2022
|
Rim, EY; Clevers, H; Nusse, R The Wnt Pathway: From Signaling Mechanisms to Synthetic Modulators. Journal Article In: Annual review of biochemistry, 2022, ISSN: 0066-4154. @article{455,
title = {The Wnt Pathway: From Signaling Mechanisms to Synthetic Modulators.},
author = {EY Rim and H Clevers and R Nusse},
url = {https://arjournals.annualreviews.org/doi/10.1146/annurev-biochem-040320-103615?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%3dpubmed},
doi = {10.1146/annurev-biochem-040320-103615},
issn = {0066-4154},
year = {2022},
date = {2022-02-01},
journal = {Annual review of biochemistry},
abstract = {The Wnt pathway is central to a host of developmental and disease-related processes. The remarkable conservation of this intercellular signaling cascade throughout metazoan lineages indicates that it coevolved with multicellularity to regulate the generation and spatial arrangement of distinct cell types. By regulating cell fate specification, mitotic activity, and cell polarity, Wnt signaling orchestrates development and tissue homeostasis, and its dysregulation is implicated in developmental defects, cancer, and degenerative disorders. We review advances in our understanding of this key pathway, from Wnt protein production and secretion to relay of the signal in the cytoplasm of the receiving cell. We discuss the evolutionary history of this pathway as well as endogenous and synthetic modulators of its activity. Finally, we highlight remaining gaps in our knowledge of Wnt signal transduction and avenues for future research. Expected final online publication date for the , Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The Wnt pathway is central to a host of developmental and disease-related processes. The remarkable conservation of this intercellular signaling cascade throughout metazoan lineages indicates that it coevolved with multicellularity to regulate the generation and spatial arrangement of distinct cell types. By regulating cell fate specification, mitotic activity, and cell polarity, Wnt signaling orchestrates development and tissue homeostasis, and its dysregulation is implicated in developmental defects, cancer, and degenerative disorders. We review advances in our understanding of this key pathway, from Wnt protein production and secretion to relay of the signal in the cytoplasm of the receiving cell. We discuss the evolutionary history of this pathway as well as endogenous and synthetic modulators of its activity. Finally, we highlight remaining gaps in our knowledge of Wnt signal transduction and avenues for future research. Expected final online publication date for the , Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates. |
2021
|
Habara, O; Logan, C Y; Kanai-Azuma, M; Nusse, R; Takase, HM WNT signaling in pre-granulosa cells is required for ovarian folliculogenesis and female fertility. Journal Article In: Development (Cambridge, England), vol. 148, 2021, ISSN: 0950-1991. @article{453,
title = {WNT signaling in pre-granulosa cells is required for ovarian folliculogenesis and female fertility.},
author = {O Habara and C Y Logan and M Kanai-Azuma and R Nusse and HM Takase},
url = {https://journals.biologists.com/dev/article-lookup/doi/10.1242/dev.198846},
doi = {10.1242/dev.198846},
issn = {0950-1991},
year = {2021},
date = {2021-05-01},
journal = {Development (Cambridge, England)},
volume = {148},
abstract = {In mammalian ovaries, immature oocytes are reserved in primordial follicles until their activation for potential ovulation. Precise control of primordial follicle activation (PFA) is essential for reproduction, but how this is achieved is unclear. Here, we show that canonical wingless-type MMTV integration site family (WNT) signaling is pivotal for pre-granulosa cell (pre-GC) activation during PFA. We identified several WNT ligands expressed in pre-GCs that act in an autocrine manner. Inhibition of WNT secretion from pre-GCs/GCs by conditional knockout (cKO) of the wntless (Wls) gene led to female infertility. In Wls cKO mice, GC layer thickness was greatly reduced in growing follicles, which resulted in impaired oocyte growth with both an abnormal, sustained nuclear localization of forkhead box O3 (FOXO3) and reduced phosphorylation of ribosomal protein S6 (RPS6). Constitutive stabilization of β-catenin (CTNNB1) in pre-GCs/GCs induced morphological changes of pre-GCs from a squamous into a cuboidal form, though it did not influence oocyte activation. Our results reveal that canonical WNT signaling plays a permissive role in the transition of pre-GCs to GCs, which is an essential step to support oocyte growth.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
In mammalian ovaries, immature oocytes are reserved in primordial follicles until their activation for potential ovulation. Precise control of primordial follicle activation (PFA) is essential for reproduction, but how this is achieved is unclear. Here, we show that canonical wingless-type MMTV integration site family (WNT) signaling is pivotal for pre-granulosa cell (pre-GC) activation during PFA. We identified several WNT ligands expressed in pre-GCs that act in an autocrine manner. Inhibition of WNT secretion from pre-GCs/GCs by conditional knockout (cKO) of the wntless (Wls) gene led to female infertility. In Wls cKO mice, GC layer thickness was greatly reduced in growing follicles, which resulted in impaired oocyte growth with both an abnormal, sustained nuclear localization of forkhead box O3 (FOXO3) and reduced phosphorylation of ribosomal protein S6 (RPS6). Constitutive stabilization of β-catenin (CTNNB1) in pre-GCs/GCs induced morphological changes of pre-GCs from a squamous into a cuboidal form, though it did not influence oocyte activation. Our results reveal that canonical WNT signaling plays a permissive role in the transition of pre-GCs to GCs, which is an essential step to support oocyte growth. |
Russell, JP; Lim, X; Santambrogio, A; Yianni, V; Kemkem, Y; Wang, B; Fish, M; Haston, S; Grabek, A; Hallang, S; Lodge, EJ; Patist, AL; Schedl, A; Mollard, P; Nusse, R; Andoniadou, CL Pituitary stem cells produce paracrine WNT signals to control the expansion of their descendant progenitor cells. Journal Article In: eLife, vol. 10, 2021. @article{449,
title = {Pituitary stem cells produce paracrine WNT signals to control the expansion of their descendant progenitor cells.},
author = {JP Russell and X Lim and A Santambrogio and V Yianni and Y Kemkem and B Wang and M Fish and S Haston and A Grabek and S Hallang and EJ Lodge and AL Patist and A Schedl and P Mollard and R Nusse and CL Andoniadou},
url = {https://doi.org/10.7554/eLife.59142},
doi = {10.7554/eLife.59142},
year = {2021},
date = {2021-01-01},
journal = {eLife},
volume = {10},
abstract = {In response to physiological demand, the pituitary gland generates new hormone-secreting cells from committed progenitor cells throughout life. It remains unclear to what extent pituitary stem cells (PSCs), which uniquely express SOX2, contribute to pituitary growth and renewal. Moreover, neither the signals that drive proliferation nor their sources have been elucidated. We have used genetic approaches in the mouse, showing that the WNT pathway is essential for proliferation of all lineages in the gland. We reveal that SOX2 stem cells are a key source of WNT ligands. By blocking secretion of WNTs from SOX2 PSCs in vivo, we demonstrate that proliferation of neighbouring committed progenitor cells declines, demonstrating that progenitor multiplication depends on the paracrine WNT secretion from SOX2 PSCs. Our results indicate that stem cells can hold additional roles in tissue expansion and homeostasis, acting as paracrine signalling centres to coordinate the proliferation of neighbouring cells.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
In response to physiological demand, the pituitary gland generates new hormone-secreting cells from committed progenitor cells throughout life. It remains unclear to what extent pituitary stem cells (PSCs), which uniquely express SOX2, contribute to pituitary growth and renewal. Moreover, neither the signals that drive proliferation nor their sources have been elucidated. We have used genetic approaches in the mouse, showing that the WNT pathway is essential for proliferation of all lineages in the gland. We reveal that SOX2 stem cells are a key source of WNT ligands. By blocking secretion of WNTs from SOX2 PSCs in vivo, we demonstrate that proliferation of neighbouring committed progenitor cells declines, demonstrating that progenitor multiplication depends on the paracrine WNT secretion from SOX2 PSCs. Our results indicate that stem cells can hold additional roles in tissue expansion and homeostasis, acting as paracrine signalling centres to coordinate the proliferation of neighbouring cells. |
Winter, TJJ; Nusse, R Running Against the Wnt: How Wnt/β-Catenin Suppresses Adipogenesis. Journal Article In: Frontiers in cell and developmental biology, vol. 9, pp. 627429, 2021. @article{451,
title = {Running Against the Wnt: How Wnt/β-Catenin Suppresses Adipogenesis.},
author = {TJJ Winter and R Nusse},
url = {https://doi.org/10.3389/fcell.2021.627429},
doi = {10.3389/fcell.2021.627429},
year = {2021},
date = {2021-00-01},
journal = {Frontiers in cell and developmental biology},
volume = {9},
pages = {627429},
abstract = {Mesenchymal stem cells (MSCs) give rise to adipocytes, osteocytes, and chondrocytes and reside in various tissues, including bone marrow and adipose tissue. The differentiation choices of MSCs are controlled by several signaling pathways, including the Wnt/β-catenin signaling. When MSCs undergo adipogenesis, they first differentiate into preadipocytes, a proliferative adipocyte precursor cell, after which they undergo terminal differentiation into mature adipocytes. These two steps are controlled by the Wnt/β-catenin pathway, in such a way that when signaling is abrogated, the next step in adipocyte differentiation can start. This sequence suggests that the main role of Wnt/β-catenin signaling is to suppress differentiation while increasing MSC and preadipocytes cell mass. During later steps of MSC differentiation, however, active Wnt signaling can promote osteogenesis instead of keeping the MSCs undifferentiated and proliferative. The exact mechanisms behind the various functions of Wnt signaling remain elusive, although recent research has revealed that during lineage commitment of MSCs into preadipocytes, Wnt signaling is inactivated by endogenous Wnt inhibitors. In part, this process is regulated by histone-modifying enzymes, which can lead to increased or decreased Wnt gene expression. The role of Wnt in adipogenesis, as well as in osteogenesis, has implications for metabolic diseases since Wnt signaling may serve as a therapeutic target.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Mesenchymal stem cells (MSCs) give rise to adipocytes, osteocytes, and chondrocytes and reside in various tissues, including bone marrow and adipose tissue. The differentiation choices of MSCs are controlled by several signaling pathways, including the Wnt/β-catenin signaling. When MSCs undergo adipogenesis, they first differentiate into preadipocytes, a proliferative adipocyte precursor cell, after which they undergo terminal differentiation into mature adipocytes. These two steps are controlled by the Wnt/β-catenin pathway, in such a way that when signaling is abrogated, the next step in adipocyte differentiation can start. This sequence suggests that the main role of Wnt/β-catenin signaling is to suppress differentiation while increasing MSC and preadipocytes cell mass. During later steps of MSC differentiation, however, active Wnt signaling can promote osteogenesis instead of keeping the MSCs undifferentiated and proliferative. The exact mechanisms behind the various functions of Wnt signaling remain elusive, although recent research has revealed that during lineage commitment of MSCs into preadipocytes, Wnt signaling is inactivated by endogenous Wnt inhibitors. In part, this process is regulated by histone-modifying enzymes, which can lead to increased or decreased Wnt gene expression. The role of Wnt in adipogenesis, as well as in osteogenesis, has implications for metabolic diseases since Wnt signaling may serve as a therapeutic target. |
2020
|
Miao, Y; Ha, A; Lau, W; Yuki, K; Santos, AJM; You, C; Geurts, MH; Puschhof, J; Pleguezuelos-Manzano, C; Peng, WC; Senlice, R; Piani, C; Buikema, JW; Gbenedio, OM; Vallon, M; Yuan, J; Haan, S; Hemrika, W; Rösch, K; Dang, LT; Baker, D; Ott, M; Depeille, P; Wu, SM; Drost, J; Nusse, R; Roose, JP; Piehler, J; Boj, SF; Janda, CY; Clevers, H; Kuo, CJ; Garcia, KC Next-Generation Surrogate Wnts Support Organoid Growth and Deconvolute Frizzled Pleiotropy In~Vivo. Journal Article In: Cell stem cell, vol. 27, pp. 840-851.e6, 2020, ISSN: 1934-5909. @article{447,
title = {Next-Generation Surrogate Wnts Support Organoid Growth and Deconvolute Frizzled Pleiotropy In~Vivo.},
author = {Y Miao and A Ha and W Lau and K Yuki and AJM Santos and C You and MH Geurts and J Puschhof and C Pleguezuelos-Manzano and WC Peng and R Senlice and C Piani and JW Buikema and OM Gbenedio and M Vallon and J Yuan and S Haan and W Hemrika and K Rösch and LT Dang and D Baker and M Ott and P Depeille and SM Wu and J Drost and R Nusse and JP Roose and J Piehler and SF Boj and CY Janda and H Clevers and CJ Kuo and KC Garcia},
url = {https://linkinghub.elsevier.com/retrieve/pii/S1934-5909(20)30358-1},
doi = {10.1016/j.stem.2020.07.020},
issn = {1934-5909},
year = {2020},
date = {2020-11-01},
journal = {Cell stem cell},
volume = {27},
pages = {840-851.e6},
abstract = {Modulation of Wnt signaling has untapped potential in regenerative medicine due to its essential functions in stem cell homeostasis. However, Wnt lipidation and Wnt-Frizzled (Fzd) cross-reactivity have hindered translational Wnt applications. Here, we designed and engineered water-soluble, Fzd subtype-specific "next-generation surrogate" (NGS) Wnts that hetero-dimerize Fzd and Lrp6. NGS Wnt supports long-term expansion of multiple different types of organoids, including kidney, colon, hepatocyte, ovarian, and breast. NGS Wnts are superior to Wnt3a conditioned media in organoid expansion and single-cell organoid outgrowth. Administration of Fzd subtype-specific NGS Wnt in~vivo reveals that adult intestinal crypt proliferation can be promoted by agonism of Fzd5 and/or Fzd8 receptors, while a broad spectrum of Fzd receptors can induce liver zonation. Thus, NGS Wnts offer a unified organoid expansion protocol and a laboratory "tool kit" for dissecting the functions of Fzd subtypes in stem cell biology.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Modulation of Wnt signaling has untapped potential in regenerative medicine due to its essential functions in stem cell homeostasis. However, Wnt lipidation and Wnt-Frizzled (Fzd) cross-reactivity have hindered translational Wnt applications. Here, we designed and engineered water-soluble, Fzd subtype-specific "next-generation surrogate" (NGS) Wnts that hetero-dimerize Fzd and Lrp6. NGS Wnt supports long-term expansion of multiple different types of organoids, including kidney, colon, hepatocyte, ovarian, and breast. NGS Wnts are superior to Wnt3a conditioned media in organoid expansion and single-cell organoid outgrowth. Administration of Fzd subtype-specific NGS Wnt in~vivo reveals that adult intestinal crypt proliferation can be promoted by agonism of Fzd5 and/or Fzd8 receptors, while a broad spectrum of Fzd receptors can induce liver zonation. Thus, NGS Wnts offer a unified organoid expansion protocol and a laboratory "tool kit" for dissecting the functions of Fzd subtypes in stem cell biology. |
Rim, EY; Kinney, LK; Nusse, R β-catenin-mediated Wnt signal transduction proceeds through an endocytosis-independent mechanism. Journal Article In: Molecular biology of the cell, vol. 31, pp. 1425-1436, 2020, ISSN: 1059-1524. @article{445,
title = {β-catenin-mediated Wnt signal transduction proceeds through an endocytosis-independent mechanism.},
author = {EY Rim and LK Kinney and R Nusse},
url = {https://www.molbiolcell.org/doi/10.1091/mbc.E20-02-0114?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%3dpubmed},
doi = {10.1091/mbc.E20-02-0114},
issn = {1059-1524},
year = {2020},
date = {2020-06-01},
journal = {Molecular biology of the cell},
volume = {31},
pages = {1425-1436},
abstract = {The Wnt pathway is a key intercellular signaling cascade that regulates development, tissue homeostasis, and regeneration. However, gaps remain in our understanding of the molecular events that take place between ligand-receptor binding and target gene transcription. We used a novel tool for quantitative, real-time assessment of endogenous pathway activation, measured in single cells, to answer an unresolved question in the field-whether receptor endocytosis is required for Wnt signal transduction. We combined knockdown or knockout of essential components of clathrin-mediated endocytosis with quantitative assessment of Wnt signal transduction in mouse embryonic stem cells (mESCs). Disruption of clathrin-mediated endocytosis did not affect accumulation and nuclear translocation of β-catenin, as measured by single-cell live imaging of endogenous β-catenin, and subsequent target gene transcription. Disruption of another receptor endocytosis pathway, caveolin-mediated endocytosis, did not affect Wnt pathway activation in mESCs. Additional results in multiple cell lines support that endocytosis is not a requirement for Wnt signal transduction. We show that off-target effects of a drug used to inhibit endocytosis may be one source of the discrepancy among reports on the role of endocytosis in Wnt signaling.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The Wnt pathway is a key intercellular signaling cascade that regulates development, tissue homeostasis, and regeneration. However, gaps remain in our understanding of the molecular events that take place between ligand-receptor binding and target gene transcription. We used a novel tool for quantitative, real-time assessment of endogenous pathway activation, measured in single cells, to answer an unresolved question in the field-whether receptor endocytosis is required for Wnt signal transduction. We combined knockdown or knockout of essential components of clathrin-mediated endocytosis with quantitative assessment of Wnt signal transduction in mouse embryonic stem cells (mESCs). Disruption of clathrin-mediated endocytosis did not affect accumulation and nuclear translocation of β-catenin, as measured by single-cell live imaging of endogenous β-catenin, and subsequent target gene transcription. Disruption of another receptor endocytosis pathway, caveolin-mediated endocytosis, did not affect Wnt pathway activation in mESCs. Additional results in multiple cell lines support that endocytosis is not a requirement for Wnt signal transduction. We show that off-target effects of a drug used to inhibit endocytosis may be one source of the discrepancy among reports on the role of endocytosis in Wnt signaling. |
2019
|
Mirabelli, CK; Nusse, R; Tuveson, DA; Williams, BO Perspectives on the role of Wnt biology in cancer. Journal Article In: Science signaling, vol. 12, 2019, ISSN: 1945-0877. @article{441,
title = {Perspectives on the role of Wnt biology in cancer.},
author = {CK Mirabelli and R Nusse and DA Tuveson and BO Williams},
url = {http://stke.sciencemag.org/cgi/pmidlookup?view=long&pmid=31289213},
doi = {10.1126/scisignal.aay4494},
issn = {1945-0877},
year = {2019},
date = {2019-07-01},
journal = {Science signaling},
volume = {12},
abstract = {Selected members of the Wnt signaling community met during a 4-day period in October 2018 to discuss the current challenges and opportunities associated with targeting the Wnt pathway for therapeutic benefit. A summary of key points of these discussions is presented in this report.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Selected members of the Wnt signaling community met during a 4-day period in October 2018 to discuss the current challenges and opportunities associated with targeting the Wnt pathway for therapeutic benefit. A summary of key points of these discussions is presented in this report. |
Zhao, L; Jin, Y; Donahue, K; Tsui, M; Fish, M; Logan, C Y; Wang, B; Nusse, R Tissue Repair in the Mouse Liver Following Acute Carbon Tetrachloride Depends on Injury-Induced Wnt/β-Catenin Signaling. Journal Article In: Hepatology (Baltimore, Md.), vol. 69, pp. 2623-2635, 2019, ISSN: 0270-9139. @article{440,
title = {Tissue Repair in the Mouse Liver Following Acute Carbon Tetrachloride Depends on Injury-Induced Wnt/β-Catenin Signaling.},
author = {L Zhao and Y Jin and K Donahue and M Tsui and M Fish and C Y Logan and B Wang and R Nusse},
url = {https://doi.org/10.1002/hep.30563},
doi = {10.1002/hep.30563},
issn = {0270-9139},
year = {2019},
date = {2019-06-01},
journal = {Hepatology (Baltimore, Md.)},
volume = {69},
pages = {2623-2635},
abstract = {In the liver, Wnt/β-catenin signaling is involved in regulating zonation and hepatocyte proliferation during homeostasis. We examined Wnt gene expression and signaling after injury, and we show by in situ hybridization that Wnts are activated by acute carbon tetrachloride (CCl ) toxicity. Following injury, peri-injury hepatocytes become Wnt-responsive, expressing the Wnt target gene axis inhibition protein 2 (Axin2). Lineage tracing of peri-injury Axin2 hepatocytes shows that during recovery the injured parenchyma becomes repopulated and repaired by Axin2 descendants. Using single-cell RNA sequencing, we show that endothelial cells are the major source of Wnts following acute CCl toxicity. Induced loss of β-catenin in peri-injury hepatocytes results in delayed repair and ultimately injury-induced lethality, while loss of Wnt production from endothelial cells leads to a delay in the proliferative response after injury. Conclusion: Our findings highlight the importance of the Wnt/β-catenin signaling pathway in restoring tissue integrity following acute liver toxicity and establish a role of endothelial cells as an important Wnt-producing regulator of liver tissue repair following localized liver injury.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
In the liver, Wnt/β-catenin signaling is involved in regulating zonation and hepatocyte proliferation during homeostasis. We examined Wnt gene expression and signaling after injury, and we show by in situ hybridization that Wnts are activated by acute carbon tetrachloride (CCl ) toxicity. Following injury, peri-injury hepatocytes become Wnt-responsive, expressing the Wnt target gene axis inhibition protein 2 (Axin2). Lineage tracing of peri-injury Axin2 hepatocytes shows that during recovery the injured parenchyma becomes repopulated and repaired by Axin2 descendants. Using single-cell RNA sequencing, we show that endothelial cells are the major source of Wnts following acute CCl toxicity. Induced loss of β-catenin in peri-injury hepatocytes results in delayed repair and ultimately injury-induced lethality, while loss of Wnt production from endothelial cells leads to a delay in the proliferative response after injury. Conclusion: Our findings highlight the importance of the Wnt/β-catenin signaling pathway in restoring tissue integrity following acute liver toxicity and establish a role of endothelial cells as an important Wnt-producing regulator of liver tissue repair following localized liver injury. |
Basham, KJ; Rodriguez, S; Turcu, AF; Lerario, AM; Logan, C Y; Rysztak, MR; Gomez-Sanchez, CE; Breault, DT; Koo, BK; Clevers, H; Nusse, R; Val, P; Hammer, GD A ZNRF3-dependent Wnt/β-catenin signaling gradient is required for adrenal homeostasis. Journal Article In: Genes & development, vol. 33, pp. 209-220, 2019, ISSN: 0890-9369. @article{438,
title = {A ZNRF3-dependent Wnt/β-catenin signaling gradient is required for adrenal homeostasis.},
author = {KJ Basham and S Rodriguez and AF Turcu and AM Lerario and C Y Logan and MR Rysztak and CE Gomez-Sanchez and DT Breault and BK Koo and H Clevers and R Nusse and P Val and GD Hammer},
url = {http://www.genesdev.org/cgi/pmidlookup?view=long&pmid=30692207},
doi = {10.1101/gad.317412.118},
issn = {0890-9369},
year = {2019},
date = {2019-02-01},
journal = {Genes & development},
volume = {33},
pages = {209-220},
abstract = {Spatiotemporal control of Wnt signaling is essential for the development and homeostasis of many tissues. The transmembrane E3 ubiquitin ligases ZNRF3 (zinc and ring finger 3) and RNF43 (ring finger protein 43) antagonize Wnt signaling by promoting degradation of frizzled receptors. ZNRF3 and RNF43 are frequently inactivated in human cancer, but the molecular and therapeutic implications remain unclear. Here, we demonstrate that adrenocortical-specific loss of ZNRF3, but not RNF43, results in adrenal hyperplasia that depends on Porcupine-mediated Wnt ligand secretion. Furthermore, we discovered a Wnt/β-catenin signaling gradient in the adrenal cortex that is disrupted upon loss of ZNRF3. Unlike β-catenin gain-of-function models, which induce high Wnt/β-catenin activation and expansion of the peripheral cortex, ZNRF3 loss triggers activation of moderate-level Wnt/β-catenin signaling that drives proliferative expansion of only the histologically and functionally distinct inner cortex. Genetically reducing β-catenin dosage significantly reverses the ZNRF3-deficient phenotype. Thus, homeostatic maintenance of the adrenal cortex is dependent on varying levels of Wnt/β-catenin activation, which is regulated by ZNRF3.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Spatiotemporal control of Wnt signaling is essential for the development and homeostasis of many tissues. The transmembrane E3 ubiquitin ligases ZNRF3 (zinc and ring finger 3) and RNF43 (ring finger protein 43) antagonize Wnt signaling by promoting degradation of frizzled receptors. ZNRF3 and RNF43 are frequently inactivated in human cancer, but the molecular and therapeutic implications remain unclear. Here, we demonstrate that adrenocortical-specific loss of ZNRF3, but not RNF43, results in adrenal hyperplasia that depends on Porcupine-mediated Wnt ligand secretion. Furthermore, we discovered a Wnt/β-catenin signaling gradient in the adrenal cortex that is disrupted upon loss of ZNRF3. Unlike β-catenin gain-of-function models, which induce high Wnt/β-catenin activation and expansion of the peripheral cortex, ZNRF3 loss triggers activation of moderate-level Wnt/β-catenin signaling that drives proliferative expansion of only the histologically and functionally distinct inner cortex. Genetically reducing β-catenin dosage significantly reverses the ZNRF3-deficient phenotype. Thus, homeostatic maintenance of the adrenal cortex is dependent on varying levels of Wnt/β-catenin activation, which is regulated by ZNRF3. |
2018
|
Wan, DC; Morgan, SL; Spencley, AL; Mariano, N; Chang, EY; Shankar, G; Luo, Y; Li, TH; Huh, D; Huynh, SK; Garcia, JM; Dovey, CM; Lumb, J; Liu, L; Brown, KV; Bermudez, A; Luong, R; Zeng, H; Mascetti, VL; Pitteri, SJ; Wang, J; Tu, H; Quarta, M; Sebastiano, V; Nusse, R; Rando, TA; Carette, JE; Bazan, JF; Wang, KC Honey bee Royalactin unlocks conserved pluripotency pathway in mammals. Journal Article In: Nature communications, vol. 9, pp. 5078, 2018. @article{436,
title = {Honey bee Royalactin unlocks conserved pluripotency pathway in mammals.},
author = {DC Wan and SL Morgan and AL Spencley and N Mariano and EY Chang and G Shankar and Y Luo and TH Li and D Huh and SK Huynh and JM Garcia and CM Dovey and J Lumb and L Liu and KV Brown and A Bermudez and R Luong and H Zeng and VL Mascetti and SJ Pitteri and J Wang and H Tu and M Quarta and V Sebastiano and R Nusse and TA Rando and JE Carette and JF Bazan and KC Wang},
url = {http://dx.doi.org/10.1038/s41467-018-06256-4},
doi = {10.1038/s41467-018-06256-4},
year = {2018},
date = {2018-12-01},
journal = {Nature communications},
volume = {9},
pages = {5078},
abstract = {Royal jelly is the queen-maker for the honey bee Apis mellifera, and has cross-species effects on longevity, fertility, and regeneration in mammals. Despite this knowledge, how royal jelly or its components exert their myriad effects has remained poorly understood. Using mouse embryonic stem cells as a platform, here we report that through its major protein component Royalactin, royal jelly can maintain pluripotency by activating a ground-state pluripotency-like gene network. We further identify Regina, a mammalian structural analog of Royalactin that also induces a naive-like state in mouse embryonic stem cells. This reveals an important innate program for stem cell self-renewal with broad implications in understanding the molecular regulation of stem cell fate across species.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Royal jelly is the queen-maker for the honey bee Apis mellifera, and has cross-species effects on longevity, fertility, and regeneration in mammals. Despite this knowledge, how royal jelly or its components exert their myriad effects has remained poorly understood. Using mouse embryonic stem cells as a platform, here we report that through its major protein component Royalactin, royal jelly can maintain pluripotency by activating a ground-state pluripotency-like gene network. We further identify Regina, a mammalian structural analog of Royalactin that also induces a naive-like state in mouse embryonic stem cells. This reveals an important innate program for stem cell self-renewal with broad implications in understanding the molecular regulation of stem cell fate across species. |
Peng, WC; Logan, C Y; Fish, M; Anbarchian, T; Aguisanda, F; Álvarez-Varela, A; Wu, P; Jin, Y; Zhu, J; Li, B; Grompe, M; Wang, B; Nusse, R Inflammatory Cytokine TNFα Promotes the Long-Term Expansion of Primary Hepatocytes in 3D Culture. Journal Article In: Cell, vol. 175, pp. 1607-1619.e15, 2018, ISSN: 0092-8674. @article{434,
title = {Inflammatory Cytokine TNFα Promotes the Long-Term Expansion of Primary Hepatocytes in 3D Culture.},
author = {WC Peng and C Y Logan and M Fish and T Anbarchian and F Aguisanda and A Álvarez-Varela and P Wu and Y Jin and J Zhu and B Li and M Grompe and B Wang and R Nusse},
url = {https://linkinghub.elsevier.com/retrieve/pii/S0092-8674(18)31504-6},
doi = {10.1016/j.cell.2018.11.012},
issn = {0092-8674},
year = {2018},
date = {2018-11-01},
journal = {Cell},
volume = {175},
pages = {1607-1619.e15},
abstract = {In the healthy adult liver, most hepatocytes proliferate minimally. However, upon physical or chemical injury to the liver, hepatocytes proliferate extensively in~vivo under the direction of multiple extracellular cues, including Wnt and pro-inflammatory signals. Currently, liver organoids can be generated readily in~vitro from bile-duct epithelial cells, but not~hepatocytes. Here, we show that TNFα, an injury-induced inflammatory cytokine, promotes the expansion of hepatocytes in 3D culture and enables serial passaging and long-term culture for more than 6~months. Single-cell RNA sequencing reveals broad expression of hepatocyte markers. Strikingly, in~vitro-expanded hepatocytes engrafted, and significantly repopulated, the injured livers of Fah mice. We anticipate that tissue repair signals can be harnessed to promote the expansion of otherwise hard-to-culture cell-types, with broad implications.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
In the healthy adult liver, most hepatocytes proliferate minimally. However, upon physical or chemical injury to the liver, hepatocytes proliferate extensively in~vivo under the direction of multiple extracellular cues, including Wnt and pro-inflammatory signals. Currently, liver organoids can be generated readily in~vitro from bile-duct epithelial cells, but not~hepatocytes. Here, we show that TNFα, an injury-induced inflammatory cytokine, promotes the expansion of hepatocytes in 3D culture and enables serial passaging and long-term culture for more than 6~months. Single-cell RNA sequencing reveals broad expression of hepatocyte markers. Strikingly, in~vitro-expanded hepatocytes engrafted, and significantly repopulated, the injured livers of Fah mice. We anticipate that tissue repair signals can be harnessed to promote the expansion of otherwise hard-to-culture cell-types, with broad implications. |
Wiese, KE; Nusse, R; Amerongen, R Wnt signalling: conquering complexity. Journal Article In: Development (Cambridge, England), vol. 145, 2018, ISSN: 0950-1991. @article{433,
title = {Wnt signalling: conquering complexity.},
author = {KE Wiese and R Nusse and R Amerongen},
url = {http://dev.biologists.org/cgi/pmidlookup?view=long&pmid=29945986},
doi = {10.1242/dev.165902},
issn = {0950-1991},
year = {2018},
date = {2018-06-01},
journal = {Development (Cambridge, England)},
volume = {145},
abstract = {The history of the Wnt pathway is an adventure that takes us from mice and flies to frogs, zebrafish and beyond, sketching the outlines of a molecular signalling cascade along the way. Here, we specifically highlight the instrumental role that developmental biology has played throughout. We take the reader on a journey, starting with developmental genetics studies that identified some of the main molecular players, through developmental model organisms that helped unravel their biochemical function and cell biological activities. Culminating in complex analyses of stem cell fate and dynamic tissue growth, these efforts beautifully illustrate how different disciplines provided missing pieces of a puzzle. Together, they have shaped our mechanistic understanding of the Wnt pathway as a conserved signalling process in development and disease. Today, researchers are still uncovering additional roles for Wnts and other members of this multifaceted signal transduction pathway, opening up promising new avenues for clinical applications.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The history of the Wnt pathway is an adventure that takes us from mice and flies to frogs, zebrafish and beyond, sketching the outlines of a molecular signalling cascade along the way. Here, we specifically highlight the instrumental role that developmental biology has played throughout. We take the reader on a journey, starting with developmental genetics studies that identified some of the main molecular players, through developmental model organisms that helped unravel their biochemical function and cell biological activities. Culminating in complex analyses of stem cell fate and dynamic tissue growth, these efforts beautifully illustrate how different disciplines provided missing pieces of a puzzle. Together, they have shaped our mechanistic understanding of the Wnt pathway as a conserved signalling process in development and disease. Today, researchers are still uncovering additional roles for Wnts and other members of this multifaceted signal transduction pathway, opening up promising new avenues for clinical applications. |
Xing, L; Anbarchian, T; Tsai, JM; Plant, GW; Nusse, R Wnt/β-catenin signaling regulates ependymal cell development and adult homeostasis. Journal Article In: Proceedings of the National Academy of Sciences of the United States of America, vol. 115, pp. E5954-E5962, 2018, ISSN: 0027-8424. @article{431,
title = {Wnt/β-catenin signaling regulates ependymal cell development and adult homeostasis.},
author = {L Xing and T Anbarchian and JM Tsai and GW Plant and R Nusse},
url = {http://www.pnas.org/cgi/pmidlookup?view=long&pmid=29891676},
doi = {10.1073/pnas.1803297115},
issn = {0027-8424},
year = {2018},
date = {2018-06-01},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
volume = {115},
pages = {E5954-E5962},
abstract = {In the adult mouse spinal cord, the ependymal cell population that surrounds the central canal is thought to be a promising source of quiescent stem cells to treat spinal cord injury. Relatively little is known about the cellular origin of ependymal cells during spinal cord development, or the molecular mechanisms that regulate ependymal cells during adult homeostasis. Using genetic lineage tracing based on the Wnt target gene , we have characterized Wnt-responsive cells during spinal cord development. Our results revealed that Wnt-responsive progenitor cells are restricted to the dorsal midline throughout spinal cord development, which gives rise to dorsal ependymal cells in a spatially restricted pattern. This is contrary to previous reports that suggested an exclusively ventral origin of ependymal cells, suggesting that ependymal cells may retain positional identities in relation to their neural progenitors. Our results further demonstrated that in the postnatal and adult spinal cord, all ependymal cells express the Wnt/β-catenin signaling target gene , as well as Wnt ligands. Genetic elimination of β-catenin or inhibition of Wnt secretion in Axin2-expressing ependymal cells in vivo both resulted in impaired proliferation, indicating that Wnt/β-catenin signaling promotes ependymal cell proliferation. These results demonstrate the continued importance of Wnt/β-catenin signaling for both ependymal cell formation and regulation. By uncovering the molecular signals underlying the formation and regulation of spinal cord ependymal cells, our findings thus enable further targeting and manipulation of this promising source of quiescent stem cells for therapeutic interventions.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
In the adult mouse spinal cord, the ependymal cell population that surrounds the central canal is thought to be a promising source of quiescent stem cells to treat spinal cord injury. Relatively little is known about the cellular origin of ependymal cells during spinal cord development, or the molecular mechanisms that regulate ependymal cells during adult homeostasis. Using genetic lineage tracing based on the Wnt target gene , we have characterized Wnt-responsive cells during spinal cord development. Our results revealed that Wnt-responsive progenitor cells are restricted to the dorsal midline throughout spinal cord development, which gives rise to dorsal ependymal cells in a spatially restricted pattern. This is contrary to previous reports that suggested an exclusively ventral origin of ependymal cells, suggesting that ependymal cells may retain positional identities in relation to their neural progenitors. Our results further demonstrated that in the postnatal and adult spinal cord, all ependymal cells express the Wnt/β-catenin signaling target gene , as well as Wnt ligands. Genetic elimination of β-catenin or inhibition of Wnt secretion in Axin2-expressing ependymal cells in vivo both resulted in impaired proliferation, indicating that Wnt/β-catenin signaling promotes ependymal cell proliferation. These results demonstrate the continued importance of Wnt/β-catenin signaling for both ependymal cell formation and regulation. By uncovering the molecular signals underlying the formation and regulation of spinal cord ependymal cells, our findings thus enable further targeting and manipulation of this promising source of quiescent stem cells for therapeutic interventions. |
Przybyl, J; Kidzinski, L; Hastie, T; Debiec-Rychter, M; Nusse, R; Rijn, M Gene expression profiling of low-grade endometrial stromal sarcoma indicates fusion protein-mediated activation of the Wnt signaling pathway. Journal Article In: Gynecologic oncology, vol. 149, pp. 388-393, 2018, ISSN: 0090-8258. @article{429,
title = {Gene expression profiling of low-grade endometrial stromal sarcoma indicates fusion protein-mediated activation of the Wnt signaling pathway.},
author = {J Przybyl and L Kidzinski and T Hastie and M Debiec-Rychter and R Nusse and M Rijn},
url = {https://linkinghub.elsevier.com/retrieve/pii/S0090-8258(18)30177-X},
doi = {10.1016/j.ygyno.2018.03.007},
issn = {0090-8258},
year = {2018},
date = {2018-05-01},
journal = {Gynecologic oncology},
volume = {149},
pages = {388-393},
abstract = {Low-grade endometrial stromal sarcomas (LGESS) harbor chromosomal translocations that affect proteins associated with chromatin remodeling Polycomb Repressive Complex 2 (PRC2), including SUZ12, PHF1 and EPC1. Roughly half of LGESS also demonstrate nuclear accumulation of β-catenin, which is a hallmark of Wnt signaling activation. However, the targets affected by the fusion proteins and the role of Wnt signaling in the pathogenesis of these tumors remain largely unknown.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Low-grade endometrial stromal sarcomas (LGESS) harbor chromosomal translocations that affect proteins associated with chromatin remodeling Polycomb Repressive Complex 2 (PRC2), including SUZ12, PHF1 and EPC1. Roughly half of LGESS also demonstrate nuclear accumulation of β-catenin, which is a hallmark of Wnt signaling activation. However, the targets affected by the fusion proteins and the role of Wnt signaling in the pathogenesis of these tumors remain largely unknown. |
Tan, SH; Nusse, R In vivo lineage tracing reveals Axin2-expressing, long-lived cortical thymic epithelial progenitors in the postnatal thymus. Journal Article In: PloS one, vol. 12, pp. e0184582, 2018. @article{423,
title = {In vivo lineage tracing reveals Axin2-expressing, long-lived cortical thymic epithelial progenitors in the postnatal thymus.},
author = {SH Tan and R Nusse},
url = {http://dx.plos.org/10.1371/journal.pone.0184582},
doi = {10.1371/journal.pone.0184582},
year = {2018},
date = {2018-00-01},
journal = {PloS one},
volume = {12},
pages = {e0184582},
abstract = {In the thymus, cortical and medullary thymic epithelial cells (TECs) are instrumental for generating a repertoire of functional T cells. Hence, there has been much interest in the ontogeny of TECs. While medullary TEC (mTEC) and bipotent progenitors have been identified, the existence of a cortical TEC (cTEC) progenitor remains ambiguous. In this study, we used lineage tracing based on a target gene of the Wnt pathway, Axin2. We found that Axin2 initially labels cells in both the cortical and medullary compartments. Using Axin2-CreERT2 mice to track the fate of labelled cells, we identified long-lived cortical TEC progenitors that give rise to expanding clones and contribute to homeostasis in postnatal thymus. In contrast, no clonal expansion was found in the medullary or in the K5K8-double positive compartments. The identification of cTEC progenitors and their regulation by Wnt signaling have important implications for our understanding of thymus physiology during homeostasis and TEC-related disorders.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
In the thymus, cortical and medullary thymic epithelial cells (TECs) are instrumental for generating a repertoire of functional T cells. Hence, there has been much interest in the ontogeny of TECs. While medullary TEC (mTEC) and bipotent progenitors have been identified, the existence of a cortical TEC (cTEC) progenitor remains ambiguous. In this study, we used lineage tracing based on a target gene of the Wnt pathway, Axin2. We found that Axin2 initially labels cells in both the cortical and medullary compartments. Using Axin2-CreERT2 mice to track the fate of labelled cells, we identified long-lived cortical TEC progenitors that give rise to expanding clones and contribute to homeostasis in postnatal thymus. In contrast, no clonal expansion was found in the medullary or in the K5K8-double positive compartments. The identification of cTEC progenitors and their regulation by Wnt signaling have important implications for our understanding of thymus physiology during homeostasis and TEC-related disorders. |
2017
|
Lippert, A; Janeczek, AA; Fürstenberg, A; Ponjavic, A; Moerner, WE; Nusse, R; Helms, J A; Evans, ND; Lee, SF Single-Molecule Imaging of Wnt3A Protein Diffusion on Living Cell Membranes. Journal Article In: Biophysical journal, vol. 113, pp. 2762-2767, 2017, ISSN: 0006-3495. @article{427,
title = {Single-Molecule Imaging of Wnt3A Protein Diffusion on