Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Mol Cell Cardiol ; 164: 69-82, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34838588

RESUMEN

The global propagation of SARS-CoV-2 leads to an unprecedented public health emergency. Despite that the lungs are the primary organ targeted by COVID-19, systemic endothelial inflammation and dysfunction is observed particularly in patients with severe COVID-19, manifested by elevated endothelial injury markers, endotheliitis, and coagulopathy. Here, we review the clinical characteristics of COVID-19 associated endothelial dysfunction; and the likely pathological mechanisms underlying the disease including direct cell entry or indirect immune overreactions after SARS-CoV-2 infection. In addition, we discuss potential biomarkers that might indicate the disease severity, particularly related to the abnormal development of thrombosis that is a fatal vascular complication of severe COVID-19. Furthermore, we summarize clinical trials targeting the direct and indirect pathological pathways after SARS-CoV-2 infection to prevent or inhibit the virus induced endothelial disorders.


Asunto(s)
COVID-19/patología , Endotelio Vascular/patología , SARS-CoV-2 , Adolescente , Adulto , Anciano , Enzima Convertidora de Angiotensina 2/fisiología , Animales , COVID-19/sangre , COVID-19/complicaciones , COVID-19/fisiopatología , COVID-19/terapia , Ensayos Clínicos como Asunto , Células Endoteliales/patología , Células Endoteliales/virología , Endotelio Vascular/inmunología , Endotelio Vascular/fisiopatología , Proteína HMGB1/fisiología , Humanos , Macaca mulatta , Ratones , Neuropilina-1/fisiología , Estrés Oxidativo , Especies Reactivas de Oxígeno , Receptores Virales/fisiología , Receptores Depuradores de Clase B/fisiología , Índice de Severidad de la Enfermedad , Transducción de Señal , Síndrome de Respuesta Inflamatoria Sistémica/patología , Síndrome de Respuesta Inflamatoria Sistémica/fisiopatología , Trombofilia/etiología , Trombofilia/fisiopatología , Factor A de Crecimiento Endotelial Vascular/fisiología , Vasculitis/etiología , Vasculitis/inmunología , Vasculitis/fisiopatología , Adulto Joven
2.
Int J Mol Sci ; 21(16)2020 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-32796510

RESUMEN

Autoimmune diabetes is a complex multifactorial disease with genetic and environmental factors playing pivotal roles. While many genes associated with the risk of diabetes have been identified to date, the mechanisms by which external triggers contribute to the genetic predisposition remain unclear. Here, we derived embryonic stem (ES) cell lines from diabetes-prone non-obese diabetic (NOD) and healthy C57BL/6 (B6) mice. While overall pluripotency markers were indistinguishable between newly derived NOD and B6 ES cells, we discovered several differentially expressed genes that normally are not expressed in ES cells. Several genes that reside in previously identified insulin-dependent diabetics (Idd) genomic regions were up-regulated in NOD ES cells. Gene set enrichment analysis showed that different groups of genes associated with immune functions are differentially expressed in NOD. Transcriptomic analysis of NOD blastocysts validated several differentially overexpressed Idd genes compared to B6. Genome-wide mapping of active histone modifications using ChIP-Seq supports active expression as the promoters and enhancers of activated genes are also marked by active histone modifications. We have also found that NOD ES cells secrete more inflammatory cytokines. Our data suggest that the known genetic predisposition of NOD to autoimmune diabetes leads to epigenetic instability of several Idd regions.


