Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Commun Med (Lond) ; 4(1): 38, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38499690

ABSTRACT

BACKGROUND: Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a familial cardiac disease associated with ventricular arrhythmias and an increased risk of sudden cardiac death. Currently, there are no approved treatments that address the underlying genetic cause of this disease, representing a significant unmet need. Mutations in Plakophilin-2 (PKP2), encoding a desmosomal protein, account for approximately 40% of ARVC cases and result in reduced gene expression. METHODS: Our goal is to examine the feasibility and the efficacy of adeno-associated virus 9 (AAV9)-mediated restoration of PKP2 expression in a cardiac specific knock-out mouse model of Pkp2. RESULTS: We show that a single dose of AAV9:PKP2 gene delivery prevents disease development before the onset of cardiomyopathy and attenuates disease progression after overt cardiomyopathy. Restoration of PKP2 expression leads to a significant extension of lifespan by restoring cellular structures of desmosomes and gap junctions, preventing or halting decline in left ventricular ejection fraction, preventing or reversing dilation of the right ventricle, ameliorating ventricular arrhythmia event frequency and severity, and preventing adverse fibrotic remodeling. RNA sequencing analyses show that restoration of PKP2 expression leads to highly coordinated and durable correction of PKP2-associated transcriptional networks beyond desmosomes, revealing a broad spectrum of biological perturbances behind ARVC disease etiology. CONCLUSIONS: We identify fundamental mechanisms of PKP2-associated ARVC beyond disruption of desmosome function. The observed PKP2 dose-function relationship indicates that cardiac-selective AAV9:PKP2 gene therapy may be a promising therapeutic approach to treat ARVC patients with PKP2 mutations.


Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a heart disease that leads to abnormal heartbeats and a higher risk of sudden cardiac death. ARVC is often caused by changes in a gene called PKP2, that then makes less PKP2 protein. PKP2 protein is important for the normal structure and function of the heart. Human ARVC characteristics can be mimicked in a mouse model missing this gene. Given no therapeutic option, our goal was to test if adding a working copy of PKP2 gene in the heart of this mouse model, using a technique called gene therapy that can deliver genes to cells, could improve heart function. Here, we show that a single dose of PKP2 gene therapy can improve heart function and heartbeats as well as extend lifespan in mice. PKP2 gene therapy may be a promising approach to treat ARVC patients with PKP2 mutations.

2.
Nat Commun ; 15(1): 1352, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38409164

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) poses therapeutic challenges due to the limited treatment options. Building upon our previous research that demonstrates the efficacy of histone deacetylase 6 (HDAC6) inhibition in a genetic cardiomyopathy model, we investigate HDAC6's role in HFpEF due to their shared mechanisms of inflammation and metabolism. Here, we show that inhibiting HDAC6 with TYA-018 effectively reverses established heart failure and its associated symptoms in male HFpEF mouse models. Additionally, in male mice lacking Hdac6 gene, HFpEF progression is delayed and they are resistant to TYA-018's effects. The efficacy of TYA-018 is comparable to a sodium-glucose cotransporter 2 (SGLT2) inhibitor, and the combination shows enhanced effects. Mechanistically, TYA-018 restores gene expression related to hypertrophy, fibrosis, and mitochondrial energy production in HFpEF heart tissues. Furthermore, TYA-018 also inhibits activation of human cardiac fibroblasts and enhances mitochondrial respiratory capacity in cardiomyocytes. In this work, our findings show that HDAC6 impacts on heart pathophysiology and is a promising target for HFpEF treatment.


Subject(s)
Cardiomyopathies , Heart Failure , Animals , Humans , Male , Mice , Heart Failure/drug therapy , Heart Failure/genetics , Heart Failure/diagnosis , Histone Deacetylase 6/genetics , Myocytes, Cardiac/metabolism , Stroke Volume/physiology
3.
Circulation ; 148(14): 1099-1112, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37602409

ABSTRACT

BACKGROUND: Cardiac reprogramming is a technique to directly convert nonmyocytes into myocardial cells using genes or small molecules. This intervention provides functional benefit to the rodent heart when delivered at the time of myocardial infarction or activated transgenically up to 4 weeks after myocardial infarction. Yet, several hurdles have prevented the advancement of cardiac reprogramming for clinical use. METHODS: Through a combination of screening and rational design, we identified a cardiac reprogramming cocktail that can be encoded in a single adeno-associated virus. We also created a novel adeno-associated virus capsid that can transduce cardiac fibroblasts more efficiently than available parental serotypes by mutating posttranslationally modified capsid residues. Because a constitutive promoter was needed to drive high expression of these cell fate-altering reprogramming factors, we included binding sites to a cardiomyocyte-restricted microRNA within the 3' untranslated region of the expression cassette that limits expression to nonmyocytes. After optimizing this expression cassette to reprogram human cardiac fibroblasts into induced cardiomyocyte-like cells in vitro, we also tested the ability of this capsid/cassette combination to confer functional benefit in acute mouse myocardial infarction and chronic rat myocardial infarction models. RESULTS: We demonstrated sustained, dose-dependent improvement in cardiac function when treating a rat model 2 weeks after myocardial infarction, showing that cardiac reprogramming, when delivered in a single, clinically relevant adeno-associated virus vector, can support functional improvement in the postremodeled heart. This benefit was not observed with GFP (green fluorescent protein) or a hepatocyte reprogramming cocktail and was achieved even in the presence of immunosuppression, supporting myocyte formation as the underlying mechanism. CONCLUSIONS: Collectively, these results advance the application of cardiac reprogramming gene therapy as a viable therapeutic approach to treat chronic heart failure resulting from ischemic injury.


Subject(s)
MicroRNAs , Myocardial Infarction , Rats , Mice , Humans , Animals , Dependovirus/genetics , Myocytes, Cardiac/metabolism , Myocardial Infarction/therapy , Myocardial Infarction/drug therapy , MicroRNAs/genetics , MicroRNAs/metabolism , Genetic Therapy/methods , Green Fluorescent Proteins/genetics , Cellular Reprogramming , Fibroblasts/metabolism
4.
PLoS One ; 15(3): e0229445, 2020.
Article in English | MEDLINE | ID: mdl-32160239

ABSTRACT

The Wnt/ß-catenin signaling pathway has been implicated in human proliferative diseases such as cancer and fibrosis. The functions of ß-catenin and several other components of this pathway have been investigated in fibrosis. However, the potential role of R-spondin proteins (RSPOs), enhancers of the Wnt/ß-catenin signaling, has not been described. A specific interventional strategy targeting this pathway for fibrosis remains to be defined. We developed monoclonal antibodies against members of the RSPO family (RSPO1, 2, and 3) and probed their potential function in fibrosis in vivo. We demonstrated that RSPO3 plays a critical role in the development of fibrosis in multiple organs. Specifically, an anti-RSPO3 antibody, OMP-131R10, when dosed therapeutically, attenuated fibrosis in carbon tetrachloride (CCl4)-induced liver fibrosis, bleomycin-induced pulmonary and skin fibrosis models. Mechanistically, we showed that RSPO3 induces multiple pro-fibrotic chemokines and cytokines in Kupffer cells and hepatocytes. We found that the anti-fibrotic activity of OMP-131R10 is associated with its inhibition of ß-catenin activation in vivo. Finally, RSPO3 was found to be highly elevated in the active lesions of fibrotic tissues in mouse models of fibrosis and in patients with idiopathic pulmonary fibrosis (IPF) and nonalcoholic steatohepatitis (NASH). Together these data provide an anti-fibrotic strategy for targeting the Wnt/ß-catenin pathway through RSPO3 blockade and support that OMP-131R10 could be an important therapeutic agent for fibrosis.


Subject(s)
Antibodies/therapeutic use , Idiopathic Pulmonary Fibrosis , Non-alcoholic Fatty Liver Disease , Thrombospondins/physiology , Animals , Cells, Cultured , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/metabolism , Male , Mice , Mice, Inbred DBA , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Wnt Signaling Pathway/drug effects
5.
J Exp Bot ; 70(5): 1627-1638, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30843586

ABSTRACT

The Arabidopsis plasma membrane-localized resistance protein RPM1 is degraded upon the induction of the hypersensitive response (HR) triggered in response to its own activation or that of other unrelated resistance (R) proteins. We investigated the role of RPM1 turnover in RPM1-mediated resistance and showed that degradation of RPM1 is not associated with HR or resistance mediated by this R protein. Likewise, the runaway cell death phenotype in the lsd1 mutant was not associated with RPM1 degradation and did not alter RPM1-derived resistance. RPM1 stability and RPM1-mediated resistance were dependent on the double-stranded RNA binding (DRB) proteins 1 and 4. Interestingly, the function of DRB1 in RPM1-mediated resistance was not associated with its role in pre-miRNA processing. The DRB3 and DRB5 proteins negatively regulated RPM1-mediated resistance and a mutation in these completely or partially restored resistance in the drb1, drb2, and drb4 mutant backgrounds. Conversely, plants overexpressing DRB5 showed attenuated RPM1-mediated resistance. A similar role for DRBs in basal and R-mediated resistance suggests that these proteins play a general role in bacterial resistance.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis/genetics , Plant Diseases/genetics , Pseudomonas syringae/physiology , RNA-Binding Proteins/genetics , Arabidopsis Proteins/metabolism , Disease Resistance/genetics , RNA-Binding Proteins/metabolism
6.
PLoS Pathog ; 14(3): e1006894, 2018 03.
Article in English | MEDLINE | ID: mdl-29513740

ABSTRACT

The E3 ubiquitin ligase COP1 (Constitutive Photomorphogenesis 1) is a well known component of the light-mediated plant development that acts as a repressor of photomorphogenesis. Here we show that COP1 positively regulates defense against turnip crinkle virus (TCV) and avrRPM1 bacteria by contributing to stability of resistance (R) protein HRT and RPM1, respectively. HRT and RPM1 levels and thereby pathogen resistance is significantly reduced in the cop1 mutant background. Notably, the levels of at least two double-stranded RNA binding (DRB) proteins DRB1 and DRB4 are reduced in the cop1 mutant background suggesting that COP1 affects HRT stability via its effect on the DRB proteins. Indeed, a mutation in either drb1 or drb4 resulted in degradation of HRT. In contrast to COP1, a multi-subunit E3 ligase encoded by anaphase-promoting complex (APC) 10 negatively regulates DRB4 and TCV resistance but had no effect on DRB1 levels. We propose that COP1-mediated positive regulation of HRT is dependent on a balance between COP1 and negative regulators that target DRB1 and DRB4.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/immunology , Carmovirus/immunology , Disease Resistance/immunology , Plant Diseases/immunology , RNA-Binding Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Arabidopsis/metabolism , Arabidopsis/virology , Arabidopsis Proteins/genetics , Gene Expression Regulation, Plant , Light , Morphogenesis , Mutation , Plant Development , Plant Diseases/virology , Nicotiana/immunology , Nicotiana/virology , Ubiquitin-Protein Ligases/genetics
7.
Sci Rep ; 7(1): 15270, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29127379

ABSTRACT

Activating mutations in the Wnt pathway are a characteristic feature of colorectal cancer (CRC). The R-spondin (RSPO) family is a group of secreted proteins that enhance Wnt signaling and RSPO2 and RSPO3 gene fusions have been reported in CRC. We have previously shown that Wnt pathway blockers exhibit potent combinatorial activity with taxanes to inhibit tumor growth. Here we show that RSPO3 antagonism synergizes with paclitaxel based chemotherapies in patient-derived xenograft models (PDX) with RSPO3 fusions and in tumors with common CRC mutations such as APC, ß-catenin, or RNF43. In these latter types of tumors that represent over 90% of CRC, RSPO3 is produced by stromal cells in the tumor microenvironment and the activating mutations appear to sensitize the tumors to Wnt-Rspo synergy. The combination of RSPO3 inhibition and taxane treatment provides an approach to effectively target oncogenic WNT signaling in a significant number of patients with colorectal and other intestinal cancers.


Subject(s)
Bridged-Ring Compounds/pharmacology , Colorectal Neoplasms , Mutation , Neoplasm Proteins , Paclitaxel/pharmacology , Taxoids/pharmacology , Thrombospondins , Tumor Microenvironment/drug effects , Wnt Signaling Pathway , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Thrombospondins/antagonists & inhibitors , Thrombospondins/genetics , Thrombospondins/metabolism , Tumor Microenvironment/genetics , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , Xenograft Model Antitumor Assays
8.
Sci Adv ; 3(6): e1700090, 2017 06.
Article in English | MEDLINE | ID: mdl-28691093

ABSTRACT

The WNT pathway mediates intercellular signaling that regulates cell fate in both normal development and cancer. It is widely appreciated that the WNT pathway is frequently dysregulated in human cancers through a variety of genetic and epigenetic mechanisms. Targets in the WNT pathway are being extensively pursued for the development of new anticancer therapies, and we have advanced two WNT antagonists for clinical development: vantictumab (anti-FZD) and ipafricept (FZD8-Fc). We examined the antitumor efficacy of these WNT antagonists in combination with various chemotherapies in a large set of patient-derived xenograft models. In responsive models, WNT blockade led to profound synergy with taxanes such as paclitaxel, and the combination activity with taxanes was consistently more effective than with other classes of chemotherapy. Taxane monotherapy increased the frequency of cells with active WNT signaling. This selection of WNT-active chemotherapy-resistant tumorigenic cells was prevented by WNT-antagonizing biologics and required sequential dosing of the WNT antagonist followed by the taxane. The WNT antagonists potentiated paclitaxel-mediated mitotic blockade and promoted widespread mitotic cell death. By blocking WNT/ß-catenin signaling before mitotic blockade by paclitaxel, we found that this treatment effectively sensitizes cancer stem cells to taxanes. This combination strategy and treatment regimen has been incorporated into ongoing clinical testing for vantictumab and ipafricept.


Subject(s)
Antineoplastic Agents/pharmacology , Mitosis/drug effects , Taxoids/pharmacology , Wnt Proteins/antagonists & inhibitors , Cell Death/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Paclitaxel/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors
9.
Nature ; 545(7654): 360-364, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28489825

ABSTRACT

The Notch signalling pathway mediates cell fate decisions and is tumour suppressive or oncogenic depending on the context. During lung development, Notch pathway activation inhibits the differentiation of precursor cells to a neuroendocrine fate. In small-cell lung cancer, an aggressive neuroendocrine lung cancer, loss-of-function mutations in NOTCH genes and the inhibitory effects of ectopic Notch activation indicate that Notch signalling is tumour suppressive. Here we show that Notch signalling can be both tumour suppressive and pro-tumorigenic in small-cell lung cancer. Endogenous activation of the Notch pathway results in a neuroendocrine to non-neuroendocrine fate switch in 10-50% of tumour cells in a mouse model of small-cell lung cancer and in human tumours. This switch is mediated in part by Rest (also known as Nrsf), a transcriptional repressor that inhibits neuroendocrine gene expression. Non-neuroendocrine Notch-active small-cell lung cancer cells are slow growing, consistent with a tumour-suppressive role for Notch, but these cells are also relatively chemoresistant and provide trophic support to neuroendocrine tumour cells, consistent with a pro-tumorigenic role. Importantly, Notch blockade in combination with chemotherapy suppresses tumour growth and delays relapse in pre-clinical models. Thus, small-cell lung cancer tumours generate their own microenvironment via activation of Notch signalling in a subset of tumour cells, and the presence of these cells may serve as a biomarker for the use of Notch pathway inhibitors in combination with chemotherapy in select patients with small-cell lung cancer.


Subject(s)
Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Receptors, Notch/metabolism , Signal Transduction , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Tumor Microenvironment , Animals , Cell Differentiation , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Lung Neoplasms/drug therapy , Male , Mice , Neoplasm Recurrence, Local/prevention & control , Receptors, Notch/agonists , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/deficiency , Repressor Proteins/metabolism , Small Cell Lung Carcinoma/drug therapy
10.
Cancer Res ; 76(3): 713-23, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26719531

ABSTRACT

Deregulation of the ß-catenin signaling has long been associated with cancer. Intracellular components of this pathway, including axin, APC, and ß-catenin, are frequently mutated in a range of human tumors, but the contribution of specific extracellular ligands that promote cancer development through this signaling axis remains unclear. We conducted a reporter-based screen in a panel of human tumors to identify secreted factors that stimulate ß-catenin signaling. Through this screen and further molecular characterization, we found that R-spondin (RSPO) proteins collaborate with Wnt proteins to activate ß-catenin. RSPO family members were expressed in several human tumors representing multiple malignancies, including ovarian, pancreatic, colon, breast, and lung cancer. We generated specific monoclonal antibody antagonists of RSPO family members and found that anti-RSPO treatment markedly inhibited tumor growth in human patient-derived tumor xenograft models, either as single agents or in combination with chemotherapy. Furthermore, blocking RSPO signaling reduced the tumorigenicity of cancer cells based on serial transplantation studies. Moreover, gene-expression analyses revealed that anti-RSPO treatment in responsive tumors strongly inhibited ß-catenin target genes known to be associated with cancer and normal stem cells. Collectively, our results suggest that the RSPO family is an important stimulator of ß-catenin activity in many human tumors and highlight a new effective approach for therapeutically modulating this fundamental signaling axis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Thrombospondins/metabolism , beta Catenin/metabolism , Animals , Carcinogenesis , Cell Line, Tumor , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction , Thrombospondins/biosynthesis , Thrombospondins/genetics , Thrombospondins/immunology , Wnt Proteins/metabolism , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 21(9): 2084-95, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25934888

ABSTRACT

PURPOSE: The Notch pathway plays an important role in both stem cell biology and cancer. Dysregulation of Notch signaling has been reported in several human tumor types. In this report, we describe the development of an antibody, OMP-59R5 (tarextumab), which blocks both Notch2 and Notch3 signaling. EXPERIMENTAL DESIGN: We utilized patient-derived xenograft tumors to evaluate antitumor effect of OMP-59R5. Immunohistochemistry, RNA microarray, real-time PCR, and in vivo serial transplantation assays were employed to investigate the mechanisms of action and pharmacodynamic readouts. RESULTS: We found that anti-Notch2/3, either as a single agent or in combination with chemotherapeutic agents was efficacious in a broad spectrum of epithelial tumors, including breast, lung, ovarian, and pancreatic cancers. Notably, the sensitivity of anti-Notch2/3 in combination with gemcitabine in pancreatic tumors was associated with higher levels of Notch3 gene expression. The antitumor effect of anti-Notch2/3 in combination with gemcitabine plus nab-paclitaxel was greater than the combination effect with gemcitabine alone. OMP-59R5 inhibits both human and mouse Notch2 and Notch3 function and its antitumor activity was characterized by a dual mechanism of action in both tumor and stromal/vascular cells in xenograft experiments. In tumor cells, anti-Notch2/3 inhibited expression of Notch target genes and reduced tumor-initiating cell frequency. In the tumor stroma, OMP-59R5 consistently inhibited the expression of Notch3, HeyL, and Rgs5, characteristic of affecting pericyte function in tumor vasculature. CONCLUSIONS: These findings indicate that blockade of Notch2/3 signaling with this cross-reactive antagonist antibody may be an effective strategy for treatment of a variety of tumor types.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Neoplasms, Experimental/drug therapy , Neoplastic Stem Cells/drug effects , Receptor, Notch2/antagonists & inhibitors , Receptors, Notch/antagonists & inhibitors , Animals , Humans , Immunohistochemistry , Mice , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Receptor, Notch3 , Xenograft Model Antitumor Assays
12.
Carcinogenesis ; 36(1): 115-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25355291

ABSTRACT

Activation of the NOTCH pathway occurs commonly in T acute lymphoblastic leukemia (T-ALL) mainly due to mutations in NOTCH1 or alterations in FBW7 and is involved in the regulation of cell proliferation and survival. Since mutations hit different domains of the receptor, they are predicted to heterogeneously perturb ligand-induced NOTCH1 activity. Moreover, T-ALL cells also co-express NOTCH3 receptors which could be triggered by different ligands. In this study, we aimed to investigate the role of DLL4 in the regulation of NOTCH signaling in T-ALL cells in the context of different types of NOTCH1 mutation or wild-type NOTCH receptor, as well as the effects of DLL4 neutralization on T-ALL engraftment in mice. We found that NOTCH signaling can be stimulated in T-ALL cells in vitro by either human or murine DLL4 with heterogeneous effects, according to NOTCH1/FBW7 mutation status, and that these effects can be blocked by antibodies neutralizing DLL4, NOTCH1 or NOTCH2/3. In vivo, DLL4 is expressed in the spleen and the bone marrow (BM) of NOD/SCID mice bearing T-ALL xenografts as well as the BM of T-ALL patients. Importantly, DLL4 blockade impaired growth of T-ALL cells in NOD/SCID mice and increased leukemia cell apoptosis. These results show that DLL4 is an important component of the tumor microenvironment which contributes to the early steps of T-ALL cell growth.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Notch/metabolism , Adaptor Proteins, Signal Transducing , Animals , Apoptosis , Blotting, Western , Calcium-Binding Proteins , Cell Proliferation , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Cancer Res ; 74(7): 2106-18, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24525742

ABSTRACT

MUSASHI-1 (MSI-1) is a well-established stem cell marker in both normal and malignant colon cells and it acts by positively regulating the NOTCH pathway through inactivation of NUMB, a NOTCH signaling repressor. To date, the mechanisms of regulation of MSI-1 levels remain largely unknown. Here, we investigated the regulation of MSI-1 by NOTCH signaling in colorectal cancer cell lines and in primary cultures of colorectal cancer metastases. Stimulation by the NOTCH ligand DLL4 was associated with an increase of MSI-1 mRNA and protein levels, and this phenomenon was prevented by the addition of an antibody neutralizing NOTCH2/3 but not NOTCH1. Moreover, forced expression of activated NOTCH3 increased MSI-1 levels, whereas silencing of NOTCH3 by short hairpin RNA reduced MSI-1 levels in both colorectal cancer cells and CRC tumor xenografts. Consistent with these findings, enforced NOTCH3 expression or stimulation by DLL4 increased levels of activated NOTCH1 in colorectal cell lines. Finally, treatment of colorectal cancer cells with anti-NOTCH2/3 antibody increased NUMB protein while significantly reducing formation of tumor cell spheroids. This novel feed-forward circuit involving DLL4, NOTCH3, MSI-1, NUMB, and NOTCH1 may be relevant for regulation of NOTCH signaling in physiologic processes as well as in tumor development. With regard to therapeutic implications, NOTCH3-specific drugs could represent a valuable strategy to limit NOTCH signaling in the context of colorectal cancers overexpressing this receptor.


Subject(s)
Colorectal Neoplasms/pathology , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics , Receptors, Notch/physiology , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cell Line, Tumor , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/analysis , Mice , Mice, SCID , Neoplasm Metastasis , Nerve Tissue Proteins/analysis , Receptor, Notch3 , Spheroids, Cellular
14.
Cancer Res ; 73(15): 4909-22, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23761328

ABSTRACT

Wnt ligand expression and activation of the Wnt/ß-catenin pathway have been associated with pancreatic ductal adenocarcinoma, but whether Wnt activity is required for the development of pancreatic cancer has remained unclear. Here, we report the results of three different approaches to inhibit the Wnt/ß-catenin pathway in a established transgenic mouse model of pancreatic cancer. First, we found that ß-catenin null cells were incapable of undergoing acinar to ductal metaplasia, a process associated with development of premalignant pancreatic intraepithelial neoplasia lesions. Second, we addressed the specific role of ligand-mediated Wnt signaling through inducible expression of Dkk1, an endogenous secreted inhibitor of the canonical Wnt pathway. Finally, we targeted the Wnt pathway with OMP-18R5, a therapeutic antibody that interacts with multiple Frizzled receptors. Together, these approaches showed that ligand-mediated activation of the Wnt/ß-catenin pathway is required to initiate pancreatic cancer. Moreover, they establish that Wnt signaling is also critical for progression of pancreatic cancer, a finding with potential therapeutic implications.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/metabolism , Pancreatic Neoplasms/metabolism , Wnt Signaling Pathway/physiology , Animals , Apoptosis/physiology , Blotting, Western , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Pancreatic Neoplasms/pathology , Real-Time Polymerase Chain Reaction , beta Catenin/metabolism
15.
Clin Cancer Res ; 18(19): 5374-86, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22952347

ABSTRACT

PURPOSE: We previously showed that targeting Delta-like ligand 4 (DLL4) in colon and breast tumors inhibited tumor growth and reduced tumor initiating cell frequency. In this report, we have extended these studies to pancreatic cancer and probed the mechanism of action in tumor and stromal cells involved in antitumor efficacy. EXPERIMENTAL DESIGN: Patient-derived pancreatic xenograft tumor models were used to evaluate the antitumor effect of anti-DLL4. To investigate the mechanism of action, we compared the activity of targeting DLL4 in tumor cells with an anti-human DLL4 antibody (anti-hDLL4) and in the host stroma/vasculature with an anti-mouse DLL4 antibody (anti-mDLL4). The effect of these antibodies on cancer stem cell frequency was examined by in vivo limiting dilution assays. RESULTS: The combination of anti-hDLL4 and anti-mDLL4 was efficacious in a broad spectrum of pancreatic tumor xenografts and showed additive antitumor activity together with gemcitabine. Treatment with either anti-hDLL4 or anti-mDLL4 delayed pancreatic tumor recurrence following termination of gemcitabine treatment, and the two together produced an additive effect. Anti-hDLL4 had a pronounced effect in reducing the tumorigenicity of pancreatic cancer cells based on serial transplantation and tumorsphere assays. In contrast, disruption of tumor angiogenesis with anti-mDLL4 alone or with anti-VEGF had minimal effects on tumorigenicity. Gene expression analyses indicated that anti-DLL4 treatment regulated genes that participate in Notch signaling, pancreatic differentiation, and epithelial-to-mesenchymal transition. CONCLUSIONS: Our findings suggest a novel therapeutic approach for pancreatic cancer treatment through antagonism of DLL4/Notch signaling.


Subject(s)
Antibodies, Anti-Idiotypic/administration & dosage , Intercellular Signaling Peptides and Proteins , Neoplastic Stem Cells , Pancreatic Neoplasms , Receptors, Notch/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neovascularization, Pathologic/drug therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Receptors, Notch/immunology , Signal Transduction/drug effects , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism , Gemcitabine
16.
Proc Natl Acad Sci U S A ; 109(29): 11717-22, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22753465

ABSTRACT

The Wnt/ß-catenin pathway, which signals through the Frizzled (Fzd) receptor family and several coreceptors, has long been implicated in cancer. Here we demonstrate a therapeutic approach to targeting the Wnt pathway with a monoclonal antibody, OMP-18R5. This antibody, initially identified by binding to Frizzled 7, interacts with five Fzd receptors through a conserved epitope within the extracellular domain and blocks canonical Wnt signaling induced by multiple Wnt family members. In xenograft studies with minimally passaged human tumors, this antibody inhibits the growth of a range of tumor types, reduces tumor-initiating cell frequency, and exhibits synergistic activity with standard-of-care chemotherapeutic agents.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Frizzled Receptors/metabolism , Neoplasms/drug therapy , Wnt Signaling Pathway/drug effects , Animals , Antibodies, Monoclonal/metabolism , Antineoplastic Agents/metabolism , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , Drug Synergism , Genetic Vectors/genetics , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/genetics , Immunohistochemistry , Lentivirus , Luciferases , Neoplasms/metabolism , Peptide Library , Wnt Signaling Pathway/physiology
17.
Vasc Cell ; 3: 18, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21831306

ABSTRACT

DLL4 is a ligand for the Notch family of receptors. DLL4 has many important functions in normal development and tissue homeostasis, including roles in the immune system, the gastro-intestinal tract, and in vascular development. Because of the importance of Notch signaling in stem cell biology, DLL4 has been investigated for its role in the maintenance and proliferation of cancer stem cells (CSC). In addition, its important role in angiogenesis has been investigated for utility as an anti-angiogenic agent. Preclinical studies have highlighted that both anti-CSC and anti-angiogenic activities contribute to its anti-tumor efficacy, and have supported the clinical development of anti-DLL4 antibody for the treatment of cancer.

18.
Cancer Res ; 71(5): 1520-5, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21193546

ABSTRACT

KRAS mutations are frequent in colorectal cancer (CRC) and are associated with clinical resistance to treatment with the epidermal growth factor receptor (EGFR)-targeted monoclonal antibodies. Delta-like 4 ligand (DLL4) is an important component of the Notch signaling pathway and mediates stem cell self-renewal and vascular development. DLL4 inhibition in colon tumor cells reduces tumor growth and stem cell frequency. Considering the need for new drugs to treat colon cancers with oncogenic KRAS mutations, we examined in this study the efficacy of anti-DLL4 antibodies in KRAS mutant tumors in a panel of early passage colon tumor xenograft models derived from patients. Consistent with clinical findings, mutant KRAS colorectal xenograft tumors were insensitive to the EGFR therapeutic antibody cetuximab, whereas KRAS wild-type tumors responded to cetuximab. In contrast, anti-DLL4 was efficacious against both wild-type and mutant KRAS colon tumors as a single agent and in combination with irinotecan. Further analysis of mutant KRAS tumors indicated that the anti-DLL4/irinotecan combination produced a significant decrease in colon cancer stem cell frequency while promoting apoptosis in tumor cells. Our findings provide a rationale for targeting DLL4-Notch signaling for improved treatment of CRC patients with activating KRAS mutations.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Mutation , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , Calcium-Binding Proteins , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Irinotecan , Mice , Mice, SCID , Neoplastic Stem Cells/drug effects , Proto-Oncogene Proteins p21(ras) , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
19.
Sci Transl Med ; 2(28): 28ps19, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20410528

ABSTRACT

The resistance of cancer cells to various therapeutic agents and modalities is a crucial issue in clinical oncology. A recent Cell paper highlights how changes at the chromatin level control drug sensitivity and provides insight into more effective strategies for treating cancer with combination therapies.


Subject(s)
Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Genetic Heterogeneity , Neoplasms/drug therapy , Neoplasms/genetics , Drug Resistance, Neoplasm/drug effects , Epigenesis, Genetic/drug effects , ErbB Receptors/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Genetic Heterogeneity/drug effects , Humans , Mutation/genetics , Neoplasms/enzymology , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology
20.
Cell Stem Cell ; 5(2): 168-77, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19664991

ABSTRACT

Previous studies have shown that blocking DLL4 signaling reduced tumor growth by disrupting productive angiogenesis. We developed selective anti-human and anti-mouse DLL4 antibodies to dissect the mechanisms involved by analyzing the contributions of selectively targeting DLL4 in the tumor or in the host vasculature and stroma in xenograft models derived from primary human tumors. We found that each antibody inhibited tumor growth and that the combination of the two antibodies was more effective than either alone. Treatment with anti-human DLL4 inhibited the expression of Notch target genes and reduced proliferation of tumor cells. Furthermore, we found that specifically inhibiting human DLL4 in the tumor, either alone or in combination with the chemotherapeutic agent irinotecan, reduced cancer stem cell frequency, as shown by flow cytometric and in vivo tumorigenicity studies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Intercellular Signaling Peptides and Proteins/immunology , Neoplasms/therapy , Neoplastic Stem Cells/immunology , Receptors, Notch/metabolism , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Chaperonin 60/agonists , Chaperonin 60/metabolism , Drug Synergism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Irinotecan , Membrane Proteins/antagonists & inhibitors , Mice , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neovascularization, Pathologic/metabolism , Secondary Prevention , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...