Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Cell ; 170(6): 1149-1163.e12, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28886383

ABSTRACT

The diversity of mesenchymal cell types in the lung that influence epithelial homeostasis and regeneration is poorly defined. We used genetic lineage tracing, single-cell RNA sequencing, and organoid culture approaches to show that Lgr5 and Lgr6, well-known markers of stem cells in epithelial tissues, are markers of mesenchymal cells in the adult lung. Lgr6+ cells comprise a subpopulation of smooth muscle cells surrounding airway epithelia and promote airway differentiation of epithelial progenitors via Wnt-Fgf10 cooperation. Genetic ablation of Lgr6+ cells impairs airway injury repair in vivo. Distinct Lgr5+ cells are located in alveolar compartments and are sufficient to promote alveolar differentiation of epithelial progenitors through Wnt activation. Modulating Wnt activity altered differentiation outcomes specified by mesenchymal cells. This identification of region- and lineage-specific crosstalk between epithelium and their neighboring mesenchymal partners provides new understanding of how different cell types are maintained in the adult lung.


Subject(s)
Lung/cytology , Mesoderm/cytology , Animals , Homeostasis , Lung/physiology , Mice , Organoids/cytology , Pulmonary Alveoli/cytology , Receptors, G-Protein-Coupled/analysis , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic
2.
Cell ; 140(4): 460-76, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20178740

ABSTRACT

The growth of lymphatic vessels (lymphangiogenesis) is actively involved in a number of pathological processes including tissue inflammation and tumor dissemination but is insufficient in patients suffering from lymphedema, a debilitating condition characterized by chronic tissue edema and impaired immunity. The recent explosion of knowledge on the molecular mechanisms governing lymphangiogenesis provides new possibilities to treat these diseases.


Subject(s)
Lymphangiogenesis , Animals , Endothelial Cells/physiology , Humans , Inflammation/physiopathology , Lymphangiogenesis/drug effects , Lymphatic Vessels/physiology , Lymphatic Vessels/physiopathology , Neoplasms/physiopathology
3.
Nature ; 571(7765): 398-402, 2019 07.
Article in English | MEDLINE | ID: mdl-31292548

ABSTRACT

A decline in stem cell function impairs tissue regeneration during ageing, but the role of the stem-cell-supporting niche in ageing is not well understood. The small intestine is maintained by actively cycling intestinal stem cells that are regulated by the Paneth cell niche1,2. Here we show that the regenerative potential of human and mouse intestinal epithelium diminishes with age owing to defects in both stem cells and their niche. The functional decline was caused by a decrease in stemness-maintaining Wnt signalling due to production of Notum, an extracellular Wnt inhibitor, in aged Paneth cells. Mechanistically, high activity of mammalian target of rapamycin complex 1 (mTORC1) in aged Paneth cells inhibits activity of peroxisome proliferator activated receptor α (PPAR-α)3, and lowered PPAR-α activity increased Notum expression. Genetic targeting of Notum or Wnt supplementation restored function of aged intestinal organoids. Moreover, pharmacological inhibition of Notum in mice enhanced the regenerative capacity of aged stem cells and promoted recovery from chemotherapy-induced damage. Our results reveal a role of the stem cell niche in ageing and demonstrate that targeting of Notum can promote regeneration of aged tissues.


Subject(s)
Aging , Cellular Senescence , Esterases/metabolism , Intestinal Mucosa/pathology , Paneth Cells/metabolism , Regeneration , Aging/physiology , Animals , Cellular Senescence/physiology , Esterases/antagonists & inhibitors , Esterases/biosynthesis , Female , Humans , Intestinal Mucosa/physiology , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , PPAR alpha/metabolism , Paneth Cells/pathology , Receptors, G-Protein-Coupled/metabolism , Stem Cell Niche , Stem Cells/pathology , Wnt Proteins/antagonists & inhibitors , Wnt Signaling Pathway
4.
Immunity ; 43(3): 579-90, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26341400

ABSTRACT

Infiltration of regulatory T (Treg) cells into many tumor types correlates with poor patient prognoses. However, mechanisms of intratumoral Treg cell function remain to be elucidated. We investigated Treg cell function in a genetically engineered mouse model of lung adenocarcinoma and found that Treg cells suppressed anti-tumor responses in tumor-associated tertiary lymphoid structures (TA-TLSs). TA-TLSs have been described in human lung cancers, but their function remains to be determined. TLSs in this model were spatially associated with >90% of tumors and facilitated interactions between T cells and tumor-antigen-presenting dendritic cells (DCs). Costimulatory ligand expression by DCs and T cell proliferation rates increased in TA-TLSs upon Treg cell depletion, leading to tumor destruction. Thus, we propose that Treg cells in TA-TLSs can inhibit endogenous immune responses against tumors, and targeting these cells might provide therapeutic benefit for cancer patients.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Animals , Cell Proliferation , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lymphocyte Activation/immunology , Lymphocyte Depletion , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Transgenic , Microscopy, Confocal , Neoplasms/genetics , Neoplasms/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/metabolism
5.
Nature ; 545(7654): 355-359, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28489818

ABSTRACT

The heterogeneity of cellular states in cancer has been linked to drug resistance, cancer progression and the presence of cancer cells with properties of normal tissue stem cells. Secreted Wnt signals maintain stem cells in various epithelial tissues, including in lung development and regeneration. Here we show that mouse and human lung adenocarcinomas display hierarchical features with two distinct subpopulations, one with high Wnt signalling activity and another forming a niche that provides the Wnt ligand. The Wnt responder cells showed increased tumour propagation ability, suggesting that these cells have features of normal tissue stem cells. Genetic perturbation of Wnt production or signalling suppressed tumour progression. Small-molecule inhibitors targeting essential posttranslational modification of Wnt reduced tumour growth and markedly decreased the proliferative potential of lung cancer cells, leading to improved survival of tumour-bearing mice. These results indicate that strategies for disrupting pathways that maintain stem-like and niche cell phenotypes can translate into effective anti-cancer therapies.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Disease Progression , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Stem Cell Niche , Wnt Proteins/biosynthesis , Wnt Signaling Pathway , Adenocarcinoma of Lung , Animals , Cell Proliferation/drug effects , Female , Humans , Male , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Processing, Post-Translational/drug effects , Small Molecule Libraries/pharmacology , Survival Rate , Wnt Proteins/chemistry , Wnt Proteins/metabolism
6.
Proc Natl Acad Sci U S A ; 117(48): 30670-30678, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33199632

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is typically diagnosed at an advanced stage, which limits surgical options and portends a dismal prognosis. Current oncologic PDAC therapies confer marginal benefit and, thus, a significant unmet clinical need exists for new therapeutic strategies. To identify effective PDAC therapies, we leveraged a syngeneic orthotopic PDAC transplant mouse model to perform a large-scale, in vivo screen of 16 single-agent and 41 two-drug targeted therapy combinations in mice. Among 57 drug conditions screened, combined inhibition of heat shock protein (Hsp)-90 and MEK was found to produce robust suppression of tumor growth, leading to an 80% increase in the survival of PDAC-bearing mice with no significant toxicity. Mechanistically, we observed that single-agent MEK inhibition led to compensatory activation of resistance pathways, including components of the PI3K/AKT/mTOR signaling axis, which was overcome with the addition of HSP90 inhibition. The combination of HSP90(i) + MEK(i) was also active in vitro in established human PDAC cell lines and in vivo in patient-derived organoid PDAC transplant models. These findings encourage the clinical development of HSP90(i) + MEK(i) combination therapy and highlight the power of clinically relevant in vivo model systems for identifying cancer therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/therapeutic use , Benzodioxoles/pharmacology , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Drug Screening Assays, Antitumor/methods , Drug Synergism , Gene Expression , Humans , Immunohistochemistry , MAP Kinase Signaling System/drug effects , Mice , Molecular Targeted Therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Signal Transduction/drug effects , Survival Rate , Treatment Outcome , Xenograft Model Antitumor Assays
7.
Gastroenterology ; 160(1): 245-259, 2021 01.
Article in English | MEDLINE | ID: mdl-32941878

ABSTRACT

BACKGROUND & AIMS: Mutations in the APC gene and other genes in the Wnt signaling pathway contribute to development of colorectal carcinomas. R-spondins (RSPOs) are secreted proteins that amplify Wnt signaling in intestinal stem cells. Alterations in RSPO genes have been identified in human colorectal tumors. We studied the effects of RSPO1 overexpression in ApcMin/+ mutant mice. METHODS: An adeno associated viral vector encoding RSPO1-Fc fusion protein, or control vector, was injected into ApcMin/+mice. Their intestinal crypts were isolated and cultured as organoids. which were incubated with or without RSPO1-Fc and an inhibitor of transforming growth factor beta receptor (TGFBR). Livers were collected from mice and analyzed by immunohistochemistry. Organoids and adenomas were analyzed by quantitative reverse-transcription PCR, single cell RNA sequencing, and immunohistochemistry. RESULTS: Intestines from Apc+/+ mice injected with the vector encoding RSPO1-Fc had significantly deeper crypts, longer villi, with increased EdU labeling, indicating increased proliferation of epithelial cells, in comparison to mice given control vector. AAV-RSPO1-Fc-transduced ApcMin/+ mice also developed fewer and smaller intestinal tumors and had significantly longer survival times. Adenomas of ApcMin/+ mice injected with the RSPO1-Fc vector showed a rapid increase in apoptosis and in the expression of Wnt target genes, followed by reduced expression of messenger RNAs and proteins regulated by the Wnt pathway, reduced cell proliferation, and less crypt branching than adenomas of mice given the control vector. Addition of RSPO1 reduced the number of adenoma organoids derived from ApcMin/+ mice and suppressed expression of Wnt target genes but increased phosphorylation of SMAD2 and transcription of genes regulated by SMAD. Inhibition of TGFBR signaling in organoids stimulated with RSPO1-Fc restored organoid formation and expression of genes regulated by Wnt. The TGFBR inhibitor restored apoptosis in adenomas from ApcMin/+ mice expressing RSPO1-Fc back to the same level as in the adenomas from mice given the control vector. CONCLUSIONS: Expression of RSPO1 in ApcMin/+ mice increases apoptosis and reduces proliferation and Wnt signaling in adenoma cells, resulting in development of fewer and smaller intestinal tumors and longer mouse survival. Addition of RSPO1 to organoids derived from adenomas inhibits their growth and promotes proliferation of intestinal stem cells that retain the APC protein; these effects are reversed by TGFB inhibitor. Strategies to increase the expression of RSPO1 might be developed for the treatment of intestinal adenomas.


Subject(s)
Adenoma/pathology , Intestinal Neoplasms/pathology , Thrombospondins/metabolism , Transforming Growth Factor beta/physiology , Wnt Signaling Pathway/physiology , Adenoma/etiology , Animals , Disease Models, Animal , Intestinal Neoplasms/etiology , Mice , Organoids
8.
Proc Natl Acad Sci U S A ; 116(6): 2232-2236, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30674677

ABSTRACT

Circulating tumor cells (CTCs) play a fundamental role in cancer progression. However, in mice, limited blood volume and the rarity of CTCs in the bloodstream preclude longitudinal, in-depth studies of these cells using existing liquid biopsy techniques. Here, we present an optofluidic system that continuously collects fluorescently labeled CTCs from a genetically engineered mouse model (GEMM) for several hours per day over multiple days or weeks. The system is based on a microfluidic cell sorting chip connected serially to an unanesthetized mouse via an implanted arteriovenous shunt. Pneumatically controlled microfluidic valves capture CTCs as they flow through the device, and CTC-depleted blood is returned back to the mouse via the shunt. To demonstrate the utility of our system, we profile CTCs isolated longitudinally from animals over 4 days of treatment with the BET inhibitor JQ1 using single-cell RNA sequencing (scRNA-Seq) and show that our approach eliminates potential biases driven by intermouse heterogeneity that can occur when CTCs are collected across different mice. The CTC isolation and sorting technology presented here provides a research tool to help reveal details of how CTCs evolve over time, allowing studies to credential changes in CTCs as biomarkers of drug response and facilitating future studies to understand the role of CTCs in metastasis.


Subject(s)
Flow Cytometry , Microfluidic Analytical Techniques , Microfluidics , Neoplasms/diagnosis , Neoplasms/metabolism , Neoplastic Cells, Circulating/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Flow Cytometry/methods , Gene Expression Profiling/methods , Mice , Microfluidics/methods , Neoplasms/genetics , Neoplastic Cells, Circulating/pathology , Single-Cell Analysis/methods , Transcriptome
9.
Nature ; 516(7531): 428-31, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25337879

ABSTRACT

Cancer is a multistep process that involves mutations and other alterations in oncogenes and tumour suppressor genes. Genome sequencing studies have identified a large collection of genetic alterations that occur in human cancers. However, the determination of which mutations are causally related to tumorigenesis remains a major challenge. Here we describe a novel CRISPR/Cas9-based approach for rapid functional investigation of candidate genes in well-established autochthonous mouse models of cancer. Using a Kras(G12D)-driven lung cancer model, we performed functional characterization of a panel of tumour suppressor genes with known loss-of-function alterations in human lung cancer. Cre-dependent somatic activation of oncogenic Kras(G12D) combined with CRISPR/Cas9-mediated genome editing of tumour suppressor genes resulted in lung adenocarcinomas with distinct histopathological and molecular features. This rapid somatic genome engineering approach enables functional characterization of putative cancer genes in the lung and other tissues using autochthonous mouse models. We anticipate that this approach can be used to systematically dissect the complex catalogue of mutations identified in cancer genome sequencing studies.


Subject(s)
Adenocarcinoma/genetics , Gene Editing/methods , Lung Neoplasms/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , CRISPR-Associated Proteins , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Disease Models, Animal , Genes, Tumor Suppressor , Genome , Humans , Lentivirus/genetics , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Models, Genetic , Mutation/genetics
10.
Nature ; 514(7522): 380-4, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25119044

ABSTRACT

The study of cancer genes in mouse models has traditionally relied on genetically-engineered strains made via transgenesis or gene targeting in embryonic stem cells. Here we describe a new method of cancer model generation using the CRISPR/Cas (clustered regularly interspaced short palindromic repeats/CRISPR-associated proteins) system in vivo in wild-type mice. We used hydrodynamic injection to deliver a CRISPR plasmid DNA expressing Cas9 and single guide RNAs (sgRNAs) to the liver that directly target the tumour suppressor genes Pten (ref. 5) and p53 (also known as TP53 and Trp53) (ref. 6), alone and in combination. CRISPR-mediated Pten mutation led to elevated Akt phosphorylation and lipid accumulation in hepatocytes, phenocopying the effects of deletion of the gene using Cre-LoxP technology. Simultaneous targeting of Pten and p53 induced liver tumours that mimicked those caused by Cre-loxP-mediated deletion of Pten and p53. DNA sequencing of liver and tumour tissue revealed insertion or deletion mutations of the tumour suppressor genes, including bi-allelic mutations of both Pten and p53 in tumours. Furthermore, co-injection of Cas9 plasmids harbouring sgRNAs targeting the ß-catenin gene and a single-stranded DNA oligonucleotide donor carrying activating point mutations led to the generation of hepatocytes with nuclear localization of ß-catenin. This study demonstrates the feasibility of direct mutation of tumour suppressor genes and oncogenes in the liver using the CRISPR/Cas system, which presents a new avenue for rapid development of liver cancer models and functional genomics.


Subject(s)
CRISPR-Cas Systems , Genes, Tumor Suppressor , Genetic Engineering/methods , Liver/metabolism , Mutagenesis/genetics , Mutation/genetics , Oncogenes/genetics , Animals , Base Sequence , Cell Transformation, Neoplastic/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Female , Genes, p53/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Lipid Metabolism , Liver/cytology , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Molecular Sequence Data , PTEN Phosphohydrolase/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/genetics
11.
Circ Res ; 120(9): 1414-1425, 2017 Apr 28.
Article in English | MEDLINE | ID: mdl-28298294

ABSTRACT

RATIONALE: Vascular endothelial growth factor (VEGF) is the main driver of angiogenesis and vascular permeability via VEGF receptor 2 (VEGFR2), whereas lymphangiogenesis signals are transduced by VEGFC/D via VEGFR3. VEGFR3 also regulates sprouting angiogenesis and blood vessel growth, but to what extent VEGFR3 signaling controls blood vessel permeability remains unknown. OBJECTIVE: To investigate the role of VEGFR3 in the regulation of VEGF-induced vascular permeability. METHODS AND RESULTS: Long-term global Vegfr3 gene deletion in adult mice resulted in increased fibrinogen deposition in lungs and kidneys, indicating enhanced vascular leakage at the steady state. Short-term deletion of Vegfr3 in blood vascular endothelial cells increased baseline leakage in various tissues, as well as in tumors, and exacerbated vascular permeability in response to VEGF, administered via intradermal adenoviral delivery or through systemic injection of recombinant protein. VEGFR3 gene silencing upregulated VEGFR2 protein levels and phosphorylation in cultured endothelial cells. Consistent with elevated VEGFR2 activity, vascular endothelial cadherin showed reduced localization at endothelial cell-cell junctions in postnatal retinas after Vegfr3 deletion, or after VEGFR3 silencing in cultured endothelial cells. Furthermore, concurrent deletion of Vegfr2 prevented VEGF-induced excessive vascular leakage in mice lacking Vegfr3. CONCLUSIONS: VEGFR3 limits VEGFR2 expression and VEGF/VEGFR2 pathway activity in quiescent and angiogenic blood vascular endothelial cells, thereby preventing excessive vascular permeability.


Subject(s)
Capillary Permeability , Carcinoma, Lewis Lung/blood supply , Endothelial Cells/metabolism , Lung/blood supply , Retinal Vessels/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Adherens Junctions/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability/drug effects , Carcinoma, Lewis Lung/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Female , Genotype , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic , Neovascularization, Physiologic , Phenotype , Retinal Vessels/drug effects , Signal Transduction/drug effects , Tight Junctions/metabolism , Transfection , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/deficiency , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-3/deficiency , Vascular Endothelial Growth Factor Receptor-3/genetics
12.
Genes Dev ; 25(8): 831-44, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21498572

ABSTRACT

Neural stem cells (NSCs) are slowly dividing astrocytes that are intimately associated with capillary endothelial cells in the subventricular zone (SVZ) of the brain. Functionally, members of the vascular endothelial growth factor (VEGF) family can stimulate neurogenesis as well as angiogenesis, but it has been unclear whether they act directly via VEGF receptors (VEGFRs) expressed by neural cells, or indirectly via the release of growth factors from angiogenic capillaries. Here, we show that VEGFR-3, a receptor required for lymphangiogenesis, is expressed by NSCs and is directly required for neurogenesis. Vegfr3:YFP reporter mice show VEGFR-3 expression in multipotent NSCs, which are capable of self-renewal and are activated by the VEGFR-3 ligand VEGF-C in vitro. Overexpression of VEGF-C stimulates VEGFR-3-expressing NSCs and neurogenesis in the SVZ without affecting angiogenesis. Conversely, conditional deletion of Vegfr3 in neural cells, inducible deletion in subventricular astrocytes, and blocking of VEGFR-3 signaling with antibodies reduce SVZ neurogenesis. Therefore, VEGF-C/VEGFR-3 signaling acts directly on NSCs and regulates adult neurogenesis, opening potential approaches for treatment of neurodegenerative diseases.


Subject(s)
Neurogenesis/physiology , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Lymphangiogenesis/genetics , Lymphangiogenesis/physiology , Mice , Mice, Mutant Strains , Microscopy, Electron, Transmission , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-3/genetics
13.
Proc Natl Acad Sci U S A ; 117(49): 30876-30878, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33199634

Subject(s)
Integrases , Animals , Mice , Software
14.
Proc Natl Acad Sci U S A ; 111(34): E3553-61, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25114235

ABSTRACT

MicroRNAs (miRNAs) and siRNAs have enormous potential as cancer therapeutics, but their effective delivery to most solid tumors has been difficult. Here, we show that a new lung-targeting nanoparticle is capable of delivering miRNA mimics and siRNAs to lung adenocarcinoma cells in vitro and to tumors in a genetically engineered mouse model of lung cancer based on activation of oncogenic Kirsten rat sarcoma viral oncogene homolog (Kras) and loss of p53 function. Therapeutic delivery of miR-34a, a p53-regulated tumor suppressor miRNA, restored miR-34a levels in lung tumors, specifically down-regulated miR-34a target genes, and slowed tumor growth. The delivery of siRNAs targeting Kras reduced Kras gene expression and MAPK signaling, increased apoptosis, and inhibited tumor growth. The combination of miR-34a and siRNA targeting Kras improved therapeutic responses over those observed with either small RNA alone, leading to tumor regression. Furthermore, nanoparticle-mediated small RNA delivery plus conventional, cisplatin-based chemotherapy prolonged survival in this model compared with chemotherapy alone. These findings demonstrate that RNA combination therapy is possible in an autochthonous model of lung cancer and provide preclinical support for the use of small RNA therapies in patients who have cancer.


Subject(s)
Lung Neoplasms/therapy , MicroRNAs/therapeutic use , RNA, Small Interfering/therapeutic use , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/therapy , Animals , Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Cisplatin/administration & dosage , Combined Modality Therapy , Gene Expression , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MAP Kinase Signaling System , Mice , Mice, Knockout , Mice, Transgenic , MicroRNAs/administration & dosage , MicroRNAs/genetics , Mutation , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Nanotechnology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , ras Proteins/genetics
15.
Blood ; 122(5): 658-65, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23803710

ABSTRACT

Vascular bypass procedures in the central nervous system (CNS) remain technically challenging, hindered by complications and often failing to prevent adverse outcome such as stroke. Thus, there is an unmet clinical need for a safe and effective CNS revascularization. Vascular endothelial growth factors (VEGFs) are promising candidates for revascularization; however, their effects appear to be tissue-specific and their potential in the CNS has not been fully explored. To test growth factors for angiogenesis in the CNS, we characterized the effects of endothelium-specific growth factors on the brain vasculature and parenchyma. Recombinant adeno-associated virus (AAV) vectors encoding the growth factors were injected transcranially to the frontoparietal cerebrum of mice. Angiogenesis, mural cell investment, leukocyte recruitment, vascular permeability, reactive gliosis and neuronal patterning were evaluated by 3-dimensional immunofluorescence, electron microscopy, optical projection tomography, and magnetic resonance imaging. Placenta growth factor (PlGF) stimulated robust angiogenesis and arteriogenesis without significant side effects, whereas VEGF and VEGF-C incited growth of aberrant vessels, severe edema, and inflammation. VEGF-B, angiopoietin-1, angiopoietin-2, and a VEGF/angiopoietin-1 chimera had minimal effects on the brain vessels or parenchyma. Of the growth factors tested, PlGF emerged as the most efficient and safe angiogenic factor, hence making it a candidate for therapeutic CNS revascularization.


Subject(s)
Central Nervous System/blood supply , Cerebral Revascularization , Intercellular Signaling Peptides and Proteins/physiology , Pregnancy Proteins/physiology , Animals , Blood Vessels/growth & development , Blood Vessels/metabolism , Central Nervous System Neoplasms/etiology , Central Nervous System Neoplasms/genetics , Encephalitis/etiology , Encephalitis/genetics , Female , Genetic Therapy/methods , Hemangioma/etiology , Hemangioma/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/genetics , Placenta Growth Factor , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Pregnancy Proteins/therapeutic use , Vascular Endothelial Growth Factor A/adverse effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor C/adverse effects , Vascular Endothelial Growth Factor C/genetics
16.
J Surg Res ; 196(1): 200-7, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25777822

ABSTRACT

BACKGROUND: Lymphedema still remains an unsolved problem. Secondary lymphedema often develops after cancer operations or radiation therapy, especially in breast cancer patients. Using a mouse model, we show here that the lymphatic network can be regenerated using lymphatic vascular growth factor therapy in combination with lymph node transfer. MATERIALS AND METHODS: We have compared the therapeutic effects of different vascular endothelial growth factors (VEGF-C, VEGF-D, VEGF-C156S, and VEGF-A), in combination with lymph node transfer in mouse axilla. The lymphangiogenic effects of the growth factor therapy were examined at 3 mo postoperatively. RESULTS: VEGF therapy with VEGF-C and VEGF-D induced growth of new lymphatic vessels in the defect area, and VEGF-C also improved lymphatic vessel function compared with that of controls. VEGF-C156S induced moderate lymphangiogenesis, but the effect remained statistically nonsignificant. Prolymphangiogenic growth factors (VEGF-C, -D, and -C156S) also improved lymph node survival as compared with those of the VEGF-A and control group. VEGF-C, which activates both vascular endothelial growth factor receptor 2 and vascular endothelial growth factor receptor 3, gave the best therapeutic effect in this experimental lymphedema model. CONCLUSIONS: These results support our goal to treat secondary lymphedema by combining lymph node transfer with the growth factor therapy. VEGF-C provides the preferred alternative for growth factor therapy of lymphedema when compared with other VEGF-family growth factors, due to the superior lymphangiogenic response and minor blood vascular effects.


Subject(s)
Lymph Nodes/transplantation , Lymphedema/therapy , Vascular Endothelial Growth Factor A/therapeutic use , Animals , Lymphangiogenesis , Lymphedema/physiopathology , Lymphography , Mice , Vascular Endothelial Growth Factor Receptor-2/physiology , Vascular Endothelial Growth Factor Receptor-3/physiology
17.
Proc Natl Acad Sci U S A ; 109(16): 6223-8, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22474390

ABSTRACT

Lymphatic vessel growth or lymphangiogenesis occurs during embryonic development and wound healing and plays an important role in tumor metastasis and inflammatory diseases. However, the possibility of noninvasive detection and quantification of lymphangiogenesis has been lacking. Here, we present the Vegfr3(EGFPLuc) mouse model, where an EGFP-luciferase fusion protein, expressed under the endogenous transcriptional control of the Vegfr3 gene, allows the monitoring of physiological and pathological lymphangiogenesis in vivo. We show tracking of lymphatic vessel development during embryogenesis as well as lymphangiogenesis induced by specific growth factors, during wound healing and in contact hypersensitivity (CHS)--induced inflammation where we also monitor down-regulation of lymphangiogenesis by the glucocorticoid dexamethasone. Importantly, the Vegfr3-reporter allowed us to tracking tumor-induced lymphangiogenesis at the tumor periphery and in lymph nodes in association with the metastatic process. This is the first reporter mouse model for luminescence imaging of lymphangiogenesis. It should provide an important tool for studying the involvement of lymphangiogenesis in pathological processes.


Subject(s)
Diagnostic Imaging/methods , Inflammation/metabolism , Lymphatic Vessels/metabolism , Wound Healing , Animals , Cell Line, Tumor , Dexamethasone/pharmacology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Glucocorticoids/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Inflammation/genetics , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements/methods , Lymphangiogenesis/drug effects , Lymphatic Metastasis , Lymphatic Vessels/embryology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Time Factors , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
18.
Circulation ; 127(4): 424-34, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23357661

ABSTRACT

BACKGROUND: There is an unmet need for proangiogenic therapeutic molecules for the treatment of tissue ischemia in cardiovascular diseases. However, major inducers of angiogenesis such as vascular endothelial growth factor (VEGF/VEGF-A) have side effects that limit their therapeutic utility in vivo, especially at high concentrations. Angiopoietin-1 has been considered to be a blood vessel stabilization factor that can inhibit the intrinsic property of VEGF to promote vessel leakiness. In this study, we have designed and tested the angiogenic properties of chimeric molecules consisting of receptor-binding parts of VEGF and angiopoietin-1. We aimed at combining the activities of both factors into 1 molecule for easy delivery and expression in target tissues. METHODS AND RESULTS: The VEGF-angiopoietin-1 (VA1) chimeric protein bound to both VEGF receptor-2 and Tie2 and induced the activation of both receptors. Detailed analysis of VA1 versus VEGF revealed differences in the kinetics of VEGF receptor-2 activation and endocytosis, downstream kinase activation, and VE-cadherin internalization. The delivery of a VA1 transgene into mouse skeletal muscle led to increased blood flow and enhanced angiogenesis. VA1 was also very efficient in rescuing ischemic limb perfusion. However, VA1 induced less plasma protein leakage and myeloid inflammatory cell recruitment than VEGF. Furthermore, angioma-like structures associated with VEGF expression were not observed with VA1. CONCLUSIONS: The VEGF-angiopoietin-1 chimera is a potent angiogenic factor that triggers a novel mode of VEGF receptor-2 activation, promoting less vessel leakiness, less tissue inflammation, and better perfusion in ischemic muscle than VEGF. These properties of VA1 make it an attractive therapeutic tool.


Subject(s)
Angiopoietin-1/pharmacology , Genetic Therapy/methods , Ischemia/drug therapy , Neovascularization, Physiologic/physiology , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , Adenoviridae/genetics , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Capillary Permeability/physiology , Cell Line, Tumor , Disease Models, Animal , Female , HEK293 Cells , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Ischemia/genetics , Leukemia, Myeloid , Mice , Mice, Inbred Strains , Muscle, Skeletal/blood supply , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, TIE-2 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
J Cell Sci ; 125(Pt 9): 2212-23, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22357955

ABSTRACT

Angiopoietin 1 (Ang1) is an activating ligand for the endothelial receptor tyrosine kinase Tie2, whereas Ang2 acts as a context-dependent agonist or antagonist that has a destabilizing effect on the vasculature. The molecular mechanisms responsible for the versatile functions of Ang2 are poorly understood. We show here that Ang2, but not Ang1, induces Tie2 translocation to the specific cell-matrix contact sites located at the distal end of focal adhesions. The Ang2-specific Tie2 translocation was associated with distinct Tie2 activation and downstream signals which differed from those of Ang1, and led to impaired cell motility and weak cell-matrix adhesion. We demonstrate that the different oligomeric or multimeric forms of the angiopoietins induce distinct patterns of Tie2 trafficking; the lower oligomerization state of native Ang2 was crucial for the Ang2-specific Tie2 redistribution, whereas multimeric structures of Ang1 and Ang2 induced similar responses. The Ang2-specific Tie2 trafficking to cell-matrix contacts was also dependent on the cell substratum, α2ß1-integrin-containing cell-matrix adhesion sites and intact microtubules. Our data indicate that the different subcellular trafficking of Tie2-Ang2 and Tie2-Ang1 complexes generates ligand-specific responses in the angiopoietin-Tie signaling pathway, including modulation of cell-matrix interactions.


Subject(s)
Angiopoietin-1/chemistry , Angiopoietin-2/chemistry , Endothelium, Vascular/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Vitreous Body/blood supply , Angiopoietin-1/genetics , Angiopoietin-1/pharmacology , Angiopoietin-2/genetics , Angiopoietin-2/pharmacology , Animals , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell-Matrix Junctions/drug effects , Cell-Matrix Junctions/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Gene Expression/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Integrin alpha2beta1/genetics , Integrin alpha2beta1/metabolism , Intravitreal Injections , Mice , Microtubules/drug effects , Microtubules/metabolism , NIH 3T3 Cells , Neovascularization, Physiologic , Protein Multimerization , Receptor Protein-Tyrosine Kinases/genetics , Receptor, TIE-2 , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects
20.
Nature ; 454(7204): 656-60, 2008 Jul 31.
Article in English | MEDLINE | ID: mdl-18594512

ABSTRACT

Angiogenesis, the growth of new blood vessels from pre-existing vasculature, is a key process in several pathological conditions, including tumour growth and age-related macular degeneration. Vascular endothelial growth factors (VEGFs) stimulate angiogenesis and lymphangiogenesis by activating VEGF receptor (VEGFR) tyrosine kinases in endothelial cells. VEGFR-3 (also known as FLT-4) is present in all endothelia during development, and in the adult it becomes restricted to the lymphatic endothelium. However, VEGFR-3 is upregulated in the microvasculature of tumours and wounds. Here we demonstrate that VEGFR-3 is highly expressed in angiogenic sprouts, and genetic targeting of VEGFR-3 or blocking of VEGFR-3 signalling with monoclonal antibodies results in decreased sprouting, vascular density, vessel branching and endothelial cell proliferation in mouse angiogenesis models. Stimulation of VEGFR-3 augmented VEGF-induced angiogenesis and sustained angiogenesis even in the presence of VEGFR-2 (also known as KDR or FLK-1) inhibitors, whereas antibodies against VEGFR-3 and VEGFR-2 in combination resulted in additive inhibition of angiogenesis and tumour growth. Furthermore, genetic or pharmacological disruption of the Notch signalling pathway led to widespread endothelial VEGFR-3 expression and excessive sprouting, which was inhibited by blocking VEGFR-3 signals. Our results implicate VEGFR-3 as a regulator of vascular network formation. Targeting VEGFR-3 may provide additional efficacy for anti-angiogenic therapies, especially towards vessels that are resistant to VEGF or VEGFR-2 inhibitors.


Subject(s)
Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Dipeptides/pharmacology , Down-Regulation , Endothelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms/drug therapy , Neovascularization, Pathologic/genetics , Receptors, Notch/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL