Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Cell Rep ; 38(10): 110490, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263600

ABSTRACT

How metastatic cells arise is unclear. Here, we search for the induction of recently characterized pro-metastatic states as a surrogate for the origin of metastasis. Since cell-death-inducing therapies can paradoxically promote metastasis, we ask if such treatments induce pro-metastatic states in human colon cancer cells. We find that post-near-death cells acquire pro-metastatic states (PAMEs) and form distant metastases in vivo. These PAME ("let's go" in Greek) cells exhibit a multifactorial cytokine storm as well as signs of enhanced endoplasmic reticulum (ER) stress and nuclear reprogramming, requiring CXCL8, INSL4, IL32, PERK-CHOP, and NANOG. PAMEs induce neighboring tumor cells to become PAME-induced migratory cells (PIMs): highly migratory cells that re-enact the storm and enhance PAME migration. Metastases are thus proposed to originate from the induction of pro-metastatic states through intrinsic and extrinsic cues in a pro-metastatic tumoral ecosystem, driven by an impending cell-death experience involving ER stress modulation, metastatic reprogramming, and paracrine recruitment via a cytokine storm.


Subject(s)
Colonic Neoplasms , Cytokine Release Syndrome , Cell Death , Colonic Neoplasms/pathology , Ecosystem , Humans , Signal Transduction
2.
Cell Rep ; 33(6): 108372, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33176137

ABSTRACT

How cells with metastatic potential, or pro-metastatic states, arise within heterogeneous primary tumors remains unclear. Here, we have used one index primary colon cancer to develop spiked-scRNAseq to link omics-defined single-cell clusters with cell behavior. Using spiked-scRNAseq we uncover cell populations with differential metastatic potential in which pro-metastatic states are correlated with the expression of signaling and vesicle-trafficking genes. Analyzing such heterogeneity, we define an anti-metastatic, non-cell-autonomous interaction originating from non-/low-metastatic cells, and identify membrane VSIG1 as a critical mediator of this interaction. VSIG1 acts to restrict the development of pro-metastatic states autonomously and non-cell autonomously, in part by inhibiting YAP/TAZ-TEAD signaling. As VSIG1 re-expression is able to reduce metastatic behavior from multiple colon cancer cell types, the regulation of VSIG1 or its effectors opens new interventional opportunities. In general, we propose that crosstalk between cancer cells, including the action of VSIG1, dynamically defines the degree of pro-metastatic intra-tumoral heterogeneity.


Subject(s)
Cell Communication/physiology , Membrane Glycoproteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , RNA, Small Cytoplasmic/metabolism , Animals , Genetic Heterogeneity , Humans , Mice , Neoplasms/genetics
3.
Oncogene ; 38(29): 5817-5837, 2019 07.
Article in English | MEDLINE | ID: mdl-31253868

ABSTRACT

How cells in primary tumors initially become pro-metastatic is not understood. A previous genome-wide RNAi screen uncovered colon cancer metastatic suppressor and WNT promoting functions of TMED3, a member of the p24 ER-to-Golgi protein secretion family. Repression of canonical WNT signaling upon knockdown (kd) of TMED3 might thus be sufficient to drive metastases. However, searching for transcriptional influences on other family members here we find that TMED3 kd leads to enhanced TMED9, that TMED9 acts downstream of TMED3 and that TMED9 kd compromises metastasis. Importantly, TMED9 pro-metastatic function is linked to but distinct from the repression of TMED3-WNT-TCF signaling. Functional rescue of the migratory deficiency of TMED9 kd cells identifies TGFα as a mediator of TMED9 pro-metastatic activity. Moreover, TMED9 kd compromises the biogenesis, and thus function, of TGFα. Analyses in three colon cancer cell types highlight a TMED9-dependent gene set that includes CNIH4, a member of the CORNICHON family of TGFα exporters. Our data indicate that TGFA and CNIH4, which display predictive value for disease-free survival, promote colon cancer cell metastatic behavior, and suggest that TMED9 pro-metastatic function involves the modulation of the secretion of TGFα ligand. Finally, TMED9/TMED3 antagonism impacts WNT-TCF and GLI signaling, where TMED9 primacy over TMED3 leads to the establishment of a positive feedback loop together with CNIH4, TGFα, and GLI1 that enhances metastases. We propose that primary colon cancer cells can transition between two states characterized by secretion-transcription regulatory loops gated by TMED3 and TMED9 that modulate their metastatic proclivities.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Neoplasm Metastasis , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Transforming Growth Factor alpha/metabolism , Vesicular Transport Proteins/metabolism , Wnt Signaling Pathway , Zinc Finger Protein GLI1/metabolism , Epistasis, Genetic , Gene Expression Regulation , Humans , RNA, Messenger/metabolism
4.
Biochim Biophys Acta Rev Cancer ; 1871(2): 434-454, 2019 04.
Article in English | MEDLINE | ID: mdl-31034926

ABSTRACT

The strategy of using existing drugs originally developed for one disease to treat other indications has found success across medical fields. Such drug repurposing promises faster access of drugs to patients while reducing costs in the long and difficult process of drug development. However, the number of existing drugs and diseases, together with the heterogeneity of patients and diseases, notably including cancers, can make repurposing time consuming and inefficient. The key question we address is how to efficiently repurpose an existing drug to treat a given indication. As drug efficacy remains the main bottleneck for overall success, we discuss the need for machine-learning computational methods in combination with specific phenotypic studies along with mechanistic studies, chemical genetics and omics assays to successfully predict disease-drug pairs. Such a pipeline could be particularly important to cancer patients who face heterogeneous, recurrent and metastatic disease and need fast and personalized treatments. Here we focus on drug repurposing for colorectal cancer and describe selected therapeutics already repositioned for its prevention and/or treatment as well as potential candidates. We consider this review as a selective compilation of approaches and methodologies, and argue how, taken together, they could bring drug repurposing to the next level.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Drug Repositioning/methods , Machine Learning , Animals , Humans , Medical Oncology/methods , Phenotype
5.
Sci Rep ; 9(1): 3891, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30846719

ABSTRACT

Targeting stemness promises new therapeutic strategies against highly invasive tumors. While a number of approaches are being tested, inhibiting the core transcription regulatory network of cancer stem cells is an attractive yet challenging possibility. Here we have aimed to provide the proof of principle for a strategy, previously used in developmental studies, to directly repress the targets of a salient stemness and pluripotency factor: NANOG. In doing so we expected to inhibit the expression of so far unknown mediators of pro-tumorigenic NANOG function. We chose NANOG since previous work showed the essential requirement for NANOG activity for human glioblastoma (GBM) growth in orthotopic xenografts, and it is apparently absent from many adult human tissues thus likely minimizing unwanted effects on normal cells. NANOG repressor chimeras, which we name NANEPs, bear the DNA-binding specificity of NANOG through its homeodomain (HD), and this is linked to transposable human repressor domains. We show that in vitro and in vivo, NANEP5, our most active NANEP with a HES1 repressor domain, mimics knock-down (kd) of NANOG function in GBM cells. Competition orthotopic xenografts also reveal the effectiveness of NANEP5 in a brain tumor context, as well as the specificity of NANEP activity through the abrogation of its function via the introduction of specific mutations in the HD. The transcriptomes of cells expressing NANEP5 reveal multiple potential mediators of pro-tumorigenic NANEP/NANOG action including intercellular signaling components. The present results encourage further studies on the regulation of context-dependent NANEP abundance and function, and the development of NANEP-based anti-cancer therapies.


Subject(s)
Brain Neoplasms/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Nanog Homeobox Protein/genetics , Neoplastic Stem Cells/pathology , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Gene Knockdown Techniques , Glioblastoma/pathology , Humans , Mice , Mice, Nude
6.
PLoS One ; 11(12): e0168170, 2016.
Article in English | MEDLINE | ID: mdl-27973612

ABSTRACT

The WNT-TCF signaling pathway participates in adult tissue homeostasis and repair, and is hyperactive in a number of human diseases including cancers of the colon. Whereas to date there are no antagonists approved for patient use, a potential problem for their sustained use is the blockade of WNT signaling in healthy tissues, thus provoking potentially serious co-lateral damage. Here we have screened a library of plant and microorganism small molecules for novel WNT signaling antagonists and describe withanolide F as a potent WNT-TCF response blocker. This steroidal lactone inhibits TCF-dependent colon cancer xenograft growth and mimics the effects of genetic blockade of TCF and of ivermectin, a previously reported WNT-TCF blocker. However, withanolide F is unique in that it imposes a long-lasting repression of tumor growth, WNT-TCF targets and cancer stem cell clonogenicity after drug treatment. These findings are paralleled by its modulation of chromatin regulators and its alteration of overall H3K4me1 levels. Our results open up the possibility to permanently repress essential signaling responses in cancer cells through limited treatments with small molecules.


Subject(s)
Epigenesis, Genetic , TCF Transcription Factors/metabolism , Withanolides/chemistry , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , Animals , Cell Line, Tumor , Chromatin/chemistry , Colonic Neoplasms/metabolism , Epistasis, Genetic , Female , HEK293 Cells , Histones/chemistry , Homeostasis , Humans , Ivermectin/chemistry , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/cytology , Signal Transduction
7.
PLoS One ; 11(8): e0160904, 2016.
Article in English | MEDLINE | ID: mdl-27489960

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0150697.].

8.
PLoS One ; 11(3): e0150697, 2016.
Article in English | MEDLINE | ID: mdl-26939070

ABSTRACT

Human colon cancers commonly harbor loss of function mutations in APC, a repressor of the canonical WNT pathway, thus leading to hyperactive WNT-TCF signaling. Re-establishment of Apc function in mice, engineered to conditionally repress Apc through RNAi, resolve the intestinal tumors formed due to hyperactivated Wnt-Tcf signaling. These and other results have prompted the search for specific WNT pathway antagonists as therapeutics for clinically problematic human colon cancers and associated metastases, which remain largely incurable. This widely accepted view seems at odds with a number of findings using patient-derived material: Canonical TCF targets are repressed, instead of being hyperactivated, in advanced colon cancers, and repression of TCF function does not generally result in tumor regression in xenografts. The results of a number of genetic mouse studies have also suggested that canonical WNT-TCF signaling drives metastases, but direct in vivo tests are lacking, and, surprisingly, TCF repression can enhance directly seeded metastatic growth. Here we have addressed the abilities of enhanced and blocked WNT-TCF signaling to alter tumor growth and distant metastases using xenografts of advanced human colon cancers in mice. We find that endogenous WNT-TCF signaling is mostly anti-metastatic since downregulation of TCF function with dnTCF generally enhances metastatic spread. Consistently, elevating the level of WNT signaling, by increasing the levels of WNT ligands, is not generally pro-metastatic. Our present and previous data reveal a heterogeneous response to modulating WNT-TCF signaling in human cancer cells. Nevertheless, the findings that a fraction of colon cancers tested require WNT-TCF signaling for tumor growth but all respond to repressed signaling by increasing metastases beg for a reevaluation of the goal of blocking WNT-TCF signaling to universally treat colon cancers. Our data suggest that WNT-TCF blockade may be effective in inhibiting tumor growth in only a subset of cases but will generally boost metastases.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Neoplasm Metastasis , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Cell Line, Tumor , Cell Proliferation , Female , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Lac Operon , Lentivirus/metabolism , Ligands , Mice , Mice, Nude , Neoplasm Transplantation , Phenotype , Polymerase Chain Reaction , Species Specificity
9.
J Mol Cell Biol ; 8(2): 157-73, 2016 04.
Article in English | MEDLINE | ID: mdl-26031752

ABSTRACT

How metastases develop is not well understood and no genetic mutations have been reported as specific metastatic drivers. Here we have addressed the idea that epigenetic reprogramming by GLI-regulated pluripotent stemness factors promotes metastases. Using primary human colon cancer cells engrafted in mice, we find that transient expression of OCT4, SOX2, KLF4 +/- cMYC establishes an enhanced pro-metastatic state in the primary tumor that is stable through sequential engraftments and is transmitted through clonogenic cancer stem cells. Metastatic reprogramming alters NANOG methylation and stably boosts NANOG and NANOGP8 expression. Metastases and reprogrammed EMT-like phenotypes require endogenous NANOG, but enhanced NANOG is not sufficient to induce these phenotypes. Finally, reprogrammed tumors enhance GLI2, and we show that GLI2(high) and AXIN2(low), which are markers of the metastatic transition of colon cancers, are prognostic of poor disease outcome in patients. We propose that metastases arise through epigenetic reprogramming of cancer stem cells within primary tumors.


Subject(s)
Cellular Reprogramming/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Epigenesis, Genetic , Animals , Cell Line, Tumor , Clone Cells , DNA Methylation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Factor 4 , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Signal Transduction/genetics , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
10.
EMBO Mol Med ; 6(10): 1263-78, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25143352

ABSTRACT

Constitutive activation of canonical WNT-TCF signaling is implicated in multiple diseases, including intestine and lung cancers, but there are no WNT-TCF antagonists in clinical use. We have performed a repositioning screen for WNT-TCF response blockers aiming to recapitulate the genetic blockade afforded by dominant-negative TCF. We report that Ivermectin inhibits the expression of WNT-TCF targets, mimicking dnTCF, and that its low concentration effects are rescued by direct activation by TCF(VP16). Ivermectin inhibits the proliferation and increases apoptosis of various human cancer types. It represses the levels of C-terminal ß-CATENIN phosphoforms and of CYCLIN D1 in an okadaic acid-sensitive manner, indicating its action involves protein phosphatases. In vivo, Ivermectin selectively inhibits TCF-dependent, but not TCF-independent, xenograft growth without obvious side effects. Analysis of single semi-synthetic derivatives highlights Selamectin, urging its clinical testing and the exploration of the macrocyclic lactone chemical space. Given that Ivermectin is a safe anti-parasitic agent used by > 200 million people against river blindness, our results suggest its additional use as a therapeutic WNT-TCF pathway response blocker to treat WNT-TCF-dependent diseases including multiple cancers.


Subject(s)
Ivermectin/pharmacology , Lactones/pharmacology , Neoplasms/drug therapy , Signal Transduction/drug effects , TCF Transcription Factors/metabolism , Wnt Proteins/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Female , HEK293 Cells , HT29 Cells , Humans , Immunohistochemistry , Insecticides/pharmacology , Macrocyclic Compounds/pharmacology , Mice, Inbred Strains , Mice, Nude , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Onchocerciasis, Ocular/drug therapy , Reverse Transcriptase Polymerase Chain Reaction , TCF Transcription Factors/genetics , Wnt Proteins/genetics , Xenograft Model Antitumor Assays , beta Catenin/genetics , beta Catenin/metabolism
11.
EMBO Mol Med ; 6(7): 882-901, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24920608

ABSTRACT

The progression of tumors to the metastatic state involves the loss of metastatic suppressor functions. Finding these, however, is difficult as in vitro assays do not fully predict metastatic behavior, and the majority of studies have used cloned cell lines, which do not reflect primary tumor heterogeneity. Here, we have designed a novel genome-wide screen to identify metastatic suppressors using primary human tumor cells in mice, which allows saturation screens. Using this unbiased approach, we have tested the hypothesis that endogenous colon cancer metastatic suppressors affect WNT-TCF signaling. Our screen has identified two novel metastatic suppressors: TMED3 and SOX12, the knockdown of which increases metastatic growth after direct seeding. Moreover, both modify the type of self-renewing spheroids, but only knockdown of TMED3 also induces spheroid cell spreading and lung metastases from a subcutaneous xenograft. Importantly, whereas TMED3 and SOX12 belong to different families involved in protein secretion and transcriptional regulation, both promote endogenous WNT-TCF activity. Treatments for advanced or metastatic colon cancer may thus not benefit from WNT blockers, and these may promote a worse outcome.


Subject(s)
Colonic Neoplasms/pathology , Neoplasm Metastasis/genetics , SOXC Transcription Factors/genetics , Signal Transduction , TCF Transcription Factors/metabolism , Vesicular Transport Proteins/genetics , Wnt Proteins/metabolism , Animals , Cell Line, Tumor , Cell Movement , Colon/metabolism , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genome , Humans , Mice , Neoplasm Metastasis/pathology , SOXC Transcription Factors/metabolism , Vesicular Transport Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
12.
Semin Cell Dev Biol ; 33: 93-104, 2014 09.
Article in English | MEDLINE | ID: mdl-24852887

ABSTRACT

Canonical Hedgehog (HH) signaling leads to the regulation of the GLI code: the sum of all positive and negative functions of all GLI proteins. In humans, the three GLI factors encode context-dependent activities with GLI1 being mostly an activator and GLI3 often a repressor. Modulation of GLI activity occurs at multiple levels, including by co-factors and by direct modification of GLI structure. Surprisingly, the GLI proteins, and thus the GLI code, is also regulated by multiple inputs beyond HH signaling. In normal development and homeostasis these include a multitude of signaling pathways that regulate proto-oncogenes, which boost positive GLI function, as well as tumor suppressors, which restrict positive GLI activity. In cancer, the acquisition of oncogenic mutations and the loss of tumor suppressors - the oncogenic load - regulates the GLI code toward progressively more activating states. The fine and reversible balance of GLI activating GLI(A) and GLI repressing GLI(R) states is lost in cancer. Here, the acquisition of GLI(A) levels above a given threshold is predicted to lead to advanced malignant stages. In this review we highlight the concepts of the GLI code, the oncogenic load, the context-dependency of GLI action, and different modes of signaling integration such as that of HH and EGF. Targeting the GLI code directly or indirectly promises therapeutic benefits beyond the direct blockade of individual pathways.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Basal Cell/metabolism , Colonic Neoplasms/metabolism , Signal Transduction , Transcription Factors/physiology , Animals , Carcinogenesis/genetics , Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/physiology , Humans , Molecular Targeted Therapy , Zinc Finger Protein GLI1
13.
Sci Signal ; 4(200): pt9, 2011 Nov 22.
Article in English | MEDLINE | ID: mdl-22114144

ABSTRACT

The Hedgehog (Hh)-Gli signaling pathway is an essential pathway involved in development and cancer. It controls the Gli code-the sum of all activator and repressor functions of the Gli transcription factors. Through the Gli code, and Gli1 in particular, it modulates the fate and behavior of stem and cancer stem cells, as well as tumor growth and survival in many human cancer types. It also affects recurrence and metastasis and is enhanced in advanced tumors, where it promotes an embryonic stem (ES) cell-like gene expression signature. A central component of this signature, Nanog, is critical for glioblastoma and cancer stem cell survival and expansion. Gli1 activity is also enhanced by several oncogenic proteins, including Ras, Myc, and Akt, and by loss of tumor suppressors, such as p53 and PTEN. The oncogenic load boosts Gli1 levels, which supports tumor progression, and promotes a critical threshold of Gli1 activity that allows cells to enter the metastatic transition. In colon cancers, this transition is defined by enhanced Hh-Gli and, surprisingly, by repressed Wnt-Tcf signaling. Together our data support a model in which the Gli code, and Gli1 in particular, acts as a key sensor that responds to both Hh signals and the oncogenic load. We hypothesize that, in turn, the Gli-regulated ES-like factors induce a reprogramming event in cancer stem cells that promotes high invasion, growth and/or metastasis. Targeting the Gli code, the autoregulatory Gli1-Nanog module and interacting partners and pathways thus offers new therapeutic possibilities.


Subject(s)
Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Neoplastic Stem Cells/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Hedgehog Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Nanog Homeobox Protein , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zinc Finger Protein GLI1
14.
J Mol Cell Biol ; 2(6): 318-32, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21098050

ABSTRACT

Human colon carcinomas (CCs) represent a growing worldwide problem. One of the pathways that has been negatively implicated in the genesis of CCs is triggered by bone morphogenetic protein (BMP) ligands, which activate BMP receptors leading to the function of SMAD proteins in the nucleus. BMP signaling is altered in familial human polyposis, and mice with compromised BMP signaling in the intestine develop tumors. Here, we have re-evaluated the presence and roles of BMP signaling in advanced sporadic human CCs, using both primary tumors and established cell lines, and directly modulating BMP pathway activity in a cell-autonomous manner using constitutively active and dominant-negative BMP receptor Ib forms. We find evidence for active endogenous BMP signaling in all primary CC samples and for its role in promoting primary CC tumor growth and CC cell survival and proliferation in vivo in xenografts. In vitro, we also document autonomous and non-autonomous effects of enhanced BMP receptor activity on gap closure in culture, suggesting possible roles in invasion. Caution should thus be exerted in trying to augment or restore its activity for therapeutic purposes. In contrast, we raise the possibility that blockade of BMP signaling might have beneficial effects against at least a subset of advanced colon cancers.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Carcinoma/metabolism , Colonic Neoplasms/metabolism , Signal Transduction , Animals , Apoptosis , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Proliferation , Cell Survival , Cells, Cultured , Humans , Mice , Mice, Nude , Oncogene Proteins/physiology , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Trans-Activators/physiology , Transplantation, Heterologous , Zinc Finger Protein GLI1
15.
EMBO Mol Med ; 2(11): 440-57, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20941789

ABSTRACT

Previous studies demonstrate the initiation of colon cancers through deregulation of WNT-TCF signalling. An accepted but untested extension of this finding is that incurable metastatic colon carcinomas (CCs) universally remain WNT-TCF-dependent, prompting the search for WNT-TCF inhibitors. CCs and their stem cells also require Hedgehog (HH)-GLI1 activity, but how these pathways interact is unclear. Here we define coincident high-to-low WNT-TCF and low-to-high HH-GLI transitions in patient CCs, most strikingly in their CD133(+) stem cells, that mark the development of metastases. We find that enhanced HH-GLI mimics this transition, driving also an embryonic stem (ES)-like stemness signature and that GLI1 can be regulated by multiple CC oncogenes. The data support a model in which the metastatic transition involves the acquisition or enhancement of a more primitive ES-like phenotype, and the downregulation of the early WNT-TCF programme, driven by oncogene-regulated high GLI1 activity. Consistently, TCF blockade does not generally inhibit tumour growth; instead, it, like enhanced HH-GLI, promotes metastatic growth in vivo. Treatments for metastatic disease should therefore block HH-GLI1 but not WNT-TCF activities.


Subject(s)
Carcinoma/physiopathology , Colonic Neoplasms/physiopathology , Hedgehog Proteins/metabolism , Neoplasm Metastasis/physiopathology , TCF Transcription Factors/metabolism , Transcription Factors/metabolism , Wnt Proteins/metabolism , AC133 Antigen , Antigens, CD/analysis , Carcinoma/pathology , Colonic Neoplasms/pathology , Gene Expression Profiling , Glycoproteins/analysis , Humans , Neoplasm Metastasis/pathology , Peptides/analysis , Stem Cells , Zinc Finger Protein GLI1
16.
EMBO J ; 29(15): 2659-74, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20581802

ABSTRACT

A cohort of genes associated with embryonic stem (ES) cell behaviour, including NANOG, are expressed in a number of human cancers. They form an ES-like signature we first described in glioblastoma multiforme (GBM), a highly invasive and incurable brain tumour. We have also shown that HEDGEHOG-GLI (HH-GLI) signalling is required for GBM growth, stem cell expansion and the expression of this (ES)-like stemness signature. Here, we address the function of NANOG in human GBMs and its relationship with HH-GLI activity. We find that NANOG modulates gliomasphere clonogenicity, CD133(+) stem cell cell behavior and proliferation, and is regulated by HH-GLI signalling. However, GLI1 also requires NANOG activity forming a positive loop, which is negatively controlled by p53 and vice versa. NANOG is essential for GBM tumourigenicity in orthotopic xenografts and it is epistatic to HH-GLI activity. Our data establish NANOG as a novel HH-GLI mediator essential for GBMs. We propose that this function is conserved and that tumour growth and stem cell behaviour rely on the status of a functional GLI1-NANOG-p53 network.


Subject(s)
Glioma/metabolism , Homeodomain Proteins/metabolism , Neoplastic Stem Cells/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Aged , Aged, 80 and over , Animals , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Glioma/pathology , Homeodomain Proteins/genetics , Humans , Male , Middle Aged , Nanog Homeobox Protein , Neoplastic Stem Cells/cytology , Signal Transduction , Tumor Cells, Cultured , Zinc Finger Protein GLI1
17.
Biochem Pharmacol ; 80(5): 712-23, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20412786

ABSTRACT

Many human sporadic cancers have been recently shown to require the activity of the Hedgehog-GLI pathway for sustained growth. The survival and expansion of cancer stem cells is also HH-GLI dependent. Here we review the advances on the modulation of HH-GLI signaling by small molecules. We focus on both natural compounds and synthetic molecules that target upstream pathway components, mostly SMOOTHENED, and those that target the last steps of the pathway, the GLI transcription factors. In this review we have sought to provide some bases for useful comparisons, listing original assays used and sources to facilitate comparisons of IC50 values. This area is a rapidly expanding field where biology, medicine and chemistry intersect, both in academia and industry. We also highlight current clinical trials, with positive results in early stages. While we have tried to be exhaustive regarding the molecules, not all data is in the public domain yet. Indeed, we have opted to avoid listing chemical structures but these can be easily found in the references given. Finally, we are hopeful that the best molecules will soon reach the patients but caution about the lack of investment on compounds that lack tight IP positions. While the market in developed nations is expected to compensate the investment and risk of making HH-GLI modulators, other sources or plans must be available for developing nations and poor patient populations. The promise of curing cancer recalls the once revered dream of El Dorado, which taught us that not everything that GLI-tters is gold.


Subject(s)
Hedgehog Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Hedgehog Proteins/agonists , Hedgehog Proteins/antagonists & inhibitors , Humans , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors , Zinc Finger Protein GLI1
18.
J Mol Cell Biol ; 2(2): 84-95, 2010 04.
Article in English | MEDLINE | ID: mdl-20083481

ABSTRACT

A surprisingly large and unrelated number of human tumors depend on sustained HEDGEHOG-GLI (HH-GLI) signaling for growth. This includes cancers of the skin, brain, colon, lungs, prostate, blood and pancreas among others. The basis of such commonality is not obvious. HH-GLI signaling has also been shown to be active in and required for cancer stem cell survival and expansion in different cancer types, and its activity is essential not only for tumor growth but also for recurrence and metastatic growth, two key medical problems. Here we review recent data on the role of HH-GLI signaling in cancer focusing on the role of the GLI code, the regulated combinatorial and cooperative function of repressive and activating forms of all Gli transcription factors, as a signaling nexus that integrates not only HH signals but also those of multiple tumor suppressors and oncogenes. Recent data support the view that the context-dependent regulation of the GLI code by oncogenes and tumor suppressors constitutes a basis for the widespread involvement of GLI1 in human cancers, representing a perversion of its normal role in the control of stem cell lineages during normal development and homeostasis.


Subject(s)
Hedgehog Proteins/metabolism , Neoplasms/metabolism , Signal Transduction , Transcription Factors/metabolism , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
19.
Mech Dev ; 127(1-2): 73-81, 2010.
Article in English | MEDLINE | ID: mdl-19861162

ABSTRACT

Several lines of evidence point to the central role of WNT signaling in the initiation of intestinal tumorigenesis, most often due to loss of APC, a negative regulator of the WNT-betaCATENIN/TCF pathway. Modeling human colon cancers in mice through loss of Apc has shown that inappropriate activation of Wnt signaling is sufficient to induce adenoma formation. More recent analyses have also demonstrated a key role for HEDGEHOG-GLI (HH-GLI) signaling in human colon cancers. However, how the WNT and HH pathways interact during intestinal development, homeostasis and cancer is not clear. Marker analyses suggest predominant paracrine signaling from rare Shh producing cells in the crypt's bottom to adjacent Gli1(+) mesenchymal cells in normal adult mice. Using conditional KO models, we show that inhibition of the function of the critical Hh mediator Smoothened (Smo) rescues the lethality and intestinal phenotypes of loss of Apc. The results uncover an essential role of the Hh pathway in tumors induced by hyperactive Wnt signaling, suggest the action of the Hh pathway in parallel or downstream of Wnt signaling, and validate this model for its use in preclinical work testing Hh pathway antagonists.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Gene Expression Regulation, Developmental , Intestines/embryology , Wnt Proteins/metabolism , Animals , Epithelium/embryology , Homeostasis , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Transgenic , Phenotype , Stem Cells/metabolism , Tamoxifen/pharmacology , Time Factors , Veratrum Alkaloids/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...