Asunto(s)
Autoinmunidad/genética , Blastocisto/metabolismo , Sistema Inmunológico/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Transcripción Genética , Animales , Quimiocinas/metabolismo , Cromatina/metabolismo , Diabetes Mellitus Experimental/genética , Epigénesis Genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Proteoma/metabolismo , Proteómica , Transcriptoma/genética
3.
J Cell Biochem ; 118(6): 1349-1360, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27862217

RESUMEN

Giant cell tumor of bone (GCTB) is the most common non-malignant primary bone tumor reported in Hong Kong. Failure of treatment in advanced GCTB with aggressive local recurrence remains a clinical challenge. In order to reveal the molecular mechanism underlying the pathogenesis of this tumor, we aimed to examine the transcriptome profiling of the neoplastic stromal cells of GCTB in this study. RNA-sequencing was performed on three GCTB stromal cell samples and one bone marrow-derived MSC sample and 174 differentially expressed genes (DEGs) were identified between these two cell types. The top five up-regulated genes are SPP1, F3, TSPAN12, MMP13, and LGALS3BP and further validated by qPCR and Western Blotting. Knockdown of SPP1 was found to induce RUNX2 and OPG expression in GCTB stromal cells but not the MSCs. Ingenuity pathway analysis (IPA) of the 174 DEGs revealed significant alternations in 23 pathways; variant calling analysis revealed 1915 somatic variants of 384 genes with high or moderate impacts. Interestingly, four canonical pathways were found overlapping in both analyses; from which VEGFA, CSF1, PLAUR, and F3 genes with somatic mutation were found up-regulated in GCTB stromal cells. The STRING diagram showed two main clusters of the DEGs; one cluster of histone genes that are down-regulated in GCTB samples and another related to osteoblast differentiation, angiogenesis, cell cycle progression, and tumor growth. The DEGs and somatic mutations found in our study warrant further investigation and validation, nevertheless, our study add new insights in the search for new therapeutic targets in treating GCTB. J. Cell. Biochem. 118: 1349-1360, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Neoplasias Óseas/genética , Perfilación de la Expresión Génica/métodos , Tumor Óseo de Células Gigantes/genética , Análisis de Secuencia de ARN/métodos , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Mutación
5.
J Leukoc Biol ; 115(1): 19-35, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-37675661

RESUMEN

Age-related immunosenescence is characterized by progressive dysfunction of adaptive immune response and increased autoimmunity. Nevertheless, the impact of aging on CD4+ regulatory T cells that are master regulators of the immune system remains largely unclear. Here, we report cellular and molecular hallmarks of regulatory T cells derived from murine lymphoid and adipose tissues at 3, 18, and 24 mo of age, respectively, by analyzing their heterogeneity that displays dynamic changes in transcriptomic effector signatures at a single-cell resolution. Although the proportion of regulatory T cells among total Cd4+ T cells, as well as their expression levels of Foxp3, did not show any global change with time, we have identified 6 transcriptomically distinct clusters of regulatory T cells with cross-tissue conserved hallmarks of aging, including increased numbers of proinflammatory regulatory T cells, reduced precursor cells, increased immature and mature T follicular regulatory cells potentially supported by a metabolic switch from oxidative phosphorylation to glycolysis, a gradual loss of CD150hi regulatory T cells that support hematopoiesis, and increased adipose tissue-specific regulatory T cells that are associated with metabolic disease. To dissect the impact of immunosenescence on humoral immunity, we propose some potential mechanisms underlying T follicular regulatory cell-mediated dysfunction by interactome analysis on T follicular regulatory cells, T follicular helper cells, and B cells during aging. Lastly, spatiotemporal analysis further revealed trajectories of regulatory T-cell aging that demonstrate the most significant changes in marrow and adipose tissues that might contribute to the development of age-related immunosenescence and type 2 diabetes. Taken together, our findings could provide a better understanding of age-associated regulatory T-cell heterogeneity in lymphoid and adipose tissues, as well as regulatory T-cell hallmarks during progressive adaptation to aging that could be therapeutically targeted for rejuvenating the aging immune system in the future.


Asunto(s)
Diabetes Mellitus Tipo 2 , Linfocitos T Reguladores , Ratones , Animales , Linfocitos T Colaboradores-Inductores , Diabetes Mellitus Tipo 2/metabolismo , Envejecimiento/genética , Perfilación de la Expresión Génica
6.
Diabetes ; 71(5): 961-977, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35113157

RESUMEN

To date, identification of nonislet-specific transcriptional factors in the regulation of insulin gene expression has been little studied. Here, we report that the expression level of the transcription factor YY1 is increased dramatically in both human and mouse pancreatic ß-cells after birth. Nevertheless, the physiological role of YY1 during ß-cell development and its regulatory mechanism in ß-cell function remain largely unknown. After ß-cell ablation of Yy1, we observed rapid onset of hyperglycemia, impaired glucose tolerance, and reduced ß-cell mass in neonatal and adult mice. These mice also had hypoinsulinemia with normal insulin sensitivity compared with their wild-type littermates, manifesting as a type 1 diabetic phenotype. Mechanistically, genome-wide RNA sequencing has defined dysregulated insulin signaling and defective glucose responsiveness in ß-cells devoid of YY1. Integrative analyses coupled with chromatin immunoprecipitation assays targeting YY1, and histone modifications, including H3K4me1, H3K27ac, and H3K27me3, have further identified Ins1 and Ins2 as direct gene targets of YY1. Luciferase reporter assays and loss- and gain-of-function experiments also demonstrated that YY1 binds to the enhancer regions in exon 2 of Ins1 and Ins2, activating insulin transcription and, therefore, proinsulin and insulin production in pancreatic ß-cells. YY1 also directly interacts with RNA polymerase II, potentially stabilizing the enhancer-promoter interaction in the multiprotein-DNA complex during transcription initiation. Taken together, our findings suggest a role for YY1 as a transcriptional activator of insulin gene expression, assisting ß-cell maturation and function after birth. These analyses may advance our understanding of ß-cell biology and provide clinically relevant insights targeting the pathophysiological origins of diabetes.


Asunto(s)
Diabetes Mellitus , Células Secretoras de Insulina , Animales , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Homeostasis , Insulina/metabolismo , Insulina Regular Humana , Células Secretoras de Insulina/metabolismo , Ratones , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo
7.
Stem Cell Reports ; 17(3): 538-555, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35180397

RESUMEN

To date, the direct causative mechanism of SARS-CoV-2-induced endotheliitis remains unclear. Here, we report that human ECs barely express surface ACE2, and ECs express less intracellular ACE2 than non-ECs of the lungs. We ectopically expressed ACE2 in hESC-ECs to model SARS-CoV-2 infection. ACE2-deficient ECs are resistant to the infection but are more activated than ACE2-expressing ones. The virus directly induces endothelial activation by increasing monocyte adhesion, NO production, and enhanced phosphorylation of p38 mitogen-associated protein kinase (MAPK), NF-κB, and eNOS in ACE2-expressing and -deficient ECs. ACE2-deficient ECs respond to SARS-CoV-2 through TLR4 as treatment with its antagonist inhibits p38 MAPK/NF-κB/ interleukin-1ß (IL-1ß) activation after viral exposure. Genome-wide, single-cell RNA-seq analyses further confirm activation of the TLR4/MAPK14/RELA/IL-1ß axis in circulating ECs of mild and severe COVID-19 patients. Circulating ECs could serve as biomarkers for indicating patients with endotheliitis. Together, our findings support a direct role for SARS-CoV-2 in mediating endothelial inflammation in an ACE2-dependent or -independent manner.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Modelos Biológicos , SARS-CoV-2/fisiología , Receptor Toll-Like 4/metabolismo , Enzima Convertidora de Angiotensina 2/genética , COVID-19/patología , COVID-19/virología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , SARS-CoV-2/aislamiento & purificación , Índice de Severidad de la Enfermedad , Análisis de la Célula Individual , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
J Leukoc Biol ; 108(5): 1593-1602, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33070367

