Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Life Sci Alliance ; 7(7)2024 Jul.
Article En | MEDLINE | ID: mdl-38740432

Subclinical vascular impairment can be exacerbated in individuals who experience sustained inflammation after COVID-19 infection. Our study explores the prevalence and impact of autoantibodies on vascular dysfunction in healthy COVID-19 survivors, an area that remains inadequately investigated. Focusing on autoantibodies against the atypical chemokine receptor 1 (ACKR1), COVID-19 survivors demonstrated significantly elevated anti-ACKR1 autoantibodies, correlating with systemic cytokines, circulating damaged endothelial cells, and endothelial dysfunction. An independent cohort linked these autoantibodies to increased vascular disease outcomes during a median 6.7-yr follow-up. We analyzed a single-cell transcriptome atlas of endothelial cells from diverse mouse tissues, identifying enriched Ackr1 expressions in venous regions of the brain and soleus muscle vasculatures, which holds intriguing implications for tissue-specific venous thromboembolism manifestations reported in COVID-19. Functionally, purified immunoglobulin G (IgG) extracted from patient plasma did not trigger cell apoptosis or increase barrier permeability in human vein endothelial cells. Instead, plasma IgG enhanced antibody-dependent cellular cytotoxicity mediated by patient PBMCs, a phenomenon alleviated by blocking peptide or liposome ACKR1 recombinant protein. The blocking peptide uncovered that purified IgG from COVID-19 survivors possessed potential epitopes in the N-terminal extracellular domain of ACKR1, which effectively averted antibody-dependent cellular cytotoxicity. Our findings offer insights into therapeutic development to mitigate autoantibody reactivity in blood vessels in chronic inflammation.


Autoantibodies , COVID-19 , SARS-CoV-2 , Humans , Autoantibodies/immunology , COVID-19/immunology , Animals , Mice , Female , Male , SARS-CoV-2/immunology , Inflammation/immunology , Middle Aged , Endothelium, Vascular/metabolism , Endothelium, Vascular/immunology , Immunoglobulin G/immunology , Immunoglobulin G/blood , Endothelial Cells/metabolism , Endothelial Cells/immunology , Adult , Aged
2.
Dev Cell ; 59(9): 1110-1131.e22, 2024 May 06.
Article En | MEDLINE | ID: mdl-38569552

The developmental origin of blood-forming hematopoietic stem cells (HSCs) is a longstanding question. Here, our non-invasive genetic lineage tracing in mouse embryos pinpoints that artery endothelial cells generate HSCs. Arteries are transiently competent to generate HSCs for 2.5 days (∼E8.5-E11) but subsequently cease, delimiting a narrow time frame for HSC formation in vivo. Guided by the arterial origins of blood, we efficiently and rapidly differentiate human pluripotent stem cells (hPSCs) into posterior primitive streak, lateral mesoderm, artery endothelium, hemogenic endothelium, and >90% pure hematopoietic progenitors within 10 days. hPSC-derived hematopoietic progenitors generate T, B, NK, erythroid, and myeloid cells in vitro and, critically, express hallmark HSC transcription factors HLF and HOXA5-HOXA10, which were previously challenging to upregulate. We differentiated hPSCs into highly enriched HLF+ HOXA+ hematopoietic progenitors with near-stoichiometric efficiency by blocking formation of unwanted lineages at each differentiation step. hPSC-derived HLF+ HOXA+ hematopoietic progenitors could avail both basic research and cellular therapies.


Cell Differentiation , Cell Lineage , Hematopoietic Stem Cells , Homeodomain Proteins , Pluripotent Stem Cells , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Humans , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Animals , Mice , Endothelial Cells/metabolism , Endothelial Cells/cytology , Transcription Factors/metabolism , Transcription Factors/genetics , Hematopoiesis
3.
Semin Cell Dev Biol ; 155(Pt C): 62-75, 2024 03 01.
Article En | MEDLINE | ID: mdl-37393122

Owing to their manifold roles in health and disease, there have been intense efforts to synthetically generate blood vessels in vitro from human pluripotent stem cells (hPSCs). However, there are multiple types of blood vessel, including arteries and veins, which are molecularly and functionally different. How can we specifically generate either arterial or venous endothelial cells (ECs) from hPSCs in vitro? Here, we summarize how arterial or venous ECs arise during embryonic development. VEGF and NOTCH arbitrate the bifurcation of arterial vs. venous ECs in vivo. While manipulating these two signaling pathways biases hPSC differentiation towards arterial and venous identities, efficiently generating these two subtypes of ECs has remained challenging until recently. Numerous questions remain to be fully addressed. What is the complete identity, timing and combination of extracellular signals that specify arterial vs. venous identities? How do these extracellular signals intersect with fluid flow to modulate arteriovenous fate? What is a unified definition for endothelial progenitors or angioblasts, and when do arterial vs. venous potentials segregate? How can we regulate hPSC-derived arterial and venous ECs in vitro, and generate organ-specific ECs? In turn, answers to these questions could avail the production of arterial and venous ECs from hPSCs, accelerating vascular research, tissue engineering, and regenerative medicine.


Endothelial Cells , Pluripotent Stem Cells , Humans , Endothelial Cells/metabolism , Pluripotent Stem Cells/metabolism , Cell Differentiation/physiology , Arteries/metabolism
4.
Nat Commun ; 14(1): 5690, 2023 09 14.
Article En | MEDLINE | ID: mdl-37709760

Generating primordial germ cell-like cells (PGCLCs) from human pluripotent stem cells (hPSCs) advances studies of human reproduction and development of infertility treatments, but often entails complex 3D aggregates. Here we develop a simplified, monolayer method to differentiate hPSCs into PGCs within 3.5 days. We use our simplified differentiation platform and single-cell RNA-sequencing to achieve further insights into PGCLC specification. Transient WNT activation for 12 h followed by WNT inhibition specified PGCLCs; by contrast, sustained WNT induced primitive streak. Thus, somatic cells (primitive streak) and PGCLCs are related-yet distinct-lineages segregated by temporally-dynamic signaling. Pluripotency factors including NANOG are continuously expressed during the transition from pluripotency to posterior epiblast to PGCs, thus bridging pluripotent and germline states. Finally, hPSC-derived PGCLCs can be easily purified by virtue of their CXCR4+PDGFRA-GARP- surface-marker profile and single-cell RNA-sequencing reveals that they harbor transcriptional similarities with fetal PGCs.


Embryonic Development , Germ Cells , Humans , Cell Differentiation , Fetus , RNA
5.
Am J Hum Genet ; 109(9): 1680-1691, 2022 09 01.
Article En | MEDLINE | ID: mdl-36007525

Neisseria meningitidis protects itself from complement-mediated killing by binding complement factor H (FH). Previous studies associated susceptibility to meningococcal disease (MD) with variation in CFH, but the causal variants and underlying mechanism remained unknown. Here we attempted to define the association more accurately by sequencing the CFH-CFHR locus and imputing missing genotypes in previously obtained GWAS datasets of MD-affected individuals of European ancestry and matched controls. We identified a CFHR3 SNP that provides protection from MD (rs75703017, p value = 1.1 × 10-16) by decreasing the concentration of FH in the blood (p value = 1.4 × 10-11). We subsequently used dual-luciferase studies and CRISPR gene editing to establish that deletion of rs75703017 increased FH expression in hepatocyte by preventing promotor inhibition. Our data suggest that reduced concentrations of FH in the blood confer protection from MD; with reduced access to FH, N. meningitidis is less able to shield itself from complement-mediated killing.


Complement Factor H , Meningococcal Infections , Blood Proteins/genetics , Complement Factor H/genetics , Complement System Proteins/genetics , Genetic Predisposition to Disease , Genotype , Humans , Meningococcal Infections/genetics
6.
Cell ; 185(14): 2523-2541.e30, 2022 07 07.
Article En | MEDLINE | ID: mdl-35738284

Stem cell research endeavors to generate specific subtypes of classically defined "cell types." Here, we generate >90% pure human artery or vein endothelial cells from pluripotent stem cells within 3-4 days. We specified artery cells by inhibiting vein-specifying signals and vice versa. These cells modeled viral infection of human vasculature by Nipah and Hendra viruses, which are extraordinarily deadly (∼57%-59% fatality rate) and require biosafety-level-4 containment. Generating pure populations of artery and vein cells highlighted that Nipah and Hendra viruses preferentially infected arteries; arteries expressed higher levels of their viral-entry receptor. Virally infected artery cells fused into syncytia containing up to 23 nuclei, which rapidly died. Despite infecting arteries and occupying ∼6%-17% of their transcriptome, Nipah and Hendra largely eluded innate immune detection, minimally eliciting interferon signaling. We thus efficiently generate artery and vein cells, introduce stem-cell-based toolkits for biosafety-level-4 virology, and explore the arterial tropism and cellular effects of Nipah and Hendra viruses.


Hendra Virus , Nipah Virus , Pluripotent Stem Cells , Arteries , Endothelial Cells , Hendra Virus/genetics , Humans , Tropism
8.
Cell Mol Gastroenterol Hepatol ; 11(1): 273-290, 2021.
Article En | MEDLINE | ID: mdl-32992051

Epithelial cells in the liver (known as hepatocytes) are high-performance engines of myriad metabolic functions and versatile responders to liver injury. As hepatocytes metabolize amino acids, alcohol, drugs, and other substrates, they produce and are exposed to a milieu of toxins and harmful byproducts that can damage themselves. In the healthy liver, hepatocytes generally divide slowly. However, after liver injury, hepatocytes can ramp up proliferation to regenerate the liver. Yet, on extensive injury, regeneration falters, and liver failure ensues. It is therefore critical to understand the mechanisms underlying liver regeneration and, in particular, which liver cells are mobilized during liver maintenance and repair. Controversies continue to surround the very existence of hepatic stem cells and, if they exist, their spatial location, multipotency, degree of contribution to regeneration, ploidy, and susceptibility to tumorigenesis. This review discusses these controversies. Finally, we highlight how insights into hepatocyte regeneration and biology in vivo can inform in vitro studies to propagate primary hepatocytes with liver regeneration-associated signals and to generate hepatocytes de novo from pluripotent stem cells.


Hepatocytes/physiology , Induced Pluripotent Stem Cells/physiology , Liver Regeneration , Liver/cytology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Liver/physiology
9.
Wiley Interdiscip Rev Dev Biol ; 9(3): e368, 2020 05.
Article En | MEDLINE | ID: mdl-31746148

Too many choices can be problematic. This is certainly the case for human pluripotent stem cells (hPSCs): they harbor the potential to differentiate into hundreds of cell types; yet it is highly challenging to exclusively differentiate hPSCs into a single desired cell type. This review focuses on unresolved and fundamental questions regarding hPSC differentiation and critiquing the identity and purity of the resultant cell populations. These are timely issues in view of the fact that hPSC-derived cell populations have or are being transplanted into patients in over 30 ongoing clinical trials. While many in vitro differentiation protocols purport to "mimic development," the exact number and identity of intermediate steps that a pluripotent cell takes to differentiate into a given cell type in vivo remains largely unknown. Consequently, most differentiation efforts inevitably generate a heterogeneous cellular population, as revealed by single-cell RNA-sequencing and other analyses. The presence of unwanted cell types in differentiated hPSC populations does not portend well for transplantation therapies. This provides an impetus to precisely control differentiation to desired ends-for instance, by logically blocking the formation of unwanted cell types or by overexpressing lineage-specifying transcription factors-or by harnessing technologies to selectively purify desired cell types. Conversely, approaches to differentiate three-dimensional "organoids" from hPSCs intentionally generate heterogeneous cell populations. While this is intended to mimic the rich cellular diversity of developing tissues, whether all such organoids are spatially organized in a manner akin to native organs (and thus, whether they fully qualify as organoids) remains to be fully resolved. This article is categorized under: Adult Stem Cells > Tissue Renewal > Regeneration: Stem Cell Differentiation and Reversion Gene Expression > Transcriptional Hierarchies: Cellular Differentiation Early Embryonic Development: Gastrulation and Neurulation.


Cellular Reprogramming Techniques/methods , Organoids/cytology , Pluripotent Stem Cells/cytology , Cell Differentiation , Humans , Organoids/metabolism , Pluripotent Stem Cells/classification , Pluripotent Stem Cells/metabolism , Primary Cell Culture/methods
10.
J Vis Exp ; (148)2019 06 11.
Article En | MEDLINE | ID: mdl-31259908

The liver detoxifies harmful substances, secretes vital proteins, and executes key metabolic activities, thus sustaining life. Consequently, liver failure-which can be caused by chronic alcohol intake, hepatitis, acute poisoning, or other insults-is a severe condition that can culminate in bleeding, jaundice, coma, and eventually death. However, approaches to treat liver failure, as well as studies of liver function and disease, have been stymied in part by the lack of a plentiful supply of human liver cells. To this end, this protocol details the efficient differentiation of human pluripotent stem cells (hPSCs) into hepatocyte-like cells, guided by a developmental roadmap that describes how liver fate is specified across six consecutive differentiation steps. By manipulating developmental signaling pathways to promote liver differentiation and to explicitly suppress the formation of unwanted cell fates, this method efficiently generates populations of human liver bud progenitors and hepatocyte-like cells by days 6 and 18 of PSC differentiation, respectively. This is achieved through the temporally-precise control of developmental signaling pathways, exerted by small molecules and growth factors in a serum-free culture medium. Differentiation in this system occurs in monolayers and yields hepatocyte-like cells that express characteristic hepatocyte enzymes and have the ability to engraft a mouse model of chronic liver failure. The ability to efficiently generate large numbers of human liver cells in vitro has ramifications for treatment of liver failure, for drug screening, and for mechanistic studies of liver disease.


Cell Differentiation , Hepatocytes/cytology , Pluripotent Stem Cells/cytology , Animals , Culture Media , Humans , Mice , Signal Transduction
11.
Cell Rep ; 22(8): 2190-2205, 2018 02 20.
Article En | MEDLINE | ID: mdl-29466743

How are closely related lineages, including liver, pancreas, and intestines, diversified from a common endodermal origin? Here, we apply principles learned from developmental biology to rapidly reconstitute liver progenitors from human pluripotent stem cells (hPSCs). Mapping the formation of multiple endodermal lineages revealed how alternate endodermal fates (e.g., pancreas and intestines) are restricted during liver commitment. Human liver fate was encoded by combinations of inductive and repressive extracellular signals at different doses. However, these signaling combinations were temporally re-interpreted: cellular competence to respond to retinoid, WNT, TGF-ß, and other signals sharply changed within 24 hr. Consequently, temporally dynamic manipulation of extracellular signals was imperative to suppress the production of unwanted cell fates across six consecutive developmental junctures. This efficiently generated 94.1% ± 7.35% TBX3+HNF4A+ human liver bud progenitors and 81.5% ± 3.2% FAH+ hepatocyte-like cells by days 6 and 18 of hPSC differentiation, respectively; the latter improved short-term survival in the Fah-/-Rag2-/-Il2rg-/- mouse model of liver failure.


Cell Differentiation , Liver/cytology , Pluripotent Stem Cells/cytology , Animals , Animals, Newborn , Biliary Tract/cytology , Biomarkers/metabolism , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Endoderm/cytology , Fibroblast Growth Factors/pharmacology , Gastrointestinal Tract/cytology , Hepatocytes/cytology , Hepatocytes/drug effects , High-Throughput Screening Assays , Humans , Liver/injuries , Liver/pathology , Mice , Pluripotent Stem Cells/drug effects , Signal Transduction , Tretinoin/pharmacology , Wnt Signaling Pathway/drug effects
12.
Nat Methods ; 14(12): 1205-1212, 2017 Dec.
Article En | MEDLINE | ID: mdl-29106405

Multiple adult tissues are maintained by stem cells of restricted developmental potential which can only form a subset of lineages within the tissue. For instance, the two adult lung epithelial compartments (airways and alveoli) are separately maintained by distinct lineage-restricted stem cells. A challenge has been to obtain multipotent stem cells and/or progenitors that can generate all epithelial cell types of a given tissue. Here we show that mouse Sox9+ multipotent embryonic lung progenitors can be isolated and expanded long term in 3D culture. Cultured Sox9+ progenitors transcriptionally resemble their in vivo counterparts and generate both airway and alveolar cell types in vitro. Sox9+ progenitors that were transplanted into injured adult mouse lungs differentiated into all major airway and alveolar lineages in vivo in a region-appropriate fashion. We propose that a single expandable embryonic lung progenitor population with broader developmental competence may eventually be used as an alternative for region-restricted adult tissue stem cells in regenerative medicine.


Lung/cytology , Multipotent Stem Cells/cytology , SOX9 Transcription Factor/genetics , Animals , Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Knock-In Techniques , Lung/embryology , Lung/growth & development , Lung/metabolism , Mice, Transgenic , Multipotent Stem Cells/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , SOX9 Transcription Factor/metabolism , Tissue Engineering
13.
Differentiation ; 98: 25-34, 2017.
Article En | MEDLINE | ID: mdl-29078082

Liver diseases afflict millions of patients worldwide. Currently, the only long-term treatment for liver failure is the transplantation of a new liver. However, intravenously transplanting a suspension of human hepatocytes might be a less-invasive approach to partially reconstitute lost liver functions in human patients as evinced by promising outcomes in clinical trials. The purpose of this essay is to emphasize outstanding questions that continue to surround hepatocyte transplantation. While adult primary human hepatocytes are the gold standard for transplantation, hepatocytes are heterogeneous. Whether all hepatocytes engraft equally and what specifically defines an "engraftable" hepatocyte capable of long-term liver reconstitution remains unclear. To this end, mouse models of liver injury enable the evaluation of human hepatocytes and their behavior upon transplantation into a complex injured liver environment. While mouse models may not be fully representative of the injured human liver and human hepatocytes tend to engraft mice less efficiently than mouse hepatocytes, valuable lessons have nonetheless been learned from transplanting human hepatocytes into mouse models. With an eye to the future, it will be crucial to eventually detail the optimal biological source (whether in vivo- or in vitro-derived) and presumptive heterogeneity of human hepatocytes and to understand the mechanisms through which they engraft and regenerate liver tissue in vivo.


Cell Differentiation/physiology , Hepatocytes/cytology , Liver/cytology , Regeneration/physiology , Animals , Cell Proliferation/physiology , Disease Models, Animal , Humans
14.
Sci Data ; 3: 160109, 2016 12 20.
Article En | MEDLINE | ID: mdl-27996962

Mesoderm is the developmental precursor to myriad human tissues including bone, heart, and skeletal muscle. Unravelling the molecular events through which these lineages become diversified from one another is integral to developmental biology and understanding changes in cellular fate. To this end, we developed an in vitro system to differentiate human pluripotent stem cells through primitive streak intermediates into paraxial mesoderm and its derivatives (somites, sclerotome, dermomyotome) and separately, into lateral mesoderm and its derivatives (cardiac mesoderm). Whole-population and single-cell analyses of these purified populations of human mesoderm lineages through RNA-seq, ATAC-seq, and high-throughput surface marker screens illustrated how transcriptional changes co-occur with changes in open chromatin and surface marker landscapes throughout human mesoderm development. This molecular atlas will facilitate study of human mesoderm development (which cannot be interrogated in vivo due to restrictions on human embryo studies) and provides a broad resource for the study of gene regulation in development at the single-cell level, knowledge that might one day be exploited for regenerative medicine.


Chromatin , Mesoderm/physiology , Pluripotent Stem Cells , Transcription, Genetic , Biomarkers , Cell Differentiation , Humans , Mesoderm/cytology , Mesoderm/embryology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology
15.
Cell ; 166(2): 451-467, 2016 Jul 14.
Article En | MEDLINE | ID: mdl-27419872

Stem-cell differentiation to desired lineages requires navigating alternating developmental paths that often lead to unwanted cell types. Hence, comprehensive developmental roadmaps are crucial to channel stem-cell differentiation toward desired fates. To this end, here, we map bifurcating lineage choices leading from pluripotency to 12 human mesodermal lineages, including bone, muscle, and heart. We defined the extrinsic signals controlling each binary lineage decision, enabling us to logically block differentiation toward unwanted fates and rapidly steer pluripotent stem cells toward 80%-99% pure human mesodermal lineages at most branchpoints. This strategy enabled the generation of human bone and heart progenitors that could engraft in respective in vivo models. Mapping stepwise chromatin and single-cell gene expression changes in mesoderm development uncovered somite segmentation, a previously unobservable human embryonic event transiently marked by HOPX expression. Collectively, this roadmap enables navigation of mesodermal development to produce transplantable human tissue progenitors and uncover developmental processes. VIDEO ABSTRACT.


Mesoderm/cytology , Signal Transduction , Bone Morphogenetic Proteins/metabolism , Bone and Bones/cytology , Bone and Bones/metabolism , Heart/growth & development , Homeodomain Proteins/metabolism , Humans , Mesoderm/metabolism , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism , Primitive Streak/cytology , Primitive Streak/metabolism , Single-Cell Analysis , Somites/metabolism , Stem Cells , Tumor Suppressor Proteins/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism
16.
Physiol Rev ; 95(1): 245-95, 2015 Jan.
Article En | MEDLINE | ID: mdl-25540144

Pluripotent cells in embryos are situated near the apex of the hierarchy of developmental potential. They are capable of generating all cell types of the mammalian body proper. Therefore, they are the exemplar of stem cells. In vivo, pluripotent cells exist transiently and become expended within a few days of their establishment. Yet, when explanted into artificial culture conditions, they can be indefinitely propagated in vitro as pluripotent stem cell lines. A host of transcription factors and regulatory genes are now known to underpin the pluripotent state. Nonetheless, how pluripotent cells are equipped with their vast multilineage differentiation potential remains elusive. Consensus holds that pluripotency transcription factors prevent differentiation by inhibiting the expression of differentiation genes. However, this does not explain the developmental potential of pluripotent cells. We have presented another emergent perspective, namely, that pluripotency factors function as lineage specifiers that enable pluripotent cells to differentiate into specific lineages, therefore endowing pluripotent cells with their multilineage potential. Here we provide a comprehensive overview of the developmental biology, transcription factors, and extrinsic signaling associated with pluripotent cells, and their accompanying subtypes, in vitro heterogeneity and chromatin states. Although much has been learned since the appreciation of mammalian pluripotency in the 1950s and the derivation of embryonic stem cell lines in 1981, we will specifically emphasize what currently remains unclear. However, the view that pluripotency factors capacitate differentiation, recently corroborated by experimental evidence, might perhaps address the long-standing question of how pluripotent cells are endowed with their multilineage differentiation potential.


Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Mammals/embryology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Animals , Embryonic Development , Humans
17.
Immunobiology ; 220(5): 614-23, 2015 May.
Article En | MEDLINE | ID: mdl-25533809

The role of Annexin A1 (ANXA1) in counter-regulating the activities of innate immune cells, such as the migration of neutrophils and monocytes, and the generation of pro-inflammatory mediators in various models of inflammatory and autoimmune diseases is well documented. However, while ANXA1 has been proposed as an important mediator of the adaptive immune response, its involvement in this respect has been less studied. Furthermore, while there have been numerous studies on the role of ANXA1 in inflammatory diseases, less has been reported on its influence in immunity against infection. A recent study reported a link between ANXA1 and tuberculosis, and proposed a model in which Mycobacterium tuberculosis exerts its virulence by manipulating the ANXA1-mediated host apoptotic response. This has prompted us to further investigate the role of ANXA1 in the pathogenesis of tuberculosis in vivo. Here, we show that ANXA1(-/-) mice are more susceptible to M. tuberculosis infection, as evidenced by a transient increase in the pulmonary bacterial burden, and exacerbated and disorganized granulomatous inflammation. These pathological manifestations correlated with an impaired ability of ANXA1(-/-) dendritic cells to activate naïve T cells, thereby supporting a role for ANXA1 in shaping the adaptive immunity against M. tuberculosis.


Annexin A1/metabolism , Dendritic Cells/immunology , Granuloma/immunology , Lung/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Tuberculosis, Pulmonary/immunology , Adaptive Immunity , Animals , Annexin A1/genetics , Cells, Cultured , Dendritic Cells/microbiology , Humans , Lung/microbiology , Lung/pathology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Knockout
18.
Cell Stem Cell ; 14(2): 237-52, 2014 Feb 06.
Article En | MEDLINE | ID: mdl-24412311

Human pluripotent stem cell (hPSC) differentiation typically yields heterogeneous populations. Knowledge of signals controlling embryonic lineage bifurcations could efficiently yield desired cell types through exclusion of alternate fates. Therefore, we revisited signals driving induction and anterior-posterior patterning of definitive endoderm to generate a coherent roadmap for endoderm differentiation. With striking temporal dynamics, BMP and Wnt initially specified anterior primitive streak (progenitor to endoderm), yet, 24 hr later, suppressed endoderm and induced mesoderm. At lineage bifurcations, cross-repressive signals separated mutually exclusive fates; TGF-ß and BMP/MAPK respectively induced pancreas versus liver from endoderm by suppressing the alternate lineage. We systematically blockaded alternate fates throughout multiple consecutive bifurcations, thereby efficiently differentiating multiple hPSC lines exclusively into endoderm and its derivatives. Comprehensive transcriptional and chromatin mapping of highly pure endodermal populations revealed that endodermal enhancers existed in a surprising diversity of "pre-enhancer" states before activation, reflecting the establishment of a permissive chromatin landscape as a prelude to differentiation.


Cell Lineage , Endoderm/embryology , Pluripotent Stem Cells/cytology , Signal Transduction , Animals , Base Sequence , Body Patterning/drug effects , Bone Morphogenetic Proteins/metabolism , Cell Lineage/drug effects , Chromatin/metabolism , Culture Media, Serum-Free/pharmacology , Digestive System/embryology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Endoderm/drug effects , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic/drug effects , Fibroblast Growth Factors/metabolism , Humans , Liver/embryology , MAP Kinase Signaling System/drug effects , Mice , Molecular Sequence Data , Pancreas/embryology , Pluripotent Stem Cells/drug effects , Primitive Streak/cytology , Primitive Streak/embryology , Protein Binding/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism
19.
Eur J Pharmacol ; 683(1-3): 310-5, 2012 May 15.
Article En | MEDLINE | ID: mdl-22387854

The high frequency of rhinovirus (RV) infection and the lack of an effective treatment, underline the importance of research on novel anti-rhinoviral agents. The present study investigated the effects of des-aspartate-angiotensin I (DAA-I) on the survival of RV14-infected H1HeLa cells; and the early inflammatory processes in RV14-infected A549 lung epithelial cells. The study rationale was based on earlier findings showing that DAA-I is an effective anti-inflammatory agent, and that symptoms and severity of rhinoviral infection are related to the underling inflammation. RV14 concentration dependently caused the death of H1HeLa cells and DAA-I, at concentrations of 10⁻¹° to 10⁻¹² M, attenuated the lethal action of RV14 indicating that that DAA-I exerts antiviral action. Unlike its action on H1HeLa cells, RV14 did not cause apparent cytopathic effect on A549 cells, and these cells were used to study the antiviral action of DAA-I. RV14 induced overexpression of ICAM-1, E-selectin and overproduction of superoxide in A549 cells, and DAA-I attenuated the three increases to basal level at concentrations of 10⁻¹° to 10⁻¹² M. Losartan, an angiotensin AT1 receptor antagonist, blocked the inhibitory action of DAA-I on superoxide overproduction indicating that the AT1 receptor mediates the action of DAA-I. The present data represent a novel demonstration of the antiviral action of an angiotensin peptide, and a possible involvement of the renin angiotensin system in viral infection. Indeed the angiotensin AT1 receptor has been reported to be obligatory for the development of virus-induced myocardial injury through the proinflammatory action of angiotensin II via the NF-κB/cytokine pathway.


Angiotensin I/analogs & derivatives , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antiviral Agents/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Respiratory Mucosa/drug effects , Rhinovirus/drug effects , Angiotensin I/adverse effects , Angiotensin I/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Antiviral Agents/adverse effects , Cell Line , Cell Survival/drug effects , Cytopathogenic Effect, Viral/drug effects , E-Selectin/metabolism , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/virology , HeLa Cells , Humans , Microbial Viability/drug effects , Osmolar Concentration , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Rhinovirus/immunology , Rhinovirus/pathogenicity , Superoxides/metabolism
20.
EMBO Mol Med ; 4(1): 27-37, 2012 Jan.
Article En | MEDLINE | ID: mdl-22147526

Mycolic acids are attractive diagnostic markers for tuberculosis (TB) infection because they are bacteria-derived, contain information about bacterial species, modulate host-pathogen interactions and are chemically inert. Here, we present a novel approach based on mass spectrometry. Quantification of specific precursor → fragment transitions of approximately 2000 individual mycolic acids (MAs) resulted in high analytical sensitivity and specificity. We next used this tool in a retrospective case-control study of patients with pulmonary TB with varying disease burdens from South Korea, Vietnam, Uganda and South Africa. MAs were extracted from small volume sputum (200 µl) and analysed without the requirement for derivatization. Infected patients (70, 19 of whom were HIV+) could be separated from controls (40, 20 of whom were HIV+) with a sensitivity and specificity of 94 and 93%, respectively. Furthermore, we quantified MA species in lung tissue of TB-infected mice and demonstrated effective clearance of MA levels following curative rifampicin treatment. Thus, our results demonstrate for the first time the feasibility and clinical relevance of direct detection of mycobacterial lipids as biomarkers of TB infection.


Antibiotics, Antitubercular/therapeutic use , Mycolic Acids/analysis , Rifampin/therapeutic use , Tuberculosis/diagnosis , Tuberculosis/drug therapy , Animals , Biomarkers/analysis , Case-Control Studies , Chromatography, High Pressure Liquid , Female , HIV Infections/complications , HIV Infections/pathology , Humans , Mice , Mice, Inbred BALB C , Retrospective Studies , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization , Sputum/metabolism , Tuberculosis/complications
...