Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(5): 104611, 2023 05.
Article in English | MEDLINE | ID: mdl-36931394

ABSTRACT

Adipose tissue plays a crucial role in maintaining metabolic homeostasis by storing lipids and glucose from circulation as intracellular fat. As peripheral tissues like adipose tissue become insulin resistant, decompensation of blood glucose levels occurs causing type 2 diabetes (T2D). Currently, modulating the glycocalyx, a layer of cell-surface glycans, is an underexplored pharmacological treatment strategy to improve glucose homeostasis in T2D patients. Here, we show a novel role for cell-surface heparan sulfate (HS) in establishing glucose uptake capacity and metabolic utilization in differentiated adipocytes. Using a combination of chemical and genetic interventions, we identified that HS modulates this metabolic phenotype by attenuating levels of Wnt signaling during adipogenesis. By engineering, the glycocalyx of pre-adipocytes with exogenous synthetic HS mimetics, we were able to enhance glucose clearance capacity after differentiation through modulation of Wnt ligand availability. These findings establish the cellular glycocalyx as a possible new target for therapeutic intervention in T2D patients by enhancing glucose clearance capacity independent of insulin secretion.


Subject(s)
Adipogenesis , Diabetes Mellitus, Type 2 , Humans , Adipogenesis/genetics , Glycocalyx/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Heparitin Sulfate , Glucose/metabolism
2.
Curr Top Dev Biol ; 153: 95-119, 2023.
Article in English | MEDLINE | ID: mdl-36967203

ABSTRACT

The role of Wnt signaling in stem cells has been mired in seemingly contradictory findings. On one hand, Wnt has been heralded as a self-renewal factor. On the other hand, Wnt's association with differentiation and lineage commitment is indisputable. This apparent contradiction is particularly evident in pluripotent stem cells, where Wnt promotes self-renewal as well as differentiation. To resolve this discrepancy one must delve into fundamental principles of pluripotency and gain an appreciation for the concept of pluripotency states, which exist in a continuum with intermediate metastable states, some of which have been stabilized in vitro. Wnt signaling is a critical regulator of transitions between pluripotent states. Here, we will discuss Wnt's roles in maintaining pluripotency, promoting differentiation, as well as stimulating reprogramming of somatic cells to an induced pluripotent state.


Subject(s)
Embryonic Stem Cells , Pluripotent Stem Cells , Wnt Signaling Pathway , Cell Differentiation/physiology
3.
Curr Top Dev Biol ; 153: xv-xvi, 2023.
Article in English | MEDLINE | ID: mdl-36967204
4.
Methods Mol Biol ; 2438: 123-131, 2022.
Article in English | MEDLINE | ID: mdl-35147939

ABSTRACT

Wnt proteins are secreted, lipid-modified growth factors with a wide range of activities across all metazoan species. Their production, secretion, and signaling range are under tight cellular control such that detection of Wnt proteins in biological samples is often extremely difficult. In this chapter, we describe a protocol to detect secreted Wnt proteins in the culture medium of cell lines that ectopically or endogenously express Wnt genes. This protocol uses an affinity resin, called Blue Sepharose, that binds and thereby enriches Wnt proteins, followed by immunoblotting for the Wnt protein of interest. This method for detecting Wnt proteins will aid in the isolation of biologically active Wnt proteins, provide an assay to study the molecular basis of Wnt secretion, and potentially offer a means to detect trace amounts of Wnt proteins associated with pathological states.


Subject(s)
Signal Transduction , Wnt Proteins , Animals , Cell Culture Techniques , Cell Line
5.
Mol Cancer Ther ; 21(1): 113-124, 2022 01.
Article in English | MEDLINE | ID: mdl-34667113

ABSTRACT

Although WNT signaling is frequently dysregulated in solid tumors, drugging this pathway has been challenging due to off-tumor effects. Current clinical pan-WNT inhibitors are nonspecific and lead to adverse effects, highlighting the urgent need for more specific WNT pathway-targeting strategies. We identified elevated expression of the WNT receptor Frizzled class receptor 7 (FZD7) in multiple solid cancers in The Cancer Genome Atlas, particularly in the mesenchymal and proliferative subtypes of ovarian serous cystadenocarcinoma, which correlate with poorer median patient survival. Moreover, we observed increased FZD7 protein expression in ovarian tumors compared with normal ovarian tissue, indicating that FZD7 may be a tumor-specific antigen. We therefore developed a novel antibody-drug conjugate, septuximab vedotin (F7-ADC), which is composed of a chimeric human-mouse antibody to human FZD7 conjugated to the microtubule-inhibiting drug monomethyl auristatin E (MMAE). F7-ADC selectively binds human FZD7, potently kills ovarian cancer cells in vitro, and induces regression of ovarian tumor xenografts in murine models. To evaluate F7-ADC toxicity in vivo, we generated mice harboring a modified Fzd7 gene where the resulting Fzd7 protein is reactive with the human-targeting F7-ADC. F7-ADC treatment of these mice did not induce acute toxicities, indicating a potentially favorable safety profile in patients. Overall, our data suggest that the antibody-drug conjugate approach may be a powerful strategy to combat FZD7-expressing ovarian cancers in the clinic.


Subject(s)
Frizzled Receptors/genetics , Immunoconjugates/metabolism , Ovarian Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , Ovarian Neoplasms/pathology
6.
N Engl J Med ; 385(14): 1292-1301, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34587386

ABSTRACT

BACKGROUND: Structural birth defects occur in approximately 3% of live births; most such defects lack defined genetic or environmental causes. Despite advances in surgical approaches, pharmacologic prevention remains largely out of reach. METHODS: We queried worldwide databases of 20,248 families that included children with neurodevelopmental disorders and that were enriched for parental consanguinity. Approximately one third of affected children in these families presented with structural birth defects or microcephaly. We performed exome or genome sequencing of samples obtained from the children, their parents, or both to identify genes with biallelic pathogenic or likely pathogenic mutations present in more than one family. After identifying disease-causing variants, we generated two mouse models, each with a pathogenic variant "knocked in," to study mechanisms and test candidate treatments. We administered a small-molecule Wnt agonist to pregnant animals and assessed their offspring. RESULTS: We identified homozygous mutations in WLS, which encodes the Wnt ligand secretion mediator (also known as Wntless or WLS) in 10 affected persons from 5 unrelated families. (The Wnt ligand secretion mediator is essential for the secretion of all Wnt proteins.) Patients had multiorgan defects, including microcephaly and facial dysmorphism as well as foot syndactyly, renal agenesis, alopecia, iris coloboma, and heart defects. The mutations affected WLS protein stability and Wnt signaling. Knock-in mice showed tissue and cell vulnerability consistent with Wnt-signaling intensity and individual and collective functions of Wnts in embryogenesis. Administration of a pharmacologic Wnt agonist partially restored embryonic development. CONCLUSIONS: Genetic variations affecting a central Wnt regulator caused syndromic structural birth defects. Results from mouse models suggest that what we have named Zaki syndrome is a potentially preventable disorder. (Funded by the National Institutes of Health and others.).


Subject(s)
Abnormalities, Multiple/genetics , Congenital Abnormalities/genetics , Genetic Pleiotropy , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Receptors, G-Protein-Coupled/genetics , Wnt Proteins/metabolism , Animals , Disease Models, Animal , Fibroblasts/metabolism , Gene Knock-In Techniques , Genes, Recessive , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Pedigree , Phenotype , Receptors, G-Protein-Coupled/metabolism , Syndrome , Wnt Signaling Pathway
7.
Handb Exp Pharmacol ; 269: 3-28, 2021.
Article in English | MEDLINE | ID: mdl-34463853

ABSTRACT

Wnt signaling is critical for proper development of the embryo and for tissue homeostasis in the adult. Activation of this signaling cascade is initiated by binding of the secreted Wnts to their receptors. With the mammalian genome encoding multiple Wnts and Wnt receptors, a longstanding question in the field has been how Wnt-receptor specificities are achieved. Emerging from these studies is a picture of exquisite control over Wnt protein production, secretion, distribution, and receptor interactions, culminating in activation of downstream signaling cascades that control a myriad of biological processes. Here we discuss mechanisms by which Wnt protein activities are tuned and illustrate how the multiple layers of regulation can be leveraged for therapeutic interventions in disease.


Subject(s)
Frizzled Receptors , Wnt Signaling Pathway , Adult , Animals , Humans , Wnt Proteins
8.
Elife ; 92020 12 17.
Article in English | MEDLINE | ID: mdl-33331818

ABSTRACT

WNT proteins are secreted symmetry breaking signals that interact with cell surface receptors of the FZD family to regulate a multitude of developmental processes. Studying selectivity between WNTs and FZDs has been hampered by the paucity of purified WNT proteins and by their apparent non-selective interactions with the FZD receptors. Here, we describe an engineered protein, called F7L6, comprised of antibody-derived single-chain variable fragments, that selectively binds to human FZD7 and the co-receptor LRP6. F7L6 potently activates WNT/ß-catenin signaling in a manner similar to Wnt3a. In contrast to Wnt3a, F7L6 engages only FZD7 and none of the other FZD proteins. Treatment of human pluripotent stem (hPS) cells with F7L6 initiates transcriptional programs similar to those observed during primitive streak formation and subsequent gastrulation in the mammalian embryo. This demonstrates that selective engagement and activation of FZD7 signaling is sufficient to promote mesendodermal differentiation of hPS cells.


Subject(s)
Cell Differentiation/physiology , Frizzled Receptors/physiology , Mesoderm/embryology , Pluripotent Stem Cells/physiology , Blotting, Western , Gene Expression Regulation , Humans , Mesoderm/cytology , Mesoderm/growth & development , Real-Time Polymerase Chain Reaction , Recombinant Proteins , Wnt Signaling Pathway/physiology
9.
Nat Cell Biol ; 21(6): 721-730, 2019 06.
Article in English | MEDLINE | ID: mdl-31110287

ABSTRACT

Wnt signalling drives many processes in development, homeostasis and disease; however, the role and mechanism of individual ligand-receptor (Wnt-Frizzled (Fzd)) interactions in specific biological processes remain poorly understood. Wnt9a is specifically required for the amplification of blood progenitor cells during development. Using genetic studies in zebrafish and human embryonic stem cells, paired with in vitro cell biology and biochemistry, we determined that Wnt9a signals specifically through Fzd9b to elicit ß-catenin-dependent Wnt signalling that regulates haematopoietic stem and progenitor cell emergence. We demonstrate that the epidermal growth factor receptor (EGFR) is required as a cofactor for Wnt9a-Fzd9b signalling. EGFR-mediated phosphorylation of one tyrosine residue on the Fzd9b intracellular tail in response to Wnt9a promotes internalization of the Wnt9a-Fzd9b-LRP signalosome and subsequent signal transduction. These findings provide mechanistic insights for specific Wnt-Fzd signals, which will be crucial for specific therapeutic targeting and regenerative medicine.


Subject(s)
Hematopoietic Stem Cells/cytology , Receptors, Neurotransmitter/genetics , Wnt Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , ErbB Receptors/genetics , Humans , Phosphorylation , Wnt Signaling Pathway , Zebrafish/growth & development , beta Catenin/genetics
10.
Wiley Interdiscip Rev Syst Biol Med ; 10(5): e1422, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29600540

ABSTRACT

The Wnt signaling pathway is a highly conserved system that regulates complex biological processes across all metazoan species. At the cellular level, secreted Wnt proteins serve to break symmetry and provide cells with positional information that is critical to the patterning of the entire body plan. At the organismal level, Wnt signals are employed to orchestrate fundamental developmental processes, including the specification of the anterior-posterior body axis, induction of the primitive streak and ensuing gastrulation movements, and the generation of cell and tissue diversity. Wnt functions extend into adulthood where they regulate stem cell behavior, tissue homeostasis, and damage repair. Disruption of Wnt signaling activity during embryonic development or in adults results in a spectrum of abnormalities and diseases, including cancer. The molecular mechanisms that underlie the myriad of Wnt-regulated biological effects have been the subject of intense research for over three decades. This review is intended to summarize our current understanding of how Wnt signals are generated and interpreted. This article is categorized under: Biological Mechanisms > Cell Signaling Developmental Biology > Stem Cell Biology and Regeneration.

11.
Prog Mol Biol Transl Sci ; 153: 321-341, 2018 01.
Article in English | MEDLINE | ID: mdl-29389522

ABSTRACT

Leukemia and lymphoma are a wide encompassing term for a diverse set of blood malignancies that affect people of all ages and result in approximately 23,000 deaths in the United States per year (Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7-30.). Hematopoietic stem cells (HSCs) are tissue-specific stem cells at the apex of the hierarchy that gives rise to all of the terminally differentiated blood cells, through progressively restricted progenitor populations, a process that is known to be Wnt-responsive. In particular, the progenitor populations are subject to uncontrolled expansion during oncogenic processes, namely the common myeloid progenitor and common lymphoid progenitor, as well as the myeloblast and lymphoblast. Unregulated growth of these cell-types leads to mainly three types of blood cancers (i.e., leukemia, lymphoma, and myeloma), which frequently exhibit deregulation of the Wnt signaling pathway. Generally, leukemia is caused by the expansion of myeloid progenitors, leading to an overproduction of white blood cells; as such, patients are unable to make sufficient numbers of red blood cells and platelets. Likewise, an overproduction of lymphocytes leads to clogging of the lymph system and impairment of the immune system in lymphomas. Finally, cancer of the plasma cells in the blood is called myeloma, which also leads to immune system failure. Within each of these three types of blood cancers, there are multiple subtypes, usually characterized by their timeline of onset and their cell type of origin. Of these, 85% of leukemias are encompassed by the four most common diseases, that is, acute myeloid leukemia (AML), chronic myeloid leukemia (CML), acute lymphoblastic leukemia (ALL), and chronic lymphocytic leukemia (CLL); AML accounts for the majority of leukemia-related deaths (Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7-30.). Through understanding how HSCs are normally developed and maintained, we can understand how the normal functions of these pathways are disrupted during blood cancer progression; the Wnt pathway is important in regulation of both normal and malignant hematopoiesis. In this chapter, we will discuss the role of Wnt signaling in normal and aberrant hematopoiesis. Our understanding the relationship between Wnt and HSCs will provide novel insights into therapeutic targets.


Subject(s)
Hematologic Neoplasms/pathology , Hematopoietic Stem Cells/cytology , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Hematologic Neoplasms/metabolism , Hematopoietic Stem Cells/metabolism , Humans
12.
Genes (Basel) ; 9(2)2018 Jan 29.
Article in English | MEDLINE | ID: mdl-29382179

ABSTRACT

Hematopoietic stem cells (HSCs) differentiate into all cell types of the blood and can be used therapeutically to treat hematopoietic cancers and disorders. Despite decades of research, it is not yet possible to derive therapy-grade HSCs from pluripotent precursors. Analysis of HSC development in model organisms has identified some of the molecular cues that are necessary to instruct hematopoiesis in vivo, including Wnt9A, which is required during an early time window in zebrafish development. Although bona fide HSCs cannot be derived in vitro, it is possible to model human hematopoietic progenitor development by differentiating human pluripotent stem cells to hematopoietic cells. Herein, we modulate WNT9A expression during the in vitro differentiation of human embryonic stem cells to hematopoietic progenitor cells and demonstrate that WNT9A also regulates human hematopoietic progenitor cell development in vitro. Overexpression of WNT9A only impacts differentiation to CD34⁺/CD45⁺ cells during early time windows and does so in a dose-dependent manner. The cells that receive the Wnt signal-not the cells that secrete WNT9A-differentiate most efficiently to hematopoietic progenitors; this mimics the paracrine action of Wnt9a during in vivo hematopoiesis. Taken together, these data indicate that WNT9A is a conserved regulator of zebrafish and human hematopoietic development.

13.
Nat Commun ; 8(1): 1034, 2017 10 18.
Article in English | MEDLINE | ID: mdl-29044119

ABSTRACT

The WNT/ß-catenin signaling pathway is a prominent player in many developmental processes, including gastrulation, anterior-posterior axis specification, organ and tissue development, and homeostasis. Here, we use human pluripotent stem cells (hPSCs) to study the dynamics of the transcriptional response to exogenous activation of the WNT pathway. We describe a mechanism involving the WNT target gene SP5 that leads to termination of the transcriptional program initiated by WNT signaling. Integration of gene expression profiles of wild-type and SP5 mutant cells with genome-wide SP5 binding events reveals that SP5 acts to diminish expression of genes previously activated by the WNT pathway. Furthermore, we show that activation of SP5 by WNT signaling is most robust in cells with developmental potential, such as stem cells. These findings indicate a mechanism by which the developmental WNT signaling pathway reins in expression of transcriptional programs.


Subject(s)
DNA-Binding Proteins/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Wnt3A Protein/metabolism , Cell Line , DNA-Binding Proteins/genetics , Gene Expression Regulation , Humans , Pluripotent Stem Cells/cytology , Transcription Factors/genetics , Wnt Signaling Pathway , Wnt3A Protein/genetics , beta Catenin/genetics , beta Catenin/metabolism
14.
Dev Neurobiol ; 77(11): 1239-1259, 2017 11.
Article in English | MEDLINE | ID: mdl-28799266

ABSTRACT

The vertebrate central nervous system (CNS) is comprised of vast number of distinct cell types arranged in a highly organized manner. This high degree of complexity is achieved by cellular communication, including direct cell-cell contact, cell-matrix interactions, and cell-growth factor signaling. Among the several developmental signals controlling the development of the CNS, Wnt proteins have emerged as particularly critical and, hence, have captivated the attention of many researchers. With Wnts' evolutionarily conserved function as primordial symmetry breaking signals, these proteins and their downstream effects are responsible for simultaneously establishing cellular diversity and tissue organization. With their expansive repertoire of secreted agonists and antagonists, cell surface receptors, signaling cascades and downstream biological effects, Wnts are ideally suited to control the complex processes underlying vertebrate neural development. In this review, we will describe the mechanisms by which Wnts exert their potent effects on cells and tissues and highlight the many roles of Wnt signaling during neural development, starting from the initial induction of the neural plate, the subsequent patterning along the embryonic axes, to the intricately organized structure of the CNS. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1239-1259, 2017.


Subject(s)
Central Nervous System , Gene Expression Regulation, Developmental , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Body Patterning/physiology , Central Nervous System/embryology , Central Nervous System/growth & development , Central Nervous System/metabolism , Embryo, Nonmammalian/physiology , Vertebrates , Wnt Proteins/genetics , beta Catenin/genetics
15.
Sci Signal ; 10(482)2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28588081

ABSTRACT

Increased protein translation in cells and various factors in the tumor microenvironment can induce endoplasmic reticulum (ER) stress, which initiates the unfolded protein response (UPR). We have previously reported that factors released from cancer cells mounting a UPR induce a de novo UPR in bone marrow-derived myeloid cells, macrophages, and dendritic cells that facilitates protumorigenic characteristics in culture and tumor growth in vivo. We investigated whether this intercellular signaling, which we have termed transmissible ER stress (TERS), also operates between cancer cells and what its functional consequences were within the tumor. We found that TERS signaling induced a UPR in recipient human prostate cancer cells that included the cell surface expression of the chaperone GRP78. TERS also activated Wnt signaling in recipient cancer cells and enhanced resistance to nutrient starvation and common chemotherapies such as the proteasome inhibitor bortezomib and the microtubule inhibitor paclitaxel. TERS-induced activation of Wnt signaling required the UPR kinase and endonuclease IRE1. However, TERS-induced enhancement of cell survival was predominantly mediated by the UPR kinase PERK and a reduction in the abundance of the transcription factor ATF4, which prevented the activation of the transcription factor CHOP and, consequently, the induction of apoptosis. When implanted in mice, TERS-primed cancer cells gave rise to faster growing tumors than did vehicle-primed cancer cells. Collectively, our data demonstrate that TERS is a mechanism of intercellular communication through which tumor cells can adapt to stressful environments.


Subject(s)
Bortezomib/pharmacology , Cell Survival/drug effects , Drug Resistance , Endoplasmic Reticulum Stress/drug effects , Paclitaxel/pharmacology , Prostatic Neoplasms/pathology , Unfolded Protein Response/drug effects , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Antineoplastic Agents/pharmacology , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tubulin Modulators/pharmacology , Tumor Cells, Cultured , Wnt Proteins/genetics , Wnt Proteins/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
17.
Dev Cell ; 40(5): 439-452.e4, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28292423

ABSTRACT

Polarization of node cells along the anterior-posterior axis of mouse embryos is responsible for left-right symmetry breaking. How node cells become polarized has remained unknown, however. Wnt5a and Wnt5b are expressed posteriorly relative to the node, whereas genes for Sfrp inhibitors of Wnt signaling are expressed anteriorly. Here we show that polarization of node cells is impaired in Wnt5a-/-Wnt5b-/- and Sfrp mutant embryos, and also in the presence of a uniform distribution of Wnt5a or Sfrp1, suggesting that Wnt5 and Sfrp proteins act as instructive signals in this process. The absence of planar cell polarity (PCP) core proteins Prickle1 and Prickle2 in individual cells or local forced expression of Wnt5a perturbed polarization of neighboring wild-type cells. Our results suggest that opposing gradients of Wnt5a and Wnt5b and of their Sfrp inhibitors, together with intercellular signaling via PCP proteins, polarize node cells along the anterior-posterior axis for breaking of left-right symmetry.


Subject(s)
Body Patterning , Cell Polarity , Signal Transduction , Wnt Proteins/metabolism , Wnt-5a Protein/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Communication , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Models, Biological , Proteins/metabolism
18.
Zebrafish ; 14(4): 383-386, 2017 08.
Article in English | MEDLINE | ID: mdl-27829120

ABSTRACT

Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 has been applied to edit genomes in a wide variety of model systems. Although this process can be quite efficient, editing at precise locations in the genome remains difficult without a suitable single guide RNA (sgRNA). We have developed a method for screening sgRNA function in vitro, using reagents that most zebrafish laboratories are already using. The results from our in vitro assay correlate with function in vivo in every sgRNA that we have examined so far. When combined with endonucleases with alternative protospacer adjacent motif site specificities and alternative sgRNAs, this method will streamline genome editing at almost any locus.


Subject(s)
CRISPR-Cas Systems , Gene Editing , RNA, Guide, Kinetoplastida/genetics , Zebrafish/genetics , Animals , Endonucleases/metabolism , Gene Targeting , In Vitro Techniques , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/genetics
19.
Proc Natl Acad Sci U S A ; 113(46): 13150-13155, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27799526

ABSTRACT

Prodigiosin, a natural red pigment produced by numerous bacterial species, has exhibited promising anticancer activity; however, the molecular mechanisms of action of prodigiosin on malignant cells remain unclear. Aberrant activation of the Wnt/ß-catenin signaling cascade is associated with numerous human cancers. In this study, we identified prodigiosin as a potent inhibitor of the Wnt/ß-catenin pathway. Prodigiosin blocked Wnt/ß-catenin signaling by targeting multiple sites of this pathway, including the low-density lipoprotein-receptor-related protein (LRP) 6, Dishevelled (DVL), and glycogen synthase kinase-3ß (GSK3ß). In breast cancer MDA-MB-231 and MDA-MB-468 cells, nanomolar concentrations of prodigiosin decreased phosphorylation of LRP6, DVL2, and GSK3ß and suppressed ß-catenin-stimulated Wnt target gene expression, including expression of cyclin D1. In MDA-MB-231 breast cancer xenografts and MMTV-Wnt1 transgenic mice, administration of prodigiosin slowed tumor progression and reduced the expression of phosphorylated LRP6, phosphorylated and unphosphorylated DVL2, Ser9 phosphorylated GSK3ß, active ß-catenin, and cyclin D1. Through its ability to inhibit Wnt/ß-catenin signaling and reduce cyclin D1 levels, prodigiosin could have therapeutic activity in advanced breast cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Prodigiosin/pharmacology , Prodigiosin/therapeutic use , Wnt Signaling Pathway/drug effects , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D1/genetics , Dishevelled Proteins/genetics , Female , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Tumor Burden/drug effects , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics
20.
Cell Rep ; 17(6): 1595-1606, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27806298

ABSTRACT

All mature blood cell types in the adult animal arise from hematopoietic stem and progenitor cells (HSPCs). However, the developmental cues regulating HSPC ontogeny are incompletely understood. In particular, the details surrounding a requirement for Wnt/ß-catenin signaling in the development of mature HSPCs are controversial and difficult to consolidate. Using zebrafish, we demonstrate that Wnt signaling is required to direct an amplification of HSPCs in the aorta. Wnt9a is specifically required for this process and cannot be replaced by Wnt9b or Wnt3a. This proliferative event occurs independently of initial HSPC fate specification, and the Wnt9a input is required prior to aorta formation. HSPC arterial amplification occurs prior to seeding of secondary hematopoietic tissues and proceeds, in part, through the cell cycle regulator myca (c-myc). Our results support a general paradigm, in which early signaling events, including Wnt, direct later HSPC developmental processes.


Subject(s)
Aorta/cytology , Aorta/embryology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Wnt Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cell Count , Cell Cycle , Cell Proliferation , Hemangioblasts/metabolism , Wnt Signaling Pathway
SELECTION OF CITATIONS
SEARCH DETAIL
...