RESUMEN

Recently, immune cell-mediated tissue repair and regeneration has been an emerging paradigm of regenerative medicine. Immune cells form an essential part of the wound as induction of inflammation is a necessary step to elicit tissue healing. Rapid progress in transcriptomic analyses by high-throughput next-generation sequencing has been developed to study gene regulatory network and establish molecular signatures of immune cells that could potentially predict their functional roles in tissue repair and regeneration. However, the identification of cellular heterogeneity especially on the rare cell subsets has been limited in transcriptomic analyses of bulk cell populations. Therefore, genome-wide, single-cell RNA sequencing (scRNA-Seq) has offered an unprecedented approach to unravel cellular diversity and to study novel immune cell populations involved in tissue repair and regeneration through unsupervised sampling of individual cells without the need to rely on prior knowledge about cell-specific markers. The analysis of gene expression patterns at a single-cell resolution also holds promises to uncover the mechanisms and therefore the development of therapeutic strategy promoting immunoregenerative medicine. In this review, we will discuss how scRNA-Seq facilitates the characterization of immune cells, including macrophages, innate lymphoid cells and T and B lymphocytes, discovery of immune cell heterogeneity, identification of novel subsets, and tracking of developmental trajectories of distinct immune cells during tissue homeostasis, repair, and regeneration.


Asunto(s)
Linfocitos B/inmunología , Inmunidad Celular/inmunología , Inmunidad Innata , RNA-Seq , Regeneración/inmunología , Análisis de la Célula Individual , Linfocitos T/inmunología , Antígenos de Diferenciación/inmunología , Humanos
9.
Theranostics ; 10(20): 8924-8938, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32802172

RESUMEN

Accumulating evidence has demonstrated that immune cells play an important role in the regulation of tissue repair and regeneration. After injury, danger signals released by the damaged tissue trigger the initial pro-inflammatory phase essential for removing pathogens or cellular debris that is later replaced by the anti-inflammatory phase responsible for tissue healing. On the other hand, impaired immune regulation can lead to excessive scarring and fibrosis that could be detrimental for the restoration of organ function. Regulatory T-cells (Treg) have been revealed as the master regulator of the immune system that have both the immune and regenerative functions. In this review, we will summarize their immune role in the induction and maintenance of self-tolerance; as well as their regenerative role in directing tissue specific response for repair and regeneration. The latter is clearly demonstrated when Treg enhance the differentiation of stem or progenitor cells such as satellite cells to replace the damaged skeletal muscle, as well as the proliferation of parenchymal cells including neonatal cardiomyocytes for functional regeneration. Moreover, we will also discuss the reparative and regenerative role of Treg with a particular focus on blood vessels and cardiac tissues. Last but not least, we will describe the ongoing clinical trials with Treg in the treatment of autoimmune diseases that could give clinically relevant insights into the development of Treg therapy targeting tissue repair and regeneration.


Asunto(s)
Miocitos Cardíacos/inmunología , Regeneración/inmunología , Linfocitos T Reguladores/inmunología , Cicatrización de Heridas/inmunología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Humanos
10.
Theranostics ; 10(18): 8018-8035, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32724455

RESUMEN

Unlike adult cardiomyocytes, neonatal cardiomyocytes can readily proliferate that contributes to a transient regenerative potential after myocardial injury in mice. We have recently reported that CD4+ regulatory T-cells promote this process; however, the role of other CD4+ T-cell subsets as well as CD8+ T-cells in postnatal heart regeneration has been less studied. Methods: by comparing the regenerating postnatal day (P) 3 and the non-regenerating P8 heart after injury, we revealed the heterogeneity of CD4+ and CD8+ T-cells in the myocardium through single cell analysis. We also specifically ablated CD4+ and CD8+ T-cells using the lytic anti-CD4 and -CD8 monoclonal antibodies, respectively, in juvenile mice at P8 after myocardial injury. Results: we observe significantly more CD4+FOXP3- conventional T-cells in the P8 heart when compared to that of the P3 heart within a week after injury. Surprisingly, such a difference is not seen in CD8+ T-cells that appear to have no function as their depletion does not reactivate heart regeneration. On the other hand, specific ablation of CD4+ T-cells contributes to mitigated cardiac fibrosis and increased cardiomyocyte proliferation after injury in juvenile mice. Single-cell transcriptomic profiling reveals a pro-fibrotic CD4+ T-cell subset in the P8 but not P3 heart. Moreover, there are likely more Th1 and Th17 cells in the P8 than P3 heart. We further demonstrate that cytokines of Th1 and Th17 cells can directly reduce the proliferation and increase the apoptosis of neonatal cardiomyocytes. Moreover, ablation of CD4+ T-cells can directly or indirectly facilitate the polarization of macrophages away from the pro-fibrotic M2-like signature in the juvenile heart. Nevertheless, ablation of CD4+ T-cells alone does not offer the same protection in the adult heart after myocardial infarction, suggesting a developmental change of immune cells including CD4+ T-cells in the regulation of age-related mammalian heart repair. Conclusions: our results demonstrate that ablation of CD4+ but not CD8+ T-cells promotes heart regeneration in juvenile mice; and CD4+ T-cells play a distinct role in the regulation of heart regeneration and repair during development.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infarto del Miocardio/inmunología , Daño por Reperfusión Miocárdica/inmunología , Regeneración/inmunología , Subgrupos de Linfocitos T/inmunología , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD4/antagonistas & inhibidores , Antígenos CD8/antagonistas & inhibidores , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica/inmunología , Corazón/crecimiento & desarrollo , Humanos , Masculino , Ratones , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/citología , Miocardio/inmunología , Miocardio/patología , Miocitos Cardíacos/fisiología , Cultivo Primario de Células , RNA-Seq , Regeneración/efectos de los fármacos , Análisis de la Célula Individual , Subgrupos de Linfocitos T/efectos de los fármacos
11.
Stem Cell Reports ; 15(5): 1111-1126, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33096048

RESUMEN

To date, it remains unclear if there are specific cell-surface markers for purifying glucose-responsive pancreatic ß-like cells derived from human pluripotent stem cells (hPSCs). In searching for this, we generated an efficient protocol for differentiating ß-like cells from human embryonic stem cells. We performed single-cell RNA sequencing and found that CD9 is a negative cell-surface marker of ß-like cells, as most INS+ cells are CD9-. We purified ß-like cells for spontaneous formation of islet-like organoids against CD9, and found significantly more NKX6.1+MAFA+C-PEPTIDE+ ß-like cells in the CD9- than in the CD9+ population. CD9- cells also demonstrate better glucose responsiveness than CD9+ cells. In humans, we observe more CD9+C-PEPTIDE+ ß cells in the fetal than in the adult cadaveric islets and more Ki67+ proliferating cells among CD9+ fetal ß cells. Taken together, our experiments show that CD9 is a cell-surface marker for negative enrichment of glucose-responsive ß-like cells differentiated from hPSCs.


Asunto(s)
Células Madre Embrionarias Humanas/metabolismo , Células Secretoras de Insulina/metabolismo , Células Madre Pluripotentes/metabolismo , Tetraspanina 29/metabolismo , Biomarcadores/metabolismo , Péptido C/genética , Péptido C/metabolismo , Diferenciación Celular , Línea Celular , Células Cultivadas , Estudio de Asociación del Genoma Completo , Glucosa/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Madre Embrionarias Humanas/citología , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Organoides/metabolismo , RNA-Seq , Análisis de la Célula Individual , Tetraspanina 29/genética , Transcriptoma
12.
Theranostics ; 10(9): 4217-4232, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32226549

RESUMEN

In this study, we observe that the ischemic tissues of type-2 diabetic (T2D) patients and mice have significantly more CD8+ T-cells than that of their normoglycemic counterparts, respectively. However, the role of CD8+ T-cells in the pathogenesis of diabetic vascular complication has been less studied. Methods: We employed loss-of-function studies in mouse models using the non-lytic anti-CD8 antibody that blocks tissue infiltration of CD8+ T-cells into the injured tissue. We also performed genome-wide, single-cell RNA-sequencing of CD8+ T-cells to uncover their role in the pathogenesis of diabetic vascular diseases. Results: The vascular density is negatively correlated with the number of CD8+ T-cells in the ischemic tissues of patients and mice after injury. CD8+ T-cells or their supernatant can directly impair human and murine angiogenesis. Compared to normoglycemic mice that can regenerate their blood vessels after injury, T2D mice fail in this regeneration. Treatment with the CD8 checkpoint blocking antibody increases the proliferation and function of endothelial cells in both Leprdb/db mice and diet-induced diabetic Cdh5-Cre;Rosa-YFP lineage-tracing mice after ischemic injury. Furthermore, single-cell transcriptomic profiling reveals that CD8+ T-cells of T2D mice showed a de novo cell fate change from the angiogenic, tissue-resident memory cells towards the effector and effector memory cells after injury. Functional revascularization by CD8 checkpoint blockade is mediated through unleashing such a poised lineage commitment of CD8+ T-cells from T2D mice. Conclusion: Our results reveal that CD8+ T-cell plasticity regulates vascular regeneration; and give clinically relevant insights into the potential development of immunotherapy targeting vascular diseases associated with obesity and diabetes.


Asunto(s)
Linfocitos T CD8-positivos/citología , Enfermedad de la Arteria Coronaria/patología , Diabetes Mellitus Tipo 2/patología , Angiopatías Diabéticas/patología , Isquemia/patología , Enfermedades Vasculares Periféricas/patología , Animales , Plasticidad de la Célula , Células Cultivadas , Células Endoteliales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Regeneración
13.
Theranostics ; 9(15): 4324-4341, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31285764

RESUMEN

The neonatal mouse heart is capable of transiently regenerating after injury from postnatal day (P) 0-7 and macrophages are found important in this process. However, whether macrophages alone are sufficient to orchestrate this regeneration; what regulates cardiomyocyte proliferation; why cardiomyocytes do not proliferate after P7; and whether adaptive immune cells such as regulatory T-cells (Treg) influence neonatal heart regeneration have less studied. Methods: We employed both loss- and gain-of-function transgenic mouse models to study the role of Treg in neonatal heart regeneration. In loss-of-function studies, we treated mice with the lytic anti-CD25 antibody that specifically depletes Treg; or we treated FOXP3DTR with diphtheria toxin that specifically ablates Treg. In gain-of-function studies, we adoptively transferred hCD2+ Treg from NOD.Foxp3hCD2 to NOD/SCID that contain Treg as the only T-cell population. Furthermore, we performed single-cell RNA-sequencing of Treg to uncover paracrine factors essential for cardiomyocyte proliferation. Results: Unlike their wild type counterparts, NOD/SCID mice that are deficient in T-cells but harbor macrophages fail to regenerate their injured myocardium at as early as P3. During the first week of injury, Treg are recruited to the injured cardiac muscle but their depletion contributes to more severe cardiac fibrosis. On the other hand, adoptive transfer of Treg results in mitigated fibrosis and enhanced proliferation and function of the injured cardiac muscle. Mechanistically, single-cell transcriptomic profiling reveals that Treg could be a source of regenerative factors. Treg directly promote proliferation of both mouse and human cardiomyocytes in a paracrine manner; and their secreted factors such as CCL24, GAS6 or AREG potentiate neonatal cardiomyocyte proliferation. By comparing the regenerating P3 and non-regenerating P8 heart, there is a significant increase in the absolute number of intracardiac Treg but the whole transcriptomes of these Treg do not differ regardless of whether the neonatal heart regenerates. Furthermore, even adult Treg, given sufficient quantity, possess the same regenerative capability. Conclusion: Our results demonstrate a regenerative role of Treg in neonatal heart regeneration. Treg can directly facilitate cardiomyocyte proliferation in a paracrine manner.


Asunto(s)
Corazón/fisiología , Miocitos Cardíacos/citología , Comunicación Paracrina , Regeneración/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Proliferación Celular , Fibrosis , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunidad Innata , Mutación con Pérdida de Función/genética , Macrófagos/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Transcriptoma/genética , Regulación hacia Arriba/genética
14.
Genome Med ; 10(1): 71, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30236153

RESUMEN

BACKGROUND: We have previously reported an antigen-specific protocol to induce transplant tolerance and linked suppression to human embryonic stem cell (hESC)-derived tissues in immunocompetent mice through coreceptor and costimulation blockade. However, the exact mechanisms of acquired immune tolerance in this model have remained unclear. METHODS: We utilize the NOD.Foxp3hCD2 reporter mouse line and an ablative anti-hCD2 antibody to ask if CD4+FOXP3+ regulatory T cells (Treg) are required for coreceptor and costimulation blockade-induced immune tolerance. We also perform genome-wide single-cell RNA-sequencing to interrogate Treg during immune rejection and tolerance and to indicate possible mechanisms involved in sustaining Treg function. RESULTS: We show that Treg are indispensable for tolerance induced by coreceptor and costimulation blockade as depletion of which with an anti-hCD2 antibody resulted in rejection of hESC-derived pancreatic islets in NOD.Foxp3hCD2 mice. Single-cell transcriptomic profiling of 12,964 intragraft CD4+ T cells derived from rejecting and tolerated grafts reveals that Treg are heterogeneous and functionally distinct in the two outcomes of transplant rejection and tolerance. Treg appear to mainly promote chemotactic and ubiquitin-dependent protein catabolism during transplant rejection while seeming to harness proliferative and immunosuppressive function during tolerance. We also demonstrate that this form of acquired transplant tolerance is associated with increased proliferation and PD-1 expression by Treg. Blocking PD-1 signaling with a neutralizing anti-PD-1 antibody leads to reduced Treg proliferation and graft rejection. CONCLUSIONS: Our results suggest that short-term coreceptor and costimulation blockade mediates immune tolerance to hESC-derived pancreatic islets by promoting Treg proliferation through engagement of PD-1. Our findings could give new insights into clinical development of hESC-derived pancreatic tissues, combined with immunotherapies that expand intragraft Treg, as a potentially sustainable alternative treatment for T1D.


Asunto(s)
Perfilación de la Expresión Génica , Tolerancia Inmunológica/genética , Receptor de Muerte Celular Programada 1/metabolismo , Análisis de la Célula Individual , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Recuento de Células , Línea Celular , Proliferación Celular/genética , Supervivencia Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Genoma , Rechazo de Injerto/inmunología , Humanos , Islotes Pancreáticos/citología , Trasplante de Islotes Pancreáticos , Ratones Endogámicos C57BL , Transducción de Señal , Bazo/citología
15.
Cell Rep ; 24(6): 1610-1626, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30089270

RESUMEN

The role of CD4+ T cells in the ischemic tissues of T2D patients remains unclear. Here, we report that T2D patients' vascular density was negatively correlated with the number of infiltrating CD4+ T cells after ischemic injury. Th1 was the predominant subset, and Th1-derived IFN-γ and TNF-α directly impaired human angiogenesis. We then blocked CD4+ T cell infiltration into the ischemic tissues of both Leprdb/db and diet-induced obese T2D mice. Genome-wide RNA sequencing shows an increased proliferative and angiogenic capability of diabetic ECs in ischemic tissues. Moreover, wire myography shows enhanced EC function and laser Doppler imaging reveals improved post-ischemic blood reperfusion. Mechanistically, functional revascularization after CD4 coreceptor blockade was mediated by Tregs. Genetic lineage tracing via Cdh5-CreER and Apln-CreER and coculture assays further illustrate that Tregs increased vascular density and induced de novo sprouting angiogenesis in a paracrine manner. Taken together, our results reveal that Th1 impaired while Tregs promoted functional post-ischemic revascularization in obesity and diabetes.


Asunto(s)
Apelina/metabolismo , Diabetes Mellitus Tipo 2/genética , Linfocitos T Reguladores/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Ratones , Neovascularización Patológica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA