Nagaoka M et al. (JAN 2010)
BMC developmental biology 10 60
Culture of human pluripotent stem cells using completely defined conditions on a recombinant E-cadherin substratum.
BACKGROUND: To maintain pluripotency of human embryonic stem (huES) cells in feeder-free culture it has been necessary to provide a Matrigel substratum,which is a complex of poorly defined extracellular matrices and growth factors derived from mouse Engelbreth-Holm-Swarm sarcoma cells. Culture of stem cells under ill-defined conditions can inhibit the effectiveness of maintaining cells in a pluripotent state and reduce reproducibility of differentiation protocols. Moreover recent batches of Matrigel have been found to be contaminated with the single stranded RNA virus,Lactate Dehydrogenase Elevating Virus (LDEV),raising concerns regarding the safety of using stem cells that have been cultured on Matrigel in a therapeutic setting. To circumvent such concerns,we attempted to identify a recombinant matrix that could be used as an alternative to Matrigel for the culture of human pluripotent stem cells. huES and human induced pluripotent stem (hiPS) cells were grown on plates coated with a fusion protein consisting of E-cadherin and the IgG Fc domain using mTeSR1 medium.backslashnbackslashnRESULTS: Cells grown under these conditions maintained similar morphology and growth rate to those grown on Matrigel and retained all pluripotent stem cell features,including an ability to differentiate into multiple cell lineages in teratoma assays. We,therefore,present a culture system that maintains the pluripotency of huES and hiPS cells under completely defined conditions.backslashnbackslashnCONCLUSIONS: We propose that this system should facilitate growth of stem cells using good manufacturing practices (GMP),which will be necessary for the clinical use of pluripotent stem cells and their derivatives.
View Publication
Mü et al. (JAN 2013)
Journal of visualized experiments : JoVE 80
Culturing of human nasal epithelial cells at the air liquid interface.
In vitro models using human primary epithelial cells are essential in understanding key functions of the respiratory epithelium in the context of microbial infections or inhaled agents. Direct comparisons of cells obtained from diseased populations allow us to characterize different phenotypes and dissect the underlying mechanisms mediating changes in epithelial cell function. Culturing epithelial cells from the human tracheobronchial region has been well documented,but is limited by the availability of human lung tissue or invasiveness associated with obtaining the bronchial brushes biopsies. Nasal epithelial cells are obtained through much less invasive superficial nasal scrape biopsies and subjects can be biopsied multiple times with no significant side effects. Additionally,the nose is the entry point to the respiratory system and therefore one of the first sites to be exposed to any kind of air-borne stressor,such as microbial agents,pollutants,or allergens. Briefly,nasal epithelial cells obtained from human volunteers are expanded on coated tissue culture plates,and then transferred onto cell culture inserts. Upon reaching confluency,cells continue to be cultured at the air-liquid interface (ALI),for several weeks,which creates more physiologically relevant conditions. The ALI culture condition uses defined media leading to a differentiated epithelium that exhibits morphological and functional characteristics similar to the human nasal epithelium,with both ciliated and mucus producing cells. Tissue culture inserts with differentiated nasal epithelial cells can be manipulated in a variety of ways depending on the research questions (treatment with pharmacological agents,transduction with lentiviral vectors,exposure to gases,or infection with microbial agents) and analyzed for numerous different endpoints ranging from cellular and molecular pathways,functional changes,morphology,etc. In vitro models of differentiated human nasal epithelial cells will enable investigators to address novel and important research questions by using organotypic experimental models that largely mimic the nasal epithelium in vivo.
View Publication
Y. Hong et al. (mar 2022)
JCI insight 7 5
Cure of syngeneic carcinomas with targeted IL-12 through obligate reprogramming of lymphoid and myeloid immunity.
Therapeutic IL-12 has demonstrated the ability to reduce local immune suppression in preclinical models,but clinical development has been limited by severe inflammation-related adverse events with systemic administration. Here,we show that potent immunologic tumor control of established syngeneic carcinomas can be achieved by i.t. administration of a tumor-targeted IL-12 antibody fusion protein (NHS-rmIL-12) using sufficiently low doses to avoid systemic toxicity. Single-cell transcriptomic analysis and ex vivo functional assays of NHS-rmIL-12-treated tumors revealed reinvigoration and enhanced proliferation of exhausted CD8+ T lymphocytes,induction of Th1 immunity,and a decrease in Treg number and suppressive capacity. Similarly,myeloid cells transitioned toward inflammatory phenotypes and displayed reduced suppressive capacity. Cell type-specific IL-12 receptor-KO BM chimera studies revealed that therapeutic modulation of both lymphoid and myeloid cells is required for maximum treatment effect and tumor cure. Study of single-cell data sets from human head and neck carcinomas revealed IL-12 receptor expression patterns similar to those observed in murine tumors. These results describing the diverse mechanisms underlying tumor-directed IL-12-induced antitumor immunity provide the preclinical rationale for the clinical study of i.t. NHS-IL-12.
View Publication
Botta G et al. ( 2012)
Current medicinal chemistry 19 34 5871--84
Current advances in the synthesis and antitumoral activity of SIRT1-2 inhibitors by modulation of p53 and pro-apoptotic proteins.
Four different classes of HDACs have been identified in humans so far. Classes I,II and IV are zinc-dependent amidohydrolases,while III is a family of phylogenetically conserved NAD-dependent protein deacetylases/ADP-ribosyltransferase with a well-defined role in modifying chromatin conformation and altering the accessibility of the damaged sites of DNA for repair enzymes. Sirtuins are histone deacetylases (HDACs) of class III that cleave off acetyl groups from acetyl-lysine residues in histones and non-histone proteins. As sirtuins are involved in many physiological and pathological processes,their activity has been associated with different human diseases,including cancer. Especially two sirtuin members,SIRT1 and SIRT2,have been found to antagonize p53-dependent transcriptional activation and apoptosis in response to DNA damage by catalyzing p53 deacetylation. The findings that SIRT1 levels are increased in a number of tumors highlight the oncogenic role of sirtuins,in particular,in the down-modulation of p53 oncosuppressor activity. Along this lane,cancers carrying wild-type (wt) p53 protein are known to deregulate its activity by other mechanisms. Therefore,inhibition of SIRT1 and SIRT2,aimed at restoring wt-p53 transcriptional activity in tumors that retain the ability to express normal p53,might represent a valid therapeutic cancer approach specially when combined with standard therapies. This review will be focused on sirtuin inhibitors,with a specific attention on inhibitors of SIRT1 and SIRT2. Among them,nicotinamide and its analogs,sirtinol,A3 and M15,splitomicin,HR73 and derivatives,cambinol and derivatives,EX 527,kinase inhibitors,suramin,4-dihydropyridine derivatives,tenovins,TRIPOS 360702,AC 93253,3-arylideneindolinones,CSC8 and CSC13 will also be described.
View Publication
Jennifer S et al. (JANUARY 2016)
Cytotherapy 18 1 1-12
Current perspectives on the use of ancillary materials for the manufacture of cellular therapies.
Continued growth in the cell therapy industry and commercialization of cell therapies that successfully advance through clinical trials has led to increased awareness around the need for specialized and complex materials utilized in their manufacture. Ancillary materials (AMs) are components or reagents used during the manufacture of cell therapy products but are not intended to be part of the final products. Commonly,there are limitations in the availability of clinical-grade reagents used as AMs. Furthermore,AMs may affect the efficacy of the cell product and subsequent safety of the cell therapy for the patient. As such,AMs must be carefully selected and appropriately qualified during the cell therapy development process. However,the ongoing evolution of cell therapy research,limited number of clinical trials and registered cell therapy products results in the current absence of specific regulations governing the composition,compliance,and qualification of AMs often leads to confusion by suppliers and users in this field. Here we provide an overview and interpretation of the existing global framework surrounding AM use and investigate some common misunderstandings within the industry,with the aim of facilitating the appropriate selection and qualification of AMs. The key message we wish to emphasize is that in order to most effectively mitigate risk around cell therapy development and patient safety,users must work with their suppliers and regulators to qualify each AM to assess source,purity,identity,safety,and suitability in a given application.
View Publication
Philonenko ES et al. (JAN 2011)
International review of cell and molecular biology 292 153--96
Current progress and potential practical application for human pluripotent stem cells.
Pluripotent stem cells are able to give rise to all cell types of the organism. There are two sources for human pluripotent stem cells: embryonic stem cells (ESCs) derived from surplus blastocysts created for in vitro fertilization and induced pluripotent stem cells (iPSCs) generated by reprogramming of somatic cells. ESCs have been an area of intense research during the past decade,and two clinical trials have been recently approved. iPSCs were created only recently,and most of the research has been focused on the iPSC generation protocols and investigation of mechanisms of direct reprogramming. The iPSC technology makes possible to derive pluripotent stem cells from any patient. However,there are a number of hurdles to be overcome before iPSCs will find a niche in practice. In this review,we discuss differences and similarities of the two pluripotent cell types and assess prospects for application of these cells in biomedicine.
View Publication
Van VQ et al. (OCT 2008)
Journal of immunology (Baltimore,Md. : 1950) 181 8 5204--8
Cutting edge: CD47 controls the in vivo proliferation and homeostasis of peripheral CD4+ CD25+ Foxp3+ regulatory T cells that express CD103.
Peripheral CD103(+)Foxp3(+) regulatory T cells (Tregs) can develop both from conventional naive T cells upon cognate Ag delivery under tolerogenic conditions and from thymic-derived,expanded/differentiated natural Tregs. We here show that CD47 expression,a marker of self on hematopoietic cells,selectively regulated CD103(+)Foxp3(+) Treg homeostasis at the steady state. First,the proportion of effector/memory-like (CD44(high)CD62L(low)) CD103(+)Foxp3(+) Tregs rapidly augmented with age in CD47-deficient mice (CD47(-/-)) as compared with age-matched control littermates. Yet,the percentage of quiescent (CD44(low)CD62L(high)) CD103(-)Foxp3(+) Tregs remained stable. Second,the increased proliferation rate (BrdU incorporation) observed within the CD47(-/-)Foxp3(+) Treg subpopulation was restricted to those Tregs expressing CD103. Third,CD47(-/-) Tregs maintained a normal suppressive function in vitro and in vivo and their increased proportion in old mice led to a decline of Ag-specific T cell responses. Thus,sustained CD47 expression throughout life is critical to avoid an excessive expansion of CD103(+) Tregs that may overwhelmingly inhibit Ag-specific T cell responses.
View Publication
D. J. Friedman et al. (apr 2022)
Journal of immunology (Baltimore,Md. : 1950) 208 8 1845--1850
Cutting Edge: Enhanced Antitumor Immunity in ST8Sia6 Knockout Mice.
Inhibitory receptors have a critical role in the regulation of immunity. Siglecs are a family of primarily inhibitory receptors expressed by immune cells that recognize specific sialic acid modifications on cell surface glycans. Many tumors have increased sialic acid incorporation. Overexpression of the sialyltransferase ST8Sia6 on tumors led to altered immune responses and increased tumor growth. In this study,we examined the role of ST8Sia6 on immune cells in regulating antitumor immunity. ST8Sia6 knockout mice had an enhanced immune response to tumors. The loss of ST8Sia6 promoted an enhanced intratumoral activation of macrophages and dendritic cells,including upregulation of CD40. Intratumoral regulatory T cells exhibited a more inflammatory phenotype in ST8Sia6 knockout mice. Using adoptive transfer studies,the change in regulatory T cell phenotype was not cell intrinsic and depended on the loss of ST8Sia6 expression in APCs. Thus,ST8Sia6 generates ligands for Siglecs that dampen antitumor immunity.
View Publication
Deets KA et al. (MAR 2016)
Journal of Immunology 196 6 2450--5
Cutting Edge: Enhanced Clonal Burst Size Corrects an Otherwise Defective Memory Response by CD8+ Recent Thymic Emigrants.
The youngest peripheral T cells (recent thymic emigrants [RTEs]) are functionally distinct from naive T cells that have completed postthymic maturation. We assessed the RTE memory response and found that RTEs produced less granzyme B than their mature counterparts during infection but proliferated more and,therefore,generated equivalent target killing in vivo. Postinfection,RTE numbers contracted less dramatically than those of mature T cells,but RTEs were delayed in their transition to central memory,displaying impaired expression of CD62L,IL-2,Eomesodermin,and CXCR4,which resulted in impaired bone marrow localization. RTE-derived and mature memory cells expanded equivalently during rechallenge,indicating that the robust proliferative capacity of RTEs was maintained independently of central memory phenotype. Thus,the diminished effector function and delayed central memory differentiation of RTE-derived memory cells are counterbalanced by their increased proliferative capacity,driving the efficacy of the RTE response to that of mature T cells.
View Publication
Jensen H et al. ( 2017)
Journal of immunology (Baltimore,Md. : 1950) 199 6 1967--1972
Cutting Edge: IL-2-Induced Expression of the Amino Acid Transporters SLC1A5 and CD98 Is a Prerequisite for NKG2D-Mediated Activation of Human NK Cells.
Priming of human NK cells with IL-2 is necessary to render them functionally competent upon NKG2D engagement. We examined the underlying mechanisms that control NKG2D responsiveness in NK cells and found that IL-2 upregulates expression of the amino acid transporters SLC1A5 and CD98. Using specific inhibitors to block SLC1A5 and CD98 function,we found that production of IFN-γ and degranulation by CD56bright and CD56dim NK cells following NKG2D stimulation were dependent on both transporters. IL-2 priming increased the activity of mTORC1,and inhibition of mTORC1 abrogated the ability of the IL-2-primed NK cells to produce IFN-γ in response to NKG2D-mediated stimulation. This study identifies a series of IL-2-induced cellular changes that regulates the NKG2D responsiveness in human NK cells.
View Publication
Glatigny S et al. (MAY 2016)
Journal of Immunology 196 9 3542--6
Cutting Edge: Integrin α4 Is Required for Regulatory B Cell Control of Experimental Autoimmune Encephalomyelitis.
The neutralization of integrin α4 (Itga4) is currently used as treatment in multiple sclerosis. Although most studies have focused on its function on lymphocyte migration to the CNS,we have uncovered the importance of Itga4 for the generation of regulatory B cells in peripheral immune organs and their control of pathogenic T cell response and CNS pathology. Our study underscores the importance of looking at the dual role of B cells in CNS autoimmunity and provides important perspectives regarding the efficacy and side effects associated with Itga4 neutralization and other B cell-targeting therapies.
View Publication
Koka R et al. (SEP 2004)
Journal of immunology (Baltimore,Md. : 1950) 173 6 3594--8
Cutting edge: murine dendritic cells require IL-15R alpha to prime NK cells.
NK cells protect hosts against viral pathogens and transformed cells,and dendritic cells (DCs) play important roles in activating NK cells. We now find that murine IL-15Ralpha-deficient DCs fail to support NK cell cytolytic activity and elaboration of IFN-gamma,despite the fact that these DCs express normal levels of costimulatory molecules and IL-12. By contrast,IL-15Ralpha expression on NK cells is entirely dispensable for their activation by DCs. In addition,blockade with anti-IL-15Ralpha and anti-IL-2Rbeta but not anti-IL-2Ralpha-specific Abs prevents NK cell activation by wild-type DCs. Finally,presentation of IL-15 by purified IL-15Ralpha/Fc in trans synergizes with IL-12 to support NK cell priming. These findings suggest that murine DCs require IL-15Ralpha to present IL-15 in trans to NK cells during NK cell priming.
View Publication