Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Proc Natl Acad Sci U S A ; 115(32): 8179-8184, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30038013

ABSTRACT

Agents that remodel the tumor microenvironment (TME), prime functional tumor-specific T cells, and block inhibitory signaling pathways are essential components of effective immunotherapy. We are evaluating live-attenuated, double-deleted Listeria monocytogenes expressing tumor antigens (LADD-Ag) in the clinic. Here we show in numerous mouse models that while treatment with nonrecombinant LADD induced some changes in the TME, no antitumor efficacy was observed, even when combined with immune checkpoint blockade. In contrast, LADD-Ag promoted tumor rejection by priming tumor-specific KLRG1+PD1loCD62L- CD8+ T cells. These IFNγ-producing effector CD8+ T cells infiltrated the tumor and converted the tumor from an immunosuppressive to an inflamed microenvironment that was characterized by a decrease in regulatory T cells (Treg) levels, a proinflammatory cytokine milieu, and the shift of M2 macrophages to an inducible nitric oxide synthase (iNOS)+CD206- M1 phenotype. Remarkably, these LADD-Ag-induced tumor-specific T cells persisted for more than 2 months after primary tumor challenge and rapidly controlled secondary tumor challenge. Our results indicate that the striking antitumor efficacy observed in mice with LADD-based immunotherapy stems from TME remodeling which is a direct consequence of eliciting potent, systemic tumor-specific CD8+ T cells.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Listeria monocytogenes/immunology , Neoplasms/therapy , Tumor Microenvironment/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/therapeutic use , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Listeria monocytogenes/genetics , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/immunology , Treatment Outcome , Vaccination/methods , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Attenuated/therapeutic use , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Xenograft Model Antitumor Assays
2.
Eur J Immunol ; 43(9): 2398-408, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23716300

ABSTRACT

Glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE) is a synthetic adjuvant TLR4 agonist that promotes potent poly-functional T(H)1 responses. Different TLR4 agonists may preferentially signal via MyD88 or TIR-domain-containing adapter inducing IFN-beta (TRIF) to exert adjuvant effects; however, the contribution of MyD88 and TRIF signaling to the induction of polyclonal T(H)1 responses by TLR4 agonist adjuvants has not been studied in vivo. To determine whether GLA-SE preferentially signals through MyD88 or TRIF, we evaluated the immune response against a candidate tuberculosis (TB) vaccine Ag following immunization of mice lacking either signaling adapter compared with that of wild-type mice. We find that both MyD88 and TRIF are necessary for GLA-SE to induce a poly-functional T(H)1 immune response characterized by CD4(+) T cells producing IFN-γ, TNF, and IL-2, as well as IgG2c class switching, when paired with the TB vaccine Ag ID93. Accordingly, the protective efficacy of ID93/GLA-SE immunization against aerosolized Mycobacterium tuberculosis was lost when either signaling molecule was ablated. We demonstrate that MyD88 and TRIF must be expressed in the same cell for the in vivo T(H)1-skewing adjuvant activity, indicating that these two signaling pathways cooperate on an intracellular level. Thus engagement of both the MyD88 and TRIF signaling pathways are essential for the effective adjuvant activity of this TLR4 agonist.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Adjuvants, Immunologic/metabolism , Myeloid Differentiation Factor 88/metabolism , Th1 Cells/immunology , Toll-Like Receptor 4/agonists , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunization , Immunoglobulin Class Switching/immunology , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium Infections/immunology , Mycobacterium tuberculosis/immunology , Receptors, IgG/metabolism , Signal Transduction/immunology , Tuberculosis Vaccines/immunology , Tumor Necrosis Factor-alpha/biosynthesis
3.
Proc Natl Acad Sci U S A ; 107(9): 4194-9, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20160075

ABSTRACT

Macrophages are required for tissue homeostasis through their role in regulation of the immune response and the resolution of injury. Here we show, using the kidney as a model, that the Wnt pathway ligand Wnt7b is produced by macrophages to stimulate repair and regeneration. When macrophages are inducibly ablated from the injured kidney, the canonical Wnt pathway response in kidney epithelial cells is reduced. Furthermore, when Wnt7b is somatically deleted in macrophages, repair of injury is greatly diminished. Finally, injection of the Wnt pathway regulator Dkk2 enhances the repair process and suggests a therapeutic option. Because Wnt7b is known to stimulate epithelial responses during kidney development, these findings suggest that macrophages are able to rapidly invade an injured tissue and reestablish a developmental program that is beneficial for repair and regeneration.


Subject(s)
Kidney/physiology , Macrophages/metabolism , Proto-Oncogene Proteins/physiology , Regeneration , Wnt Proteins/physiology , Animals , Base Sequence , Cell Cycle , DNA Primers , Intercellular Signaling Peptides and Proteins/physiology , Kidney/cytology , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Proto-Oncogene Proteins/metabolism , Signal Transduction , Wnt Proteins/metabolism
4.
Circulation ; 121(20): 2211-20, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20458011

ABSTRACT

BACKGROUND: Understanding the mechanisms of repair and regeneration of the kidney after injury is of great interest because there are currently no therapies that promote repair, and kidneys frequently do not repair adequately. We studied the capacity of human CD34(+) hematopoietic stem/progenitor cells (HSPCs) to promote kidney repair and regeneration using an established ischemia/reperfusion injury model in mice, with particular focus on the microvasculature. METHODS AND RESULTS: Human HSPCs administered systemically 24 hours after kidney injury were selectively recruited to injured kidneys of immunodeficient mice (Jackson Labs, Bar Harbor, Me) and localized prominently in and around vasculature. This recruitment was associated with enhanced repair of the kidney microvasculature, tubule epithelial cells, enhanced functional recovery, and increased survival. HSPCs recruited to kidney expressed markers consistent with circulating endothelial progenitors and synthesized high levels of proangiogenic cytokines, which promoted proliferation of both endothelial and epithelial cells. Although purified HSPCs acquired endothelial progenitor markers once recruited to the kidney, engraftment of human endothelial cells in the mouse capillary walls was an extremely rare event, indicating that human stem cell mediated renal repair is by paracrine mechanisms rather than replacement of vasculature. CONCLUSIONS: These studies advance human HSPCs as a promising therapeutic strategy for promoting renal repair after injury.


Subject(s)
Cell Movement , Hematopoietic Stem Cells , Kidney/blood supply , Kidney/physiopathology , Regeneration , Reperfusion Injury/physiopathology , Wound Healing , Animals , Antigens, CD34/metabolism , Biomarkers/metabolism , Capillaries/pathology , Colony-Stimulating Factors/pharmacology , Epithelial Cells , Fibrosis/prevention & control , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Immunocompetence , Kidney/pathology , Kidney Tubules/blood supply , Kidney Tubules/physiopathology , Mice/immunology , Microcirculation , Paracrine Communication , Recovery of Function , Survival Analysis
5.
Am J Pathol ; 176(1): 85-97, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20008127

ABSTRACT

Understanding the origin of myofibroblasts in kidney is of great interest because these cells are responsible for scar formation in fibrotic kidney disease. Recent studies suggest epithelial cells are an important source of myofibroblasts through a process described as the epithelial-to-mesenchymal transition; however, confirmatory studies in vivo are lacking. To quantitatively assess the contribution of renal epithelial cells to myofibroblasts, we used Cre/Lox techniques to genetically label and fate map renal epithelia in models of kidney fibrosis. Genetically labeled primary proximal epithelial cells cultured in vitro from these mice readily induce markers of myofibroblasts after transforming growth factor beta(1) treatment. However, using either red fluorescent protein or beta-galactosidase as fate markers, we found no evidence that epithelial cells migrate outside of the tubular basement membrane and differentiate into interstitial myofibroblasts in vivo. Thus, although renal epithelial cells can acquire mesenchymal markers in vitro, they do not directly contribute to interstitial myofibroblast cells in vivo. Lineage analysis shows that during nephrogenesis, FoxD1-positive((+)) mesenchymal cells give rise to adult CD73(+), platelet derived growth factor receptor beta(+), smooth muscle actin-negative interstitial pericytes, and these FoxD1-derivative interstitial cells expand and differentiate into smooth muscle actin(+) myofibroblasts during fibrosis, accounting for a large majority of myofibroblasts. These data indicate that therapeutic strategies directly targeting pericyte differentiation in vivo may productively impact fibrotic kidney disease.


Subject(s)
Cell Lineage , Epithelial Cells/pathology , Fibroblasts/pathology , Kidney/pathology , Pericytes/pathology , Actins/metabolism , Animals , Cell Movement , Cells, Cultured , Disease Models, Animal , Epithelial Cells/metabolism , Fibroblasts/metabolism , Fibrosis , Forkhead Transcription Factors/metabolism , Green Fluorescent Proteins , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Integrases/metabolism , Kidney/metabolism , Kidney Tubules/metabolism , Kidney Tubules/pathology , Mesoderm/metabolism , Mesoderm/pathology , Mice , Pericytes/metabolism , Promoter Regions, Genetic/genetics , S100 Calcium-Binding Protein A4 , S100 Proteins/metabolism , Transcription Factors/genetics , Up-Regulation
6.
FASEB J ; 24(12): 4767-81, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20709912

ABSTRACT

Kidney damage due to injury rarely resolves completely, and there are currently no therapies capable of promoting repair. In addition to understanding mechanisms by which tissues are damaged, illuminating mechanisms of repair and regeneration is also of great importance. Here we show that the melanoma-associated, transmembrane glycoprotein, Gpnmb, is up-regulated 15-fold following ischemic damage in kidney tissue and by more than 10-fold in macrophages and 3-fold in surviving epithelial cells. Gpnmb-expressing macrophages and epithelial cells were found to contain apoptotic bodies at 3 times the rate of nonexpressing cells. Either mutation of Gpnmb or ablation of inflammatory macrophages prevents normal repair of the kidney. Significantly, the kidneys from postischemic Gpnmb mutant mice exhibited a 5-fold increase in apoptotic cellular debris compared to wild-type mice. These mice also experienced an 85% increase in mortality following bilateral ischemic kidney. Finally, we demonstrate that Gpnmb is a phagocytic protein that is necessary for recruitment of the autophagy protein LC3 to the phagosome where these proteins are colocalized and for lysosomal fusion with the phagosome and hence bulk degradation of their content. Therefore, Gpnmb is a novel prorepair gene that is necessary for crosstalk between the macroautophagic degradation pathway and phagocytosis.


Subject(s)
Biological Transport/physiology , Eye Proteins/metabolism , Membrane Glycoproteins/metabolism , Animals , Cell Line , Eye Proteins/genetics , Flow Cytometry , Humans , Immunoblotting , Kidney/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Membrane Glycoproteins/genetics , Mice , Mice, Mutant Strains , Microscopy, Fluorescence , Phagocytosis/physiology , Plaque, Atherosclerotic/chemically induced , Plaque, Atherosclerotic/metabolism , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism
7.
Cell Rep ; 29(7): 1832-1847.e8, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31722201

ABSTRACT

Iterative liver injury results in progressive fibrosis disrupting hepatic architecture, regeneration potential, and liver function. Hepatic stellate cells (HSCs) are a major source of pathological matrix during fibrosis and are thought to be a functionally homogeneous population. Here, we use single-cell RNA sequencing to deconvolve the hepatic mesenchyme in healthy and fibrotic mouse liver, revealing spatial zonation of HSCs across the hepatic lobule. Furthermore, we show that HSCs partition into topographically diametric lobule regions, designated portal vein-associated HSCs (PaHSCs) and central vein-associated HSCs (CaHSCs). Importantly we uncover functional zonation, identifying CaHSCs as the dominant pathogenic collagen-producing cells in a mouse model of centrilobular fibrosis. Finally, we identify LPAR1 as a therapeutic target on collagen-producing CaHSCs, demonstrating that blockade of LPAR1 inhibits liver fibrosis in a rodent NASH model. Taken together, our work illustrates the power of single-cell transcriptomics to resolve the key collagen-producing cells driving liver fibrosis with high precision.


Subject(s)
Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , Single-Cell Analysis , Transcriptome , Animals , Disease Models, Animal , Hepatic Stellate Cells/pathology , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice , Mice, Transgenic , Rats , Rats, Wistar , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism
8.
Cell Rep ; 25(11): 3074-3085.e5, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30540940

ABSTRACT

Intratumoral (IT) STING activation results in tumor regression in preclinical models, yet factors dictating the balance between innate and adaptive anti-tumor immunity are unclear. Here, clinical candidate STING agonist ADU-S100 (S100) is used in an IT dosing regimen optimized for adaptive immunity to uncover requirements for a T cell-driven response compatible with checkpoint inhibitors (CPIs). In contrast to high-dose tumor ablative regimens that result in systemic S100 distribution, low-dose immunogenic regimens induce local activation of tumor-specific CD8+ effector T cells that are responsible for durable anti-tumor immunity and can be enhanced with CPIs. Both hematopoietic cell STING expression and signaling through IFNAR are required for tumor-specific T cell activation, and in the context of optimized T cell responses, TNFα is dispensable for tumor control. In a poorly immunogenic model, S100 combined with CPIs generates a survival benefit and durable protection. These results provide fundamental mechanistic insights into STING-induced anti-tumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity , Membrane Proteins/metabolism , Neoplasms/immunology , Animals , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Cytokines/metabolism , Dose-Response Relationship, Immunologic , Drug Resistance, Neoplasm , Hematopoiesis , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/pathology , Programmed Cell Death 1 Receptor/metabolism , S100 Proteins/administration & dosage , S100 Proteins/immunology
9.
PLoS One ; 11(1): e0146372, 2016.
Article in English | MEDLINE | ID: mdl-26731269

ABSTRACT

The Toll-like receptor 4 agonist glucopyranosyl lipid adjuvant formulated in a stable emulsion (GLA-SE) promotes strong TH1 and balanced IgG1/IgG2 responses to protein vaccine antigens. This enhanced immunity is sufficient to provide protection against many diseases including tuberculosis and leishmaniasis. To better characterize the adjuvant action it is important to understand how the different cytokines and transcription factors contribute to the initiation of immunity. In the present study using T-bet-/- and IL-12-/- mice and a blocking anti-IFNαR1 monoclonal antibody, we define mechanisms of adjuvant activity of GLA-SE. In accordance with previous studies of TLR4 agonist based adjuvants, we found that TH1 induction via GLA-SE was completely dependent upon T-bet, a key transcription factor for IFNγ production and TH1 differentiation. Consistent with this, deficiency of IL-12, a cytokine canonical to TH1 induction, ablated TH1 induction via GLA-SE. Finally we demonstrate that the innate immune response to GLA-SE, including rapid IFNγ production by memory CD8+ T cells and NK cells, was contingent on type I interferon, a cytokine group whose association with TH1 induction is contextual, and that they contributed to the adjuvant activity of GLA-SE.


Subject(s)
Glucosides/pharmacology , Lipid A/pharmacology , Th1 Cells/drug effects , Adjuvants, Immunologic/pharmacology , Animals , Immunity, Innate/drug effects , Mice , Th1 Cells/immunology , Tuberculosis/immunology
10.
Vaccine ; 33(48): 6570-8, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26541135

ABSTRACT

Pulmonary tuberculosis (TB) remains one of the leading causes of infectious disease death despite widespread usage of the BCG vaccine. A number of new TB vaccines have moved into clinical evaluation to replace or boost the BCG vaccine including ID93+GLA-SE, an adjuvanted subunit vaccine. The vast majority of new TB vaccines in trials are delivered parenterally even though intranasal delivery can augment lung-resident immunity and protective efficacy in small animal models. Parenteral immunization with the adjuvanted subunit vaccine ID93+GLA-SE elicits robust TH1 immunity and protection against aerosolized Mycobacterium tuberculosis in mice and guinea pigs. Here we describe the immunogenicity and efficacy of this vaccine when delivered intranasally. Intranasal delivery switches the CD4 T cell response from a TH1 to a TH17 dominated tissue-resident response with increased frequencies of ID93-specific cells in both the lung tissue and at the lung surface. Surprisingly these changes do not affect the protective efficacy of ID93+GLA-SE. Unlike intramuscular immunization, ID93+GLA does not require the squalene-based oil-in-water emulsion SE to elicit protective CD4 T cells when delivered intranasally. Finally we demonstrate that TNF and the IL-17 receptor are dispensable for the efficacy of the intranasal vaccine suggesting an alternative mechanism of protection.


Subject(s)
Th1 Cells/immunology , Th17 Cells/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Adjuvants, Immunologic/administration & dosage , Administration, Mucosal , Animals , Antigens, Bacterial/immunology , BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Guinea Pigs , Humans , Mice , Mycobacterium tuberculosis/immunology , Peptide Fragments/immunology , Tumor Necrosis Factor-alpha/immunology
11.
PLoS One ; 9(1): e83884, 2014.
Article in English | MEDLINE | ID: mdl-24404140

ABSTRACT

With over eight million cases of tuberculosis each year there is a pressing need for the development of new vaccines against Mycobacterium tuberculosis. Subunit vaccines consisting of recombinant proteins are an attractive vaccine approach due to their inherent safety compared to attenuated live vaccines and the uniformity of manufacture. Addition of properly formulated TLR agonist-containing adjuvants to recombinant protein vaccines enhances the antigen-specific CD4(+) T cell response characterized by IFN-γ and TNF, both of which are critical for the control of TB. We have developed a clinical stage vaccine candidate consisting of a recombinant fusion protein ID93 adjuvanted with the TLR4 agonist GLA-SE. Here we examine whether ID93+GLA-SE can be improved by the addition of a second TLR agonist. Addition of CpG containing DNA to ID93+GLA-SE enhanced the magnitude of the multi-functional TH1 response against ID93 characterized by co-production of IFN-γ, TNF, and IL-2. Addition of CpG also improved the protective efficacy of ID93+GLA-SE. Finally we demonstrate that this adjuvant synergy between GLA and CpG is independent of TRIF signaling, whereas TRIF is necessary for the adjuvant activity of GLA-SE in the absence of CpG.


Subject(s)
Adjuvants, Immunologic , Mycobacterium tuberculosis/immunology , Toll-Like Receptors/agonists , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Antigens/immunology , Antimicrobial Cationic Peptides/immunology , Disease Models, Animal , Female , Mice , Mice, Knockout , Signal Transduction , Th1 Cells/immunology , Tuberculosis/immunology , Tuberculosis/metabolism , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL