Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
Cell ; 173(2): 515-528.e17, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29625057

ABSTRACT

Bladder cancer is the fifth most prevalent cancer in the U.S., yet is understudied, and few laboratory models exist that reflect the biology of the human disease. Here, we describe a biobank of patient-derived organoid lines that recapitulates the histopathological and molecular diversity of human bladder cancer. Organoid lines can be established efficiently from patient biopsies acquired before and after disease recurrence and are interconvertible with orthotopic xenografts. Notably, organoid lines often retain parental tumor heterogeneity and exhibit a spectrum of genomic changes that are consistent with tumor evolution in culture. Analyses of drug response using bladder tumor organoids show partial correlations with mutational profiles, as well as changes associated with treatment resistance, and specific responses can be validated using xenografts in vivo. Our studies indicate that patient-derived bladder tumor organoids represent a faithful model system for studying tumor evolution and treatment response in the context of precision cancer medicine.


Subject(s)
Urinary Bladder Neoplasms/pathology , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Survival/drug effects , DNA Copy Number Variations , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred NOD , Middle Aged , Mutation , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Precision Medicine , Transplantation, Heterologous , Tumor Cells, Cultured , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism
2.
Cell ; 159(2): 402-14, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25303533

ABSTRACT

Identification of driver mutations in human diseases is often limited by cohort size and availability of appropriate statistical models. We propose a framework for the systematic discovery of genetic alterations that are causal determinants of disease, by prioritizing genes upstream of functional disease drivers, within regulatory networks inferred de novo from experimental data. We tested this framework by identifying the genetic determinants of the mesenchymal subtype of glioblastoma. Our analysis uncovered KLHL9 deletions as upstream activators of two previously established master regulators of the subtype, C/EBPß and C/EBPδ. Rescue of KLHL9 expression induced proteasomal degradation of C/EBP proteins, abrogated the mesenchymal signature, and reduced tumor viability in vitro and in vivo. Deletions of KLHL9 were confirmed in > 50% of mesenchymal cases in an independent cohort, thus representing the most frequent genetic determinant of the subtype. The method generalized to study other human diseases, including breast cancer and Alzheimer's disease.


Subject(s)
Algorithms , Gene Regulatory Networks , Glioblastoma/genetics , Mutation , Alzheimer Disease/genetics , Animals , Breast Neoplasms/genetics , CCAAT-Enhancer-Binding Protein-delta/metabolism , DNA Copy Number Variations , Glioblastoma/pathology , Heterografts , Humans , Mice , Neoplasm Transplantation , Proteasome Endopeptidase Complex/metabolism , Proteins/metabolism , Quantitative Trait Loci , Ubiquitination
4.
Proc Natl Acad Sci U S A ; 120(31): e2301972120, 2023 08.
Article in English | MEDLINE | ID: mdl-37487079

ABSTRACT

PARP1 (poly-ADP ribose polymerase 1) is recruited and activated by DNA strand breaks, catalyzing the generation of poly-ADP-ribose (PAR) chains from NAD+. PAR relaxes chromatin and recruits other DNA repair factors, including XRCC1 and DNA Ligase 3, to maintain genomic stability. Here we show that, in contrast to the normal development of Parp1-null mice, heterozygous expression of catalytically inactive Parp1 (E988A, Parp1+/A) acts in a dominant-negative manner to disrupt murine embryogenesis. As such, all the surviving F1 Parp1+/A mice are chimeras with mixed Parp1+/AN (neoR retention) cells that act similarly to Parp1+/-. Pure F2 Parp1+/A embryos were found at Mendelian ratios at the E3.5 blastocyst stage but died before E9.5. Compared to Parp1-/- cells, genotype and expression-validated pure Parp1+/A cells retain significant ADP-ribosylation and PARylation activities but accumulate markedly higher levels of sister chromatid exchange and mitotic bridges. Despite proficiency for homologous recombination and nonhomologous end-joining measured by reporter assays and supported by normal lymphocyte and germ cell development, Parp1+/A cells are hypersensitive to base damages, radiation, and Topoisomerase I and II inhibition. The sensitivity of Parp1+/A cells to base damages and Topo inhibitors exceed Parp1-/- controls. The findings show that the enzymatically inactive PARP1 dominant negatively blocks DNA repair in selective pathways beyond wild-type PARP1 and establishes a crucial physiological difference between PARP1 inactivation vs. deletion. As a result, the expression of enzymatically inactive PARP1 from one allele is sufficient to abrogate murine embryonic development, providing a mechanism for the on-target side effect of PARP inhibitors used for cancer therapy.


