Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Carcinog ; 62(6): 845-854, 2023 06.
Article in English | MEDLINE | ID: mdl-36994661

ABSTRACT

Cutaneous squamous cell carcinoma (CSCC), which develops in response to ultraviolet irradiation exposure, is among the most common cancers. CSCC lesions can be removed by surgical excision, but 4.5% of these cancers reappear as aggressive and therapy-resistant tumors. CSCC tumors display a high mutation burden, and tumor frequency is dramatically increased in immune-suppressed patients, indicating a vital role for the immune system in controlling cancer development. Natural killer cells (NK cells) play a key role in cancer immune surveillance, and recent studies suggest that NK cells from healthy donors can be expanded from peripheral blood for use in therapy. In the present study, we test the ability of ex vivo expanded human NK cells to suppress the CSCC cell cancer phenotype and reduce tumor growth. We expanded human NK cells from multiple healthy donors, in the presence of IL-2, and tested their ability to suppress the CSCC cell cancer phenotype. NK cell treatment produced a dose-dependent reduction in SCC-13 and HaCaT cell spheroid growth and matrigel invasion and induced SCC-13 and HaCaT cell apoptosis as evidenced by increased procaspase 9, procaspase 3, and PARP cleavage. Moreover, two important CSCC cell pro-cancer signaling pathways, YAP1/TAZ/TEAD and MEK1/2-ERK1/2, were markedly reduced. Furthermore, tail-vein injection of NK cells markedly suppressed the growth of SCC-13 xenograft tumors in NSG mice, which was also associated with a reduction in YAP1 and MEK1/2-P levels and enhanced apoptosis. These findings show that NK cell treatment suppresses CSCC cell spheroid formation, invasion, viability, and tumor growth, suggesting NK cell treatment may be a candidate therapy for CSCC.


Subject(s)
Carcinoma, Squamous Cell , Skin Neoplasms , Humans , Animals , Mice , Cell Survival , Killer Cells, Natural , Apoptosis
2.
Front Immunol ; 12: 732135, 2021.
Article in English | MEDLINE | ID: mdl-34925314

ABSTRACT

Natural killer cells (NK cells) are the first line of the innate immune defense system, primarily located in peripheral circulation and lymphoid tissues. They kill virally infected and malignant cells through a balancing play of inhibitory and stimulatory receptors. In pre-clinical investigational studies, NK cells show promising anti-tumor effects and are used in adoptive transfer of activated and expanded cells, ex-vivo. NK cells express co-stimulatory molecules that are attractive targets for the immunotherapy of cancers. Recent clinical trials are investigating the use of CAR-NK for different cancers to determine the efficiency. Herein, we review NK cell therapy approaches (NK cell preparation from tissue sources, ways of expansion ex-vivo for "off-the-shelf" allogeneic cell-doses for therapies, and how different vector delivery systems are used to engineer NK cells with CARs) for cancer immunotherapy.


Subject(s)
Allogeneic Cells/immunology , Cell- and Tissue-Based Therapy/methods , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , Cell Engineering/methods , Fetal Blood/cytology , Humans , Induced Pluripotent Stem Cells/metabolism , Neoplasms/immunology , Receptors, Chimeric Antigen/genetics , Treatment Outcome
3.
Methods Mol Biol ; 2321: 137-154, 2021.
Article in English | MEDLINE | ID: mdl-34048013

ABSTRACT

Mice are a suitable animal model for sepsis studies because they recapitulate many aspects of the pathophysiology observed in septic human patients. It is ethically preferable to use mice for research over higher sentient species, when scientifically appropriate. Mice are also advantageous for research due to their small size, modest housing needs, the availability of genetically modified strains, and the broad range of reagents available for scientific assays on this species. Nevertheless, there are some intrinsic differences between mice and humans that should be recognized when considering the translational potential of sepsis therapies. It is often wise to complement traditional mouse studies with animal models that exhibit even greater similarity to humans, and in particular, models that better recapitulate the human immune response. Humanized mice are a promising tool to bridge this interspecies research gap. Herein, we provide a protocol to generate BLT humanized mice and describe their sepsis phenotype after cecal ligation and puncture (CLP).


Subject(s)
Sepsis/pathology , Animals , Cecum/immunology , Cecum/metabolism , Cecum/pathology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Immunity/immunology , Ligation/methods , Mice , Mice, Inbred NOD , Mice, SCID , Punctures/methods , Sepsis/immunology , Sepsis/metabolism
4.
J Immunol ; 206(2): 398-409, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33239421

ABSTRACT

IFN regulatory factor 3 (IRF3) is a transcription factor that is activated by multiple pattern-recognition receptors. We demonstrated previously that IRF3 plays a detrimental role in a severe mouse model of sepsis, induced by cecal ligation and puncture. In this study, we found that IRF3-knockout (KO) mice were greatly protected from sepsis in a clinically relevant version of the cecal ligation and puncture model incorporating crystalloid fluids and antibiotics, exhibiting improved survival, reduced disease score, lower levels of serum cytokines, and improved phagocytic function relative to wild-type (WT) mice. Computational modeling revealed that the overall complexity of the systemic inflammatory/immune network was similar in IRF3-KO versus WT septic mice, although the tempo of connectivity differed. Furthermore, the mediators driving the network differed: TNF-α, IL-1ß, and IL-6 predominated in WT mice, whereas MCP-1 and IL-6 predominated in IRF3-KO mice. Network analysis also suggested differential IL-6-related inflammatory programs in WT versus IRF3-KO mice. We created bone marrow chimeras to test the role of IRF3 within leukocytes versus stroma. Surprisingly, chimeras with IRF3-KO bone marrow showed little protection from sepsis, whereas chimeras with IRF3-KO stroma showed a substantial degree of protection. We found that WT and IRF3-KO macrophages had a similar capacity to produce IL-6 and phagocytose bacteria in vitro. Adoptive transfer experiments demonstrated that the genotype of the host environment affected the capacity of monocytes to produce IL-6 during sepsis. Thus, IRF3 acts principally within the stromal compartment to exacerbate sepsis pathogenesis via differential impacts on IL-6-related inflammatory programs.

5.
Cytotherapy ; 22(6): 322-328, 2020 06.
Article in English | MEDLINE | ID: mdl-32278551

ABSTRACT

BACKGROUND: Optimal expansion of therapeutic natural killer (NK) cell products has required media supplementation with human or fetal bovine serum, which raises safety and regulatory concerns for clinical manufacturing. Serum-free media (SFM) have been optimized for T-cell expansion, but few SFM systems have been developed for NK cells. Here, we compare six commercial clinical-grade SFM with our standard fetal bovine serum-containing medium for their ability to support NK cell expansion and function. METHODS: Human peripheral blood NK cells were expanded in selected media by recursive weekly stimulation with K562-based feeder cells expressing membrane-bound interleukin-21 and CD137L. Expansion was the primary readout, and the best-performing SFM was then compared with standard medium for cytotoxicity, phenotype, degranulation and cytokine secretion. Multiple lots were compared for consistency, and media was analyzed throughout for nutrient consumption and metabolic byproducts. RESULTS: TexMACS, OpTmizer, SCGM, ABS-001 and StemXVivo demonstrated equal or inferior NK cell expansion kinetics compared with standard medium, but expansion was markedly superior with AIM V + 5% Immune Cell Serum Replacement (ICSR; mean 5448 vs. 2621-fold expansion in 14 days). Surprisingly, NK cells expanded in AIM V + ICSR also showed increased cytotoxicity, tumor necrosis factor α secretion and DNAM-1, NKG2D, NKp30, FasL, granzyme B and perforin expression. Lot-to-lot variability was minimal. Glucose and glutamine consumption were inversely related to lactate and ammonia production. DISCUSSION: The AIM V + ICSR SFM system supports excellent ex vivo expansion of clinical-grade NK cells with the phenotype and function needed for adoptive immunotherapy.


Subject(s)
Culture Media, Serum-Free/pharmacology , Feeder Cells/drug effects , Killer Cells, Natural/drug effects , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Culture Techniques/methods , Cell Proliferation/drug effects , Culture Media/chemistry , Culture Media/pharmacology , Culture Media, Serum-Free/chemistry , Cytotoxicity, Immunologic , Fas Ligand Protein/metabolism , Humans , K562 Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Natural Cytotoxicity Triggering Receptor 3/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
J Immunol ; 201(1): 193-201, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29760192

ABSTRACT

Circadian rhythms coordinate an organism's activities and biological processes to the optimal time in the 24-h daylight cycle. We previously demonstrated that male C57BL/6 mice develop sepsis more rapidly when the disease is induced in the nighttime versus the daytime. In this report, we elucidate the mechanism of this diurnal difference. Sepsis was induced via cecal ligation and puncture (CLP) at zeitgeber time (ZT)-19 (2 am) or ZT-7 (2 pm). Like the males used in our prior study, female C57BL/6 mice had a worse outcome when CLP was induced at ZT-19 versus ZT-7, and these effects persisted when we pooled the data from both sexes. In contrast, mice with a mutated Period 2 (Per2) gene had a similar outcome when CLP was induced at ZT-19 versus ZT-7. Bone marrow chimeras reconstituted with C57BL/6 immune cells exhibited a worse outcome when sepsis was induced at ZT-19 versus ZT-7, whereas chimeras with Per2-mutated immune cells did not. Next, murine macrophages were subjected to serum shock to synchronize circadian rhythms and exposed to bacteria cultured from the mouse cecum at 4-h intervals for 48 h. We observed that IL-6 production oscillated with a 24-h period in C57BL/6 cells exposed to cecal bacteria. Interestingly, we observed a similar pattern when cells were exposed to the TLR2 agonist lipoteichoic acid. Furthermore, TLR2-knockout mice exhibited a similar sepsis phenotype when CLP was induced at ZT-19 versus ZT-7. Together, these data suggest that circadian rhythms in immune cells mediate diurnal variations in murine sepsis severity via a TLR2-dependent mechanism.


Subject(s)
Circadian Rhythm/physiology , Macrophages, Peritoneal/immunology , Sepsis/immunology , Sepsis/pathology , Toll-Like Receptor 2/metabolism , Animals , Cecum/surgery , Female , Interleukin-6/biosynthesis , Leukocytes/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Period Circadian Proteins/genetics , Teichoic Acids/pharmacology , Time Factors , Toll-Like Receptor 2/agonists
7.
Shock ; 47(5): 621-631, 2017 05.
Article in English | MEDLINE | ID: mdl-27755506

ABSTRACT

IFN regulatory factor (IRF)3 plays a detrimental role in the cecal ligation and puncture (CLP) mouse model of sepsis. However, it is unclear which pathway activates IRF3 in this context. In this report, we investigate two pathways that activate IRF3: the Stimulator of Interferon Genes (STING) pathway (that senses cytosolic DNA) and the TIR-domain-containing adapter-inducing interferon-ß (TRIF) pathway (that senses dsRNA and LPS via Toll-like receptor 3 and 4). Initially, we examine the impact of these pathways using a severe CLP model (∼90% mortality). Both STING-KO and TRIF-KO mice are protected from severe sepsis, exhibiting reduced mortality, disease score, hypothermia, and inflammatory cytokines relative to WT counterparts. STING/TRIF-DKO mice exhibit a similar phenotype to each of the single KO strains, suggesting that these pathways have an interrelated function. Subsequently, we examine the impact of these pathways using a moderate CLP model incorporating clinical treatments (Lactated Ringer Solution and antibiotics, ∼36% mortality). In this case, STING-KO mice show a similar phenotype to WT counterparts, while TRIF-KO mice show improved disease score and hypothermia. During sepsis, innate immune receptors recognize bacterial ligands and host-derived danger signals, including cell-free DNA released into the circulation. We show that IRF3 is activated in cultured macrophages treated with bacteria derived from the mouse cecum, dependent on TRIF, and in macrophages treated with mouse genomic DNA/Lipofectamine 2000, dependent on STING. Together, our data demonstrate that both the STING and TRIF pathways can promote sepsis pathogenesis; however, their contribution depends on the severity of the disease model. We show that bacteria are abundant in the peritoneum following both severe and moderate CLP, while cell-free DNA is more highly elevated in the serum following severe CLP compared with sham and moderate CLP. Hence, the presence of bacteria and cell-free DNA may explain the variable phenotypes in our severe CLP model (dependent on TRIF and STING) versus our moderate CLP model (dependent on TRIF only).


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Immunity, Innate/physiology , Membrane Proteins/metabolism , Sepsis/metabolism , Sepsis/pathology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Immunity, Innate/genetics , Inflammation/genetics , Inflammation/immunology , Membrane Proteins/genetics , Mice , Mice, Knockout , Resuscitation , Sepsis/immunology
8.
J Immunol ; 193(8): 4169-77, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25225666

ABSTRACT

Antiviral CD8(+) T cell recognition of MHC class I-peptide complexes on the surface of professional APCs is a requisite step in an effective immune response following many potentially lethal infections. Although MHC class I-peptide production is thought to be closely linked to the continued presence of virus, several studies have shown that the persistence of Ag presentation occurs for an extended period of time following the clearance of RNA viruses. However, the mechanism responsible for Ag presentation persistence following viral clearance was unknown until now. In this study, we used a recombinant DNA virus expressing different forms of a model Ag to study the mechanism of prolonged Ag presentation in mice. We determined that the persistence of Ag presentation consists of three distinct mechanistic phases, as follows: ongoing viral replication, persistence of virally infected cells, and cross-presentation of Ag. These data will allow manipulation of the form of Ag contained within viral vectors to produce the most effective and protective CD8(+) T cell response to be generated following vaccination.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Vaccinia virus/immunology , Animals , Cells, Cultured , Cross-Priming/immunology , Histocompatibility Antigens Class I/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Recombinant Proteins/immunology , Vaccinia virus/genetics , Virus Replication/immunology
9.
J Biol Chem ; 288(34): 24286-92, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23846690

ABSTRACT

The antigen processing compartments in antigen-presenting cells (APCs) have well known characteristics of multivesicular bodies (MVBs). However, the importance of MVB integrity to APC function remains unknown. In this study, we have altered the ultrastructure of the MVB by perturbing cholesterol content genetically through the use of a deletion of the lipid transporter Niemann-Pick type C1 (NPC1). Immunofluorescence and electron microscopic analyses reveal that the antigen processing compartments in NPC1(-/-) dendritic cells (DCs) have an abnormal ultrastructure in that the organelles are enlarged and the intraluminal vesicles are almost completely absent and those remaining are completely disorganized. MHC-II is restricted to the limiting membrane of these enlarged MVBs where it colocalizes with the peptide editor H2-DM. Curiously, proteolytic removal of the chaperone protein Invariant chain from MHC-II, degradation of internalized foreign antigens, and antigenic-peptide binding to nascent MHC-II are normal in NPC1(-/-) DCs. Antigen-pulsed NPC1(-/-) DCs are able to effectively activate antigen-specific CD4 T cells in vitro, and immunization of NPC1(-/-) mice reveals surprisingly normal CD4 T cell activation in vivo. Our data thus reveal that the localization of MHC-II on the intraluminal vesicles of multivesicular antigen processing compartments is not required for efficient antigen presentation by DCs.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Peptides/immunology , Proteins/immunology , Animals , Antigen Presentation/genetics , Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/genetics , Intracellular Signaling Peptides and Proteins , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Niemann-Pick C1 Protein , Peptides/genetics , Proteins/genetics
10.
J Biol Chem ; 288(19): 13236-42, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23532855

ABSTRACT

BACKGROUND: Antigen-specific CD4 T cells are activated by small numbers of antigenic peptide-MHC class II (pMHC-II) complexes on dendritic cells (DCs). RESULTS: Newly generated pMHC-II complexes are present in small clusters on the DC surface. CONCLUSION: pMHC-II clusters permit efficient T cell activation. SIGNIFICANCE: The appearance of clustered pMHC-II reveals the organization of the T cell antigen receptor ligand on the DC surface. Dendritic cells (DCs) function by stimulating naive antigen-specific CD4 T cells to proliferate and secrete a variety of immunomodulatory factors. The ability to activate naive T cells comes from the capacity of DCs to internalize, degrade, and express peptide fragments of antigenic proteins on their surface bound to MHC class II molecules (MHC-II). Although DCs express tens of thousands of distinct MHC-II, very small amounts of specific peptide-MHC-II complexes are required to interact with and activate T cells. We now show that stimulatory MHC-II I-A(k)-HEL(46-61) complexes that move from intracellular antigen-processing compartments to the plasma membrane are not randomly distributed on the DC surface. Confocal immunofluorescence microscopy and quantitative immunoelectron microscopy reveal that the majority of newly generated MHC-II I-A(k)-HEL(46-61) complexes are expressed in sub-100-nm microclusters on the DC membrane. These microclusters are stabilized in cholesterol-containing microdomains, and cholesterol depletion inhibits the stability of these clusters as well as the ability of the DCs to function as antigen-presenting cells. These results demonstrate that specific cohorts of peptide-MHC-II complexes expressed on the DC surface are present in cholesterol-dependent microclusters and that cluster integrity is important for antigen-specific naive CD4 T cell activation by DCs.


Subject(s)
Cell Membrane/metabolism , Cholesterol/metabolism , Histocompatibility Antigens Class II/metabolism , Membrane Microdomains/metabolism , Animals , Antigen Presentation , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lymphocyte Activation , Membrane Lipids/metabolism , Mice , Mice, Transgenic , Protein Transport
11.
PLoS Pathog ; 7(11): e1002374, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22102816

ABSTRACT

The goal of the innate immune system is containment of a pathogen at the site of infection prior to the initiation of an effective adaptive immune response. However, effector mechanisms must be kept in check to combat the pathogen while simultaneously limiting undesirable destruction of tissue resulting from these actions. Here we demonstrate that innate immune effector cells contain a peripheral poxvirus infection, preventing systemic spread of the virus. These innate immune effector cells are comprised primarily of CD11b⁺Ly6C⁺Ly6G⁻ monocytes that accumulate initially at the site of infection, and are then supplemented and eventually replaced by CD11b⁺Ly6C⁺Ly6G⁺ cells. The phenotype of the CD11b⁺Ly6C⁺Ly6G⁺ cells resembles neutrophils, but the infiltration of neutrophils typically occurs prior to, rather than following, accumulation of monocytes. Indeed, it appears that the CD11b⁺Ly6C⁺Ly6G⁺ cells that infiltrated the site of VACV infection in the ear are phenotypically distinct from the classical description of both neutrophils and monocyte/macrophages. We found that CD11b⁺Ly6C⁺Ly6G⁺ cells produce Type I interferons and large quantities of reactive oxygen species. We also observed that depletion of Ly6G⁺ cells results in a dramatic increase in tissue damage at the site of infection. Tissue damage is also increased in the absence of reactive oxygen species, although reactive oxygen species are typically thought to be damaging to tissue rather than protective. These data indicate the existence of a specialized population of CD11b⁺Ly6C⁺Ly6G⁺ cells that infiltrates a site of virus infection late and protects the infected tissue from immune-mediated damage via production of reactive oxygen species. Regulation of the action of this population of cells may provide an intervention to prevent innate immune-mediated tissue destruction.


Subject(s)
Immunity, Innate , Interferon Type I/biosynthesis , Monocytes/immunology , Neutrophils/immunology , Vaccinia virus/immunology , Vaccinia/immunology , Animals , Antigens, Ly/analysis , CD11b Antigen/analysis , Interferon Type I/immunology , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/virology , Neutrophils/virology , Reactive Oxygen Species/metabolism , Vaccinia/virology , Vaccinia virus/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...