair strategies. However, their generation and subsequent differentiation into specific cells or tissues entail cell line-specific manufacturing challenges and form a lengthy process that precludes acute treatment modalities. These shortcomings could be overcome by using prefabricated allogeneic cell or tissue products, but the vigorous immune response against histo-incompatible cells has prevented the successful implementation of this approach. Here we show that both mouse and human iPSCs lose their immunogenicity when major histocompatibility complex (MHC) class I and II genes are inactivated and CD47 is over-expressed. These hypoimmunogenic iPSCs retain their pluripotent stem cell potential and differentiation capacity. Endothelial cells, smooth muscle cells, and cardiomyocytes derived from hypoimmunogenic mouse or human iPSCs reliably evade immune rejection in fully MHC-mismatched allogeneic recipients and survive long-term without the use of immunosuppression. These findings suggest that hypoimmunogenic cell grafts can be engineered for universal transplantation.


Subject(s)
Cell Differentiation/immunology , Graft Rejection/immunology , HLA Antigens/genetics , Induced Pluripotent Stem Cells/transplantation , Animals , Cell Differentiation/genetics , Graft Rejection/genetics , HLA Antigens/immunology , Histocompatibility Antigens Class I/genetics , Humans , Mice , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Transplantation, Homologous/methods
17.
Thorax ; 74(3): 219-228, 2019 03.
Article in English | MEDLINE | ID: mdl-30636196

ABSTRACT

BACKGROUND: In acute respiratory distress syndrome (ARDS), pulmonary perfusion failure increases physiologic dead space ventilation (VD/VT), leading to a decline of the alveolar CO2 concentration [CO2]iA. Although it has been shown that alveolar hypocapnia contributes to formation of atelectasis and surfactant depletion, a typical complication in ARDS, the underlying mechanism has not been elucidated so far. METHODS: In isolated perfused rat lungs, cytosolic or mitochondrial Ca2+ concentrations ([Ca2+]cyt or [Ca2+]mito, respectively) of alveolar epithelial cells (AECs), surfactant secretion and the projected area of alveoli were quantified by real-time fluorescence or bright-field imaging (n=3-7 per group). In ventilated White New Zealand rabbits, the left pulmonary artery was ligated and the size of subpleural alveoli was measured by intravital microscopy (n=4 per group). Surfactant secretion was determined in the bronchoalveolar lavage (BAL) by western blot. RESULTS: Low [CO2]iA decreased [Ca2+]cyt and increased [Ca2+]mito in AECs, leading to reduction of Ca2+-dependent surfactant secretion, and alveolar ventilation in situ. Mitochondrial inhibition by ruthenium red or rotenone blocked these responses indicating that mitochondria are key players in CO2 sensing. Furthermore, ligature of the pulmonary artery of rabbits decreased alveolar ventilation, surfactant secretion and lung compliance in vivo. Addition of 5% CO2 to the inspiratory gas inhibited these responses. CONCLUSIONS: Accordingly, we provide evidence that alveolar hypocapnia leads to a Ca2+ shift from the cytosol into mitochondria. The subsequent decline of [Ca2+]cyt reduces surfactant secretion and thus regional ventilation in lung regions with high VD/VT. Additionally, the regional hypoventilation provoked by perfusion failure can be inhibited by inspiratory CO2 application.


Subject(s)
Hypocapnia/etiology , Mitochondria/physiology , Pulmonary Surfactants/metabolism , Respiratory Distress Syndrome/etiology , Tidal Volume/physiology , Animals , Disease Models, Animal , Pulmonary Alveoli/blood supply , Rats , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/physiopathology
19.
Pharmacol Res Perspect ; 6(3): e00407, 2018 06.
Article in English | MEDLINE | ID: mdl-29864245

ABSTRACT

The transcription factor Islet-1 marks a progenitor cell population of the second heart field during cardiogenesis. In the adult heart Islet-1 expression is limited to the sinoatrial node, the ventricular outflow tract, and parasympathetic ganglia. The regenerative effect in the injured mouse ventricle of thymosin beta-4 (TB4), a 43-aminoacid peptide, was associated with increased Islet-1 immunostaining, suggesting the induction of an Islet-1-positive progenitor state by TB4. Here we aimed to reassess this effect in a genetic model. Mice from the reporter mouse line Isl1-nLacZ were primed with TB4 and subsequently underwent myocardial infarction. Islet-1 expression was assessed 2, 7, and 14 days after infarction. We detected only a single Islet-1+ cell in 8 TB4 treated and infarcted hearts which located outside of the sinoatrial node, the outflow tract or cardiac ganglia (in ~2500 sections). Two cells were identified in 5 control infarcted hearts. TB4 did not induce LacZ positivity in ventricular explants cultures of Isl1-nLacZ mice nor did it affect the density of LacZ+ cells in explant cultures of nLacZ+ regions of the heart. In summary, we found no evidence that TB4 reactivates Islet-1 expression in adult mouse ventricle.


Subject(s)
Heart Ventricles/drug effects , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Myocardial Infarction/genetics , Thymosin/administration & dosage , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Heart Ventricles/cytology , Heart Ventricles/metabolism , Mice , Mice, Transgenic , Sinoatrial Node/cytology , Sinoatrial Node/drug effects , Sinoatrial Node/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Thymosin/pharmacology
20.
J Vasc Surg ; 68(6S): 201S-207S, 2018 12.
Article in English | MEDLINE | ID: mdl-29804740

ABSTRACT

OBJECTIVE: The main objective of this study was to define a role of sphingosine-1-phosphate receptor 1 (S1PR1) in the arterial injury response of a human artery. The hypotheses were tested that injury induces an expansion of S1PR1-positive cells and that these cells accumulate toward the lumen because they follow the sphingosine-1-phosphate gradient from arterial wall tissue (low) to plasma (high). METHODS: A humanized rat model was used in which denuded human internal mammary artery (IMA) was implanted into the position of the abdominal aorta of immunosuppressed Rowett nude rats. This injury model is characterized by medial as well as intimal hyperplasia, whereby intimal cells are of human origin. At 7, 14, and 28 days after implantation, grafts were harvested and processed for fluorescent immunostaining for S1PR1 and smooth muscle α-actin. Nuclei were stained with 4',6-diamidine-2'-phenylindole dihydrochloride. Using digitally reconstructed, complete cross sections of grafts, intimal and medial areas were measured, whereby the medial area had virtually been divided into an outer (toward adventitia) and inner (toward lumen) layer. The fraction of S1PR1-positive cells was determined in each layer by counting S1PR1-positive and S1PR1-negative cells. RESULTS: The fraction of S1PR1-postive cells in naive IMA is 58.9% ± 6.0% (mean ± standard deviation). At day 28 after implantation, 81.6% ± 4.4% of medial cells were scored S1PR1 positive (P < .01). At day 14, the ratio between S1PR1-positive and S1PR1-negative cells was significantly higher in the lumen-oriented inner layer (9.3 ± 2.1 vs 6.0 ± 1.0; P < .01). Cells appearing in the intima at day 7 and day 14 were almost all S1PR1 positive. At day 28, however, about one-third of intimal cells were scored S1PR1 negative. CONCLUSIONS: From these data, we conclude that denudation of IMA specifically induces the expansion of S1PR1-positive cells. Based on the nonrandom distribution of S1PR1-positive cells, we consider the possibility that much like lymphocytes, S1PR1-positive smooth muscle cells also use S1PR1 to recognize the sphingosine-1-phosphate gradient from tissue (low) to plasma (high) and so migrate out of the media toward the intima of the injured IMA.


Subject(s)
Aorta, Abdominal/surgery , Graft Occlusion, Vascular/metabolism , Mammary Arteries/transplantation , Muscle, Smooth, Vascular/transplantation , Myocytes, Smooth Muscle/transplantation , Neointima , Receptors, Lysosphingolipid/metabolism , Animals , Cell Movement , Cell Proliferation , Disease Models, Animal , Graft Occlusion, Vascular/etiology , Graft Occlusion, Vascular/pathology , Humans , Lysophospholipids/metabolism , Male , Mammary Arteries/metabolism , Mammary Arteries/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Rats, Nude , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...