Subject(s)
ADP-Ribosylation , Genomic Instability , Female , Pregnancy , Animals , Mice , Causality , Alleles , Genotype
5.
EMBO Rep ; 23(5): e54049, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35253958

ABSTRACT

The healthy prostate is a relatively quiescent tissue. Yet, prostate epithelium overgrowth is a common condition during aging, associated with urinary dysfunction and tumorigenesis. For over thirty years, TGF-ß ligands have been known to induce cytostasis in a variety of epithelia, but the intracellular pathway mediating this signal in the prostate, and its relevance for quiescence, have remained elusive. Here, using mouse prostate organoids to model epithelial progenitors, we find that intra-epithelial non-canonical Activin A signaling inhibits cell proliferation in a Smad-independent manner. Mechanistically, Activin A triggers Tak1 and p38 ΜAPK activity, leading to p16 and p21 nuclear import. Spontaneous evasion from this quiescent state occurs upon prolonged culture, due to reduced Activin A secretion, a condition associated with DNA replication stress and aneuploidy. Organoids capable to escape quiescence in vitro are also able to implant with increased frequency into immunocompetent mice. This study demonstrates that non-canonical Activin A signaling safeguards epithelial quiescence in the healthy prostate, with potential implications for the understanding of cancer initiation, and the development of therapies targeting quiescent tumor progenitors.


Subject(s)
Activins , Prostate , Activins/metabolism , Animals , Male , Mice , Prostate/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
6.
Genes Dev ; 30(4): 399-407, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26883359

ABSTRACT

Understanding new therapeutic paradigms for both castrate-sensitive and more aggressive castrate-resistant prostate cancer is essential to improve clinical outcomes. As a critically important cellular process, autophagy promotes stress tolerance by recycling intracellular components to sustain metabolism important for tumor survival. To assess the importance of autophagy in prostate cancer, we generated a new autochthonous genetically engineered mouse model (GEMM) with inducible prostate-specific deficiency in the Pten tumor suppressor and autophagy-related-7 (Atg7) genes. Atg7 deficiency produced an autophagy-deficient phenotype and delayed Pten-deficient prostate tumor progression in both castrate-naïve and castrate-resistant cancers. Atg7-deficient tumors display evidence of endoplasmic reticulum (ER) stress, suggesting that autophagy may promote prostate tumorigenesis through management of protein homeostasis. Taken together, these data support the importance of autophagy for both castrate-naïve and castrate-resistant growth in a newly developed GEMM, suggesting a new paradigm and model to study approaches to inhibit autophagy in combination with known and new therapies for advanced prostate cancer.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Microtubule-Associated Proteins/metabolism , Prostatic Neoplasms/physiopathology , Animals , Animals, Genetically Modified , Autophagy/genetics , Autophagy-Related Protein 7 , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Gene Deletion , Male , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/physiopathology , Signal Transduction/genetics
7.
Semin Cancer Biol ; 82: 60-67, 2022 07.
Article in English | MEDLINE | ID: mdl-34147640

ABSTRACT

Although prostate adenocarcinoma lacks distinguishable histopathological subtypes, prostate cancer displays significant inter- and intratumor heterogeneity at the molecular level and with respect to disease prognosis and treatment response. In principle, understanding the basis for prostate cancer heterogeneity can help distinguish aggressive from indolent disease, and help overcome castration-resistance in advanced prostate cancer. In this review, we will discuss recent advances in understanding the cell types of origin, putative cancer stem cells, and tumor plasticity in prostate cancer, focusing on insights from studies of genetically engineered mouse models (GEMMs). We will also outline future directions for investigating tumor heterogeneity using mouse models of prostate cancer.


Subject(s)
Prostate , Prostatic Neoplasms , Animals , Disease Models, Animal , Humans , Male , Mice , Neoplastic Stem Cells/pathology , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy
8.
Proc Natl Acad Sci U S A ; 117(49): 30907-30917, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33219121

ABSTRACT

Myostatin (MSTN) is a transforming growth factor-ß (TGF-ß) family member that normally acts to limit muscle growth. The function of MSTN is partially redundant with that of another TGF-ß family member, activin A. MSTN and activin A are capable of signaling through a complex of type II and type I receptors. Here, we investigated the roles of two type II receptors (ACVR2 and ACVR2B) and two type I receptors (ALK4 and ALK5) in the regulation of muscle mass by these ligands by genetically targeting these receptors either alone or in combination specifically in myofibers in mice. We show that targeting signaling in myofibers is sufficient to cause significant increases in muscle mass, showing that myofibers are the direct target for signaling by these ligands in the regulation of muscle growth. Moreover, we show that there is functional redundancy between the two type II receptors as well as between the two type I receptors and that all four type II/type I receptor combinations are utilized in vivo. Targeting signaling specifically in myofibers also led to reductions in overall body fat content and improved glucose metabolism in mice fed either regular chow or a high-fat diet, demonstrating that these metabolic effects are the result of enhanced muscling. We observed no effect, however, on either bone density or muscle regeneration in mice in which signaling was targeted in myofibers. The latter finding implies that MSTN likely signals to other cells, such as satellite cells, in addition to myofibers to regulate muscle homeostasis.


Subject(s)
Activin Receptors, Type II/metabolism , Activin Receptors, Type I/metabolism , Activins/metabolism , Muscle Development , Myostatin/metabolism , Animals , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Muscles/metabolism , Organ Size
9.
Development ; 144(8): 1382-1398, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28400434

ABSTRACT

Prostate organogenesis is a complex process that is primarily mediated by the presence of androgens and subsequent mesenchyme-epithelial interactions. The investigation of prostate development is partly driven by its potential relevance to prostate cancer, in particular the apparent re-awakening of key developmental programs that occur during tumorigenesis. However, our current knowledge of the mechanisms that drive prostate organogenesis is far from complete. Here, we provide a comprehensive overview of prostate development, focusing on recent findings regarding sexual dimorphism, bud induction, branching morphogenesis and cellular differentiation.


Subject(s)
Hormones/metabolism , Organogenesis , Prostate/cytology , Prostate/embryology , Animals , Cell Differentiation , Epithelium/embryology , Humans , Male , Prostate/anatomy & histology , Sex Characteristics
10.
Development ; 142(22): 3821-32, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26417042

ABSTRACT

The secreted glycoprotein sonic hedgehog (Shh) is expressed in the prechordal mesoderm, where it plays a crucial role in induction and patterning of the ventral forebrain. Currently little is known about how Shh is regulated in prechordal tissue. Here we show that in the embryonic chick, Shh is expressed transiently in prechordal mesoderm, and is governed by unprocessed Nodal. Exposure of prechordal mesoderm microcultures to Nodal-conditioned medium, the Nodal inhibitor CerS, or to an ALK4/5/7 inhibitor reveals that Nodal is required to maintain both Shh and Gsc expression, but whereas Gsc is largely maintained through canonical signalling, Nodal signals through a non-canonical route to maintain Shh. Further, Shh expression can be maintained by a recombinant Nodal cleavage mutant, proNodal, but not by purified mature Nodal. A number of lines of evidence suggest that proNodal acts via FGFR3. ProNodal and FGFR3 co-immunoprecipitate and proNodal increases FGFR3 tyrosine phosphorylation. In microcultures, soluble FGFR3 abolishes Shh without affecting Gsc expression. Further, prechordal mesoderm cells in which Fgfr3 expression is reduced by Fgfr3 siRNA fail to bind to proNodal. Finally, targeted electroporation of Fgfr3 siRNA to prechordal mesoderm in vivo results in premature Shh downregulation without affecting Gsc. We report an inverse correlation between proNodal-FGFR3 signalling and pSmad1/5/8, and show that proNodal-FGFR3 signalling antagonises BMP-mediated pSmad1/5/8 signalling, which is poised to downregulate Shh. Our studies suggest that proNodal/FGFR3 signalling governs Shh duration by repressing canonical BMP signalling, and that local BMPs rapidly silence Shh once endogenous Nodal-FGFR3 signalling is downregulated.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/metabolism , Mesoderm/embryology , Nodal Protein/metabolism , Prosencephalon/embryology , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Signal Transduction/physiology , Animals , Chick Embryo , Electroporation , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Mesoderm/metabolism , Nodal Protein/antagonists & inhibitors , RNA, Small Interfering/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Smad Proteins/metabolism
11.
Genes Dev ; 24(18): 1967-2000, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20844012

ABSTRACT

Despite much recent progress, prostate cancer continues to represent a major cause of cancer-related mortality and morbidity in men. Since early studies on the role of the androgen receptor that led to the advent of androgen deprivation therapy in the 1940s, there has long been intensive interest in the basic mechanisms underlying prostate cancer initiation and progression, as well as the potential to target these processes for therapeutic intervention. Here, we present an overview of major themes in prostate cancer research, focusing on current knowledge of principal events in cancer initiation and progression. We discuss recent advances, including new insights into the mechanisms of castration resistance, identification of stem cells and tumor-initiating cells, and development of mouse models for preclinical evaluation of novel therapuetics. Overall, we highlight the tremendous research progress made in recent years, and underscore the challenges that lie ahead.


Subject(s)
Prostatic Neoplasms/genetics , Animals , Castration , Disease Progression , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy
12.
Genes Dev ; 23(6): 675-80, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19261747

ABSTRACT

Although bladder cancer represents a serious health problem worldwide, relevant mouse models for investigating disease progression or therapeutic targets have been lacking. We show that combined deletion of p53 and Pten in bladder epithelium leads to invasive cancer in a novel mouse model. Inactivation of p53 and PTEN promotes tumorigenesis in human bladder cells and is correlated with poor survival in human tumors. Furthermore, the synergistic effects of p53 and Pten deletion are mediated by deregulation of mammalian target of rapamycin (mTOR) signaling, consistent with the ability of rapamycin to block bladder tumorigenesis in preclinical studies. Our integrated analyses of mouse and human bladder cancer provide a rationale for investigating mTOR inhibition for treatment of patients with invasive disease.


Subject(s)
Carcinoma, Transitional Cell/pathology , Cell Transformation, Neoplastic , Disease Models, Animal , PTEN Phosphohydrolase/metabolism , Tumor Suppressor Protein p53/metabolism , Urinary Bladder Neoplasms/pathology , Animals , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/metabolism , Humans , Lymphatic Metastasis , Male , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Invasiveness , PTEN Phosphohydrolase/genetics , Protein Kinases/physiology , Rats , Signal Transduction , TOR Serine-Threonine Kinases , Tumor Suppressor Protein p53/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
13.
Am J Pathol ; 185(11): 2907-22, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26429739

ABSTRACT

Cripto-1, a member of the epidermal growth factor-Cripto-1/FRL-1/Cryptic family, is critical for early embryonic development. Together with its ligand Nodal, Cripto-1 has been found to be associated with the undifferentiated status of mouse and human embryonic stem cells. Several studies have clearly shown that Cripto-1 is involved in regulating branching morphogenesis and epithelial-mesenchymal transition of the mammary gland both in vitro and in vivo and together with the cofactor GRP78 is critical for the maintenance of mammary stem cells ex vivo. Our previous studies showed that mammary-specific overexpression of human Cripto-1 exhibited dramatic morphological alterations in nulliparous mice mammary glands. The present study shows a novel mechanism for Cripto-1 regulation of mammary gland development through direct effects on progesterone receptor expression and pathways regulated by progesterone in the mammary gland. We demonstrate a strict temporal regulation of mouse Cripto-1 (mCripto-1) expression that occurs during mammary gland development and a stage-specific function of mCripto-1 signaling during mammary gland development. Our data suggest that Cripto-1, like the progesterone receptor, is not required for the initial ductal growth but is essential for subsequent side branching and alveologenesis during the initial stages of pregnancy. Dissection of the mechanism by which this occurs indicates that mCripto-1 activates receptor activator NF-κB/receptor activator NF-κB ligand, and NF-κB signaling pathways.


Subject(s)
Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , NF-kappa B p50 Subunit/metabolism , Neoplasm Proteins/metabolism , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Animals , Cell Proliferation , Endoplasmic Reticulum Chaperone BiP , Epidermal Growth Factor/genetics , Epithelial Cells , Epithelial-Mesenchymal Transition , Female , Humans , Mammary Glands, Animal/cytology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Models, Biological , NF-kappa B p50 Subunit/genetics , Neoplasm Proteins/genetics , Organ Specificity , Pregnancy , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptors, Progesterone/genetics
15.
Proc Natl Acad Sci U S A ; 110(37): E3506-15, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23918374

ABSTRACT

Combinatorial activation of PI3-kinase and RAS signaling occurs frequently in advanced prostate cancer and is associated with adverse patient outcome. We now report that the oncogenic Ets variant 4 (Etv4) promotes prostate cancer metastasis in response to coactivation of PI3-kinase and Ras signaling pathways in a genetically engineered mouse model of highly penetrant, metastatic prostate cancer. Using an inducible Cre driver to simultaneously inactivate Pten while activating oncogenic Kras and a fluorescent reporter allele in the prostate epithelium, we performed lineage tracing in vivo to define the temporal and spatial occurrence of prostate tumors, disseminated tumor cells, and metastases. These analyses revealed that though disseminated tumors cells arise early following the initial occurrence of prostate tumors, there is a significant temporal lag in metastasis, which is temporally coincident with the up-regulation of Etv4 expression in primary tumors. Functional studies showed that knockdown of Etv4 in a metastatic cell line derived from the mouse model abrogates the metastatic phenotype but does not affect tumor growth. Notably, expression and activation of ETV4, but not other oncogenic ETS genes, is correlated with activation of both PI3-kinase and Ras signaling in human prostate tumors and metastases. Our findings indicate that ETV4 promotes metastasis in prostate tumors that have activation of PI3-kinase and Ras signaling, and therefore, ETV4 represents a potential target of therapeutic intervention for metastatic prostate cancer.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/secondary , Proto-Oncogene Proteins c-ets/metabolism , ras Proteins/metabolism , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Knockdown Techniques , Genes, ras , Genetic Engineering , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Oncogenes , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/genetics , Signal Transduction , Transcription Factors/genetics , Up-Regulation
16.
Cancer Cell ; 12(6): 495-7, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18068626

ABSTRACT

Members of the fibroblast growth factor (FGF) family are believed to play critical roles during organogenesis and carcinogenesis via signaling between epithelial and stromal compartments. Two new studies in this issue of Cancer Cell underscore the importance of FGF signaling in mediating epithelial-stromal interactions during prostate carcinogenesis. These papers show that deregulated FGF signaling in mouse models of prostate cancer leads to cancer progression and promotes an epithelial-mesenchymal transition, suggesting that FGF receptor inhibitors may have therapeutic value for prostate cancer treatment.


Subject(s)
Cell Communication , Epithelial Cells/pathology , Fibroblast Growth Factors/metabolism , Prostatic Neoplasms/pathology , Signal Transduction , Stromal Cells/pathology , Animals , Fibroblast Growth Factor 10/metabolism , Humans , Male , Mice , Receptor, Fibroblast Growth Factor, Type 1/metabolism
17.
Nature ; 461(7263): 495-500, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19741607

ABSTRACT

In epithelial tissues, the lineage relationship between normal progenitor cells and cell type(s) of origin for cancer has been poorly understood. Here we show that a known regulator of prostate epithelial differentiation, the homeobox gene Nkx3-1, marks a stem cell population that functions during prostate regeneration. Genetic lineage-marking demonstrates that rare luminal cells that express Nkx3-1 in the absence of testicular androgens (castration-resistant Nkx3-1-expressing cells, CARNs) are bipotential and can self-renew in vivo, and single-cell transplantation assays show that CARNs can reconstitute prostate ducts in renal grafts. Functional assays of Nkx3-1 mutant mice in serial prostate regeneration suggest that Nkx3-1 is required for stem cell maintenance. Furthermore, targeted deletion of the Pten tumour suppressor gene in CARNs results in rapid carcinoma formation after androgen-mediated regeneration. These observations indicate that CARNs represent a new luminal stem cell population that is an efficient target for oncogenic transformation in prostate cancer.


Subject(s)
Cell Lineage , Epithelial Cells/pathology , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology , Androgens/deficiency , Androgens/metabolism , Animals , Castration , Cell Differentiation , Cell Division , Cell Transformation, Neoplastic , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Kidney , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/transplantation , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Regeneration , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Bioessays ; 35(3): 253-60, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23027425

ABSTRACT

Recent studies of prostate cancer and other tumor types have revealed significant support, as well as unexpected complexities, for the application of concepts from normal stem cell biology to cancer. In particular, the cell of origin and cancer stem cell models have been proposed to explain the heterogeneity of tumors during the initiation, propagation, and evolution of cancer. Thus, a basis of intertumor heterogeneity has emerged from studies investigating whether stem cells and/or non-stem cells can serve as cells of origin for cancer and give rise to tumor subtypes that vary in disease outcome. Furthermore, analyses of putative cancer stem cells have revealed the genetically diverse nature of cancers and expanded our understanding of intratumor heterogeneity and clonal evolution. Overall, the principles that have emerged from these stem cell studies highlight the challenges to be surmounted to develop effective treatment strategies for cancer.


Subject(s)
Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Clone Cells , Humans , Models, Biological
19.
Proc Natl Acad Sci U S A ; 109(47): E3231-40, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23129614

ABSTRACT

Skeletal muscle regeneration mainly depends on satellite cells, a population of resident muscle stem cells. However, our understanding of the molecular mechanisms underlying satellite cell activation is still largely undefined. Here, we show that Cripto, a regulator of early embryogenesis, is a novel regulator of muscle regeneration and satellite cell progression toward the myogenic lineage. Conditional inactivation of cripto in adult satellite cells compromises skeletal muscle regeneration, whereas gain of function of Cripto accelerates regeneration, leading to muscle hypertrophy. Moreover, we provide evidence that Cripto modulates myogenic cell determination and promotes proliferation by antagonizing the TGF-ß ligand myostatin. Our data provide unique insights into the molecular and cellular basis of Cripto activity in skeletal muscle regeneration and raise previously undescribed implications for stem cell biology and regenerative medicine.


Subject(s)
Cell Lineage , Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , Muscle, Skeletal/physiology , Myostatin/antagonists & inhibitors , Neoplasm Proteins/metabolism , Regeneration , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Aging/metabolism , Animals , Cell Proliferation , Gene Deletion , Gene Targeting , Hypertrophy , Mice , Mice, Inbred C57BL , Models, Animal , Muscle Development , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts/metabolism , Myoblasts/pathology , Myostatin/metabolism , Signal Transduction
20.
Development ; 138(18): 3885-95, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21862554

ABSTRACT

The signaling pathway for Nodal, a ligand of the TGFß superfamily, plays a central role in regulating the differentiation and/or maintenance of stem cell types that can be derived from the peri-implantation mouse embryo. Extra-embryonic endoderm stem (XEN) cells resemble the primitive endoderm of the blastocyst, which normally gives rise to the parietal and the visceral endoderm in vivo, but XEN cells do not contribute efficiently to the visceral endoderm in chimeric embryos. We have found that XEN cells treated with Nodal or Cripto (Tdgf1), an EGF-CFC co-receptor for Nodal, display upregulation of markers for visceral endoderm as well as anterior visceral endoderm (AVE), and can contribute to visceral endoderm and AVE in chimeric embryos. In culture, XEN cells do not express Cripto, but do express the related EGF-CFC co-receptor Cryptic (Cfc1), and require Cryptic for Nodal signaling. Notably, the response to Nodal is inhibited by the Alk4/Alk5/Alk7 inhibitor SB431542, but the response to Cripto is unaffected, suggesting that the activity of Cripto is at least partially independent of type I receptor kinase activity. Gene set enrichment analysis of genome-wide expression signatures generated from XEN cells under these treatment conditions confirmed the differing responses of Nodal- and Cripto-treated XEN cells to SB431542. Our findings define distinct pathways for Nodal and Cripto in the differentiation of visceral endoderm and AVE from XEN cells and provide new insights into the specification of these cell types in vivo.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/physiology , Endoderm/cytology , Endoderm/physiology , Epidermal Growth Factor/physiology , Membrane Glycoproteins/physiology , Neoplasm Proteins/physiology , Nodal Protein/physiology , Animals , Cell Differentiation/drug effects , Cell Line , Embryo, Mammalian , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endoderm/drug effects , Endoderm/metabolism , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Extraembryonic Membranes/cytology , Extraembryonic Membranes/physiology , Female , Gene Expression Profiling , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred ICR , Mice, Transgenic , Microarray Analysis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/pharmacology , Nodal Protein/genetics , Nodal Protein/metabolism , Nodal Protein/pharmacology , Pregnancy , Signal Transduction/drug effects , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL