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










Publication year range
2.
Cell ; 184(8): 2033-2052.e21, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33765443

ABSTRACT

Metastasis is the leading cause of cancer-related deaths, and greater knowledge of the metastatic microenvironment is necessary to effectively target this process. Microenvironmental changes occur at distant sites prior to clinically detectable metastatic disease; however, the key niche regulatory signals during metastatic progression remain poorly characterized. Here, we identify a core immune suppression gene signature in pre-metastatic niche formation that is expressed predominantly by myeloid cells. We target this immune suppression program by utilizing genetically engineered myeloid cells (GEMys) to deliver IL-12 to modulate the metastatic microenvironment. Our data demonstrate that IL12-GEMy treatment reverses immune suppression in the pre-metastatic niche by activating antigen presentation and T cell activation, resulting in reduced metastatic and primary tumor burden and improved survival of tumor-bearing mice. We demonstrate that IL12-GEMys can functionally modulate the core program of immune suppression in the pre-metastatic niche to successfully rebalance the dysregulated metastatic microenvironment in cancer.


Subject(s)
Immunosuppression Therapy , Myeloid Cells/metabolism , Adaptive Immunity , Animals , Cell Line, Tumor , Genetic Engineering , Humans , Interleukin-12/genetics , Interleukin-12/metabolism , Lung/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/immunology , Neoplasm Metastasis , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment
3.
Nat Med ; 23(10): 1176-1190, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28920957

ABSTRACT

A deeper understanding of the metastatic process is required for the development of new therapies that improve patient survival. Metastatic tumor cell growth and survival in distant organs is facilitated by the formation of a pre-metastatic niche that is composed of hematopoietic cells, stromal cells and extracellular matrix (ECM). Perivascular cells, including vascular smooth muscle cells (vSMCs) and pericytes, are involved in new vessel formation and in promoting stem cell maintenance and proliferation. Given the well-described plasticity of perivascular cells, we hypothesized that perivascular cells similarly regulate tumor cell fate at metastatic sites. We used perivascular-cell-specific and pericyte-specific lineage-tracing models to trace the fate of perivascular cells in the pre-metastatic and metastatic microenvironments. We show that perivascular cells lose the expression of traditional vSMC and pericyte markers in response to tumor-secreted factors and exhibit increased proliferation, migration and ECM synthesis. Increased expression of the pluripotency gene Klf4 in these phenotypically switched perivascular cells promoted a less differentiated state, characterized by enhanced ECM production, that established a pro-metastatic fibronectin-rich environment. Genetic inactivation of Klf4 in perivascular cells decreased formation of a pre-metastatic niche and metastasis. Our data revealed a previously unidentified role for perivascular cells in pre-metastatic niche formation and uncovered novel strategies for limiting metastasis.


Subject(s)
Cell Plasticity/genetics , Kruppel-Like Transcription Factors/genetics , Myocytes, Smooth Muscle/metabolism , Neoplasm Metastasis/genetics , Pericytes/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Gene Knockdown Techniques , In Vitro Techniques , Kruppel-Like Factor 4 , Melanoma, Experimental , Mice , Muscle, Smooth, Vascular/cytology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Real-Time Polymerase Chain Reaction , Tumor Microenvironment
4.
Nat Med ; 22(6): 657-65, 2016 06.
Article in English | MEDLINE | ID: mdl-27183216

ABSTRACT

Although somatic cell activation of the embryonic stem cell (ESC) pluripotency factor OCT4 has been reported, this previous work has been controversial and has not demonstrated a functional role for OCT4 in somatic cells. Here we demonstrate that smooth muscle cell (SMC)-specific conditional knockout of Oct4 in Apoe(-/-) mice resulted in increased lesion size and changes in lesion composition that are consistent with decreased plaque stability, including a thinner fibrous cap, increased necrotic core area, and increased intraplaque hemorrhage. Results of SMC-lineage-tracing studies showed that these effects were probably the result of marked reductions in SMC numbers within lesions and SMC investment within the fibrous cap, which may result from impaired SMC migration. The reactivation of Oct4 within SMCs was associated with hydroxymethylation of the Oct4 promoter and was hypoxia inducible factor-1α (HIF-1α, encoded by HIF1A) and Krüppel-like factor-4 (KLF4)-dependent. These results provide the first direct evidence that OCT4 has a functional role in somatic cells, and they highlight the potential role of OCT4 in normal and diseased somatic cells.


Subject(s)
Atherosclerosis/genetics , Cell Movement/genetics , Myocytes, Smooth Muscle/metabolism , Octamer Transcription Factor-3/genetics , Plaque, Atherosclerotic/genetics , Animals , Aorta/metabolism , Apolipoproteins E/genetics , Blotting, Western , Cell Lineage , Cell Survival , Chromatin Immunoprecipitation , Coronary Artery Disease/metabolism , Diet, Western , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Mutagenesis, Site-Directed , Myocytes, Smooth Muscle/cytology , Octamer Transcription Factor-3/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
5.
Cancer Res ; 76(6): 1335-47, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26719537

ABSTRACT

Metastatic tumors have been shown to establish microenvironments in distant tissues that are permissive to disseminated tumor cells. Hematopoietic cells contribute to this microenvironment, yet the precise initiating events responsible for establishing the pre-metastatic niche remain unclear. Here, we tracked the developmental fate of hematopoietic stem and progenitor cells (HSPC) in tumor-bearing mice. We show that a distant primary tumor drives the expansion of HSPCs within the bone marrow and their mobilization to the bloodstream. Treatment of purified HSPCs cultured ex vivo with tumor-conditioned media induced their proliferation as well as their differentiation into immunosuppressive myeloid cells. We furthered tracked purified HSPCs in vivo and found they differentiated into myeloid-derived suppressor cells in early metastatic sites of tumor-bearing mice. The number of CD11b(+)Ly6g(+) cells in metastatic sites was significantly increased by HSPC mobilization and decreased if tumor-mediated mobilization was inhibited. Moreover, pharmacologic mobilization of HSPCs increased metastasis, whereas depletion of Gr1(+) cells abrogated the metastasis-promoting effects of HSPC mobilization. Finally, we detected elevated levels of HSPCs in the circulation of newly diagnosed cancer patients, which correlated with increased risk for metastatic progression. Taken together, our results highlight bone marrow activation as one of the earliest steps of the metastatic process and identify circulating HSPCs as potential clinical indicators of metastatic niche formation.


Subject(s)
Bone Marrow Cells/pathology , Hematopoietic Stem Cells/pathology , Neoplasm Metastasis/pathology , Stem Cells/pathology , Adolescent , Adult , Animals , Bone Marrow/pathology , Cell Differentiation/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Child , Child, Preschool , Humans , Immune Tolerance/physiology , Immunosuppression Therapy/methods , Infant , Mice , Mice, Inbred C57BL , Myeloid Cells/pathology , Young Adult
6.
Crit Rev Oncog ; 20(3-4): 301-14, 2015.
Article in English | MEDLINE | ID: mdl-26349421

ABSTRACT

The primary tumor niche and the related but distinct premetastatic/metastatic niche comprise a number of essential players, including immune cells, stromal cells, and extracellular matrix. The cross-talk between these components is key to tumor progression. Many of these cell types and signaling pathways in the tumor microenvironment also are found in physiological and stem cell niches, such as the bone marrow, colonic crypt, and skin bulge. Here they play tightly regulated roles in wound healing and tissue homeostasis. Understanding the similarities and differences between these distinct niches may better inform our ability to therapeutically target the tumor microenvironment. In this review we discuss a number of tumor and metastatic niche components as they relate to stem cell niches and highlight potential therapeutic strategies in pediatric cancers.


Subject(s)
Gene Targeting/methods , Neoplasms/genetics , Neoplasms/therapy , Stem Cell Niche/genetics , Tumor Microenvironment/genetics , Animals , Gene Targeting/trends , Humans , Immunotherapy/methods , Immunotherapy/trends , Neoplasms/diagnosis
7.
Nat Med ; 21(6): 581-90, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25939063

ABSTRACT

Chimeric antigen receptors (CARs) targeting CD19 have mediated dramatic antitumor responses in hematologic malignancies, but tumor regression has rarely occurred using CARs targeting other antigens. It remains unknown whether the impressive effects of CD19 CARs relate to greater susceptibility of hematologic malignancies to CAR therapies, or superior functionality of the CD19 CAR itself. We show that tonic CAR CD3-ζ phosphorylation, triggered by antigen-independent clustering of CAR single-chain variable fragments, can induce early exhaustion of CAR T cells that limits antitumor efficacy. Such activation is present to varying degrees in all CARs studied, except the highly effective CD19 CAR. We further determine that CD28 costimulation augments, whereas 4-1BB costimulation reduces, exhaustion induced by persistent CAR signaling. Our results provide biological explanations for the antitumor effects of CD19 CARs and for the observations that CD19 CAR T cells incorporating the 4-1BB costimulatory domain are more persistent than those incorporating CD28 in clinical trials.


Subject(s)
Hematologic Neoplasms/immunology , Receptors, Antigen/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Antigens, CD19/immunology , Antigens, CD19/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , Cell Line, Tumor , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Immunotherapy, Adoptive , Interleukin-2/immunology , Lymphocyte Activation/immunology , Receptors, Antigen/metabolism , T-Lymphocytes/pathology , Tumor Necrosis Factor Receptor Superfamily, Member 9/biosynthesis
8.
Retrovirology ; 10: 34, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23537062

ABSTRACT

BACKGROUND: Xenotropic Murine leukemia virus-Related Virus (XMRV) is a γ-retrovirus initially reported to be present within familial human prostate tumors and the blood of patients with chronic fatigue syndrome. Subsequent studies however were unable to replicate these findings, and there is now compelling evidence that the virus evolved through rare retroviral recombination events in human tumor cell lines established through murine xenograft experiments. There is also no direct evidence that XMRV infection has any functional effects that contribute to tumor pathogenesis. RESULTS: Herein we describe an additional xenotropic MLV, "B4rv", found in a cell line derived from xenograft experiments with the human prostate cancer LNCaP cell line. When injected subcutaneously in nude mice, LNCaP cells infected with XMRV or B4rv formed larger tumors that were highly hemorrhagic and displayed poor pericyte/smooth muscle cell (SMC) investment, markers of increased metastatic potential. Conditioned media derived from XMRV- or B4rv-infected LNCaPs, but not an amphotropic MLV control virus infected LNCaPs, profoundly decreased expression of marker genes in cultured SMC, consistent with inhibition of SMC differentiation/maturation. Similar effects were seen with a chimeric virus of the amphotropic MLV control virus containing the XMRV env gene, but not with an XMRV chimeric virus containing the amphotropic MLV env gene. UV-inactivated XMRV and pseudovirions that were pseudotyped with XMRV envelope protein also produce conditioned media that down-regulated SMC marker gene expression in vitro. CONCLUSIONS: Together these results indicate that xenotropic MLV envelope proteins are sufficient to induce the production of factors by tumor cells that suppress vascular SMC differentiation, providing evidence for a novel mechanism by which xenotropic MLVs might alter tumor pathogenesis by disrupting tumor vascular maturation. Although it is highly unlikely that either XMRV or B4Rv themselves infect humans and are pathogenic, the results suggest that xenograft approaches commonly used in the study of human cancer promote the evolution of novel retroviruses with pathogenic properties.


Subject(s)
Gene Products, env/metabolism , Neovascularization, Pathologic , Prostatic Neoplasms/pathology , Prostatic Neoplasms/virology , Xenotropic murine leukemia virus-related virus/pathogenicity , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred BALB C
9.
Physiol Genomics ; 44(7): 417-29, 2012 Apr 02.
Article in English | MEDLINE | ID: mdl-22318995

ABSTRACT

Smooth muscle cell (SMC) phenotypic modulation in atherosclerosis and in response to PDGF in vitro involves repression of differentiation marker genes and increases in SMC proliferation, migration, and matrix synthesis. However, SMCs within atherosclerotic plaques can also express a number of proinflammatory genes, and in cultured SMCs the inflammatory cytokine IL-1ß represses SMC marker gene expression and induces inflammatory gene expression. Studies herein tested the hypothesis that IL-1ß modulates SMC phenotype to a distinct inflammatory state relative to PDGF-DD. Genome-wide gene expression analysis of IL-1ß- or PDGF-DD-treated SMCs revealed that although both stimuli repressed SMC differentiation marker gene expression, IL-1ß distinctly induced expression of proinflammatory genes, while PDGF-DD primarily induced genes involved in cell proliferation. Promoters of inflammatory genes distinctly induced by IL-1ß exhibited over-representation of NF-κB binding sites, and NF-κB inhibition in SMCs reduced IL-1ß-induced upregulation of proinflammatory genes as well as repression of SMC differentiation marker genes. Interestingly, PDGF-DD-induced SMC marker gene repression was not NF-κB dependent. Finally, immunofluorescent staining of mouse atherosclerotic lesions revealed the presence of cells positive for the marker of an IL-1ß-stimulated inflammatory SMC, chemokine (C-C motif) ligand 20 (CCL20), but not the PDGF-DD-induced gene, regulator of G protein signaling 17 (RGS17). Results demonstrate that IL-1ß- but not PDGF-DD-induced phenotypic modulation of SMC is characterized by NF-κB-dependent activation of proinflammatory genes, suggesting the existence of a distinct inflammatory SMC phenotype. In addition, studies provide evidence for the possible utility of CCL20 and RGS17 as markers of inflammatory and proliferative state SMCs within atherosclerotic plaques in vivo.


Subject(s)
Inflammation/genetics , Interleukin-1beta/pharmacology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Platelet-Derived Growth Factor/pharmacology , Actins/metabolism , Animals , Binding Sites , Biomarkers/metabolism , Cell Proliferation/drug effects , Cluster Analysis , Female , Gene Expression Regulation/drug effects , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Phenotype , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism
10.
J Am Soc Mass Spectrom ; 21(1): 80-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19850495

ABSTRACT

Peptide sequence identification using tandem mass spectroscopy remains a major challenge for complex proteomic studies. Peptide matching algorithms require the accurate determination of both the mass and charge of the precursor ion and accommodate uncertainties in these properties by using a wide precursor mass tolerance and by testing, for each spectrum, several possible candidate charges. Using a data acquisition strategy that includes obtaining narrow mass-range MS(1) "zoom" scans, we describe here a post-acquisition algorithm dubbed mass and charge (Z) inference engine (MAZIE), which accurately determines the charge and monoisotopic mass of precursor ions on a low-resolution Thermo LTQ-XL mass spectrometer. This is achieved by examining the isotopic distribution obtained in the preceding MS(1) zoom spectrum and comparing to theoretical distributions for candidate charge states from +1 to +4. MAZIE then writes modified data files with the corrected monoisotopic mass and charge. We have validated MAZIE results by comparing the sequence search results obtained with the MAZIE-generated data files to results using the unmodified data files. Using two different search algorithms and a false discovery rate filter, we found that MAZIE-interpreted data resulted in 80% (using SEQUEST) and 30% (using OMSSA) more high-confidence sequence identifications. Analyses of these results indicate that the accurate determination of the precursor ion mass greatly facilitates the ability to differentiate between true and false positive matches, while the determination of the precursor ion charge reduces the overall search time but does not significantly reduce the ambiguity of interpreting the search results. MAZIE is distributed as an open-source PERL script.


Subject(s)
Algorithms , Peptides/chemistry , Proteomics/methods , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Databases, Protein , Humans , Peptides/analysis , Sequence Analysis, Protein , Urine/chemistry
11.
Diagn Mol Pathol ; 12(2): 63-70, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12766610

ABSTRACT

The prognosis of men with moderate-grade prostate cancer is uncertain. At present, there are few if any reliable molecular markers that can distinguish moderate-grade tumors from those that behave more aggressively. To better understand the molecular basis of human prostate cancer and potentially provide information toward more accurate prognosis, we measured and analyzed gene expression profiles of 13 high- and moderate-grade human prostate tumors using cDNA microarrays. The expression of 136 genes was observed to differ significantly (P < 0.001) between normal prostate and tumors using one-sample t testing and Wilcoxon ranking. Hierarchical clustering of genes demonstrated a relatively similar pattern of differential expression across the tumors. However, importantly, permutation t tests (two-tailed P < 0.001) revealed 21 genes whose expression profiles segregated moderate- and high-grade tumors from each other, which was significantly (P < 0.03) greater than what was expected by chance. These results were compared in silico with prostate cancer profiling efforts performed by other groups, including a meta-analysis of four data sets, which validated many of the dysregulated genes. We suggest that these data provide insight into the molecular nature of clinically aggressive prostate cancer.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis/methods , Prostatic Neoplasms/genetics , Adenocarcinoma/classification , Adenocarcinoma/pathology , Biomarkers, Tumor , DNA, Complementary/analysis , DNA, Neoplasm/analysis , Diagnosis, Differential , Humans , Male , Prognosis , Prostatic Neoplasms/classification , Prostatic Neoplasms/pathology , RNA, Neoplasm/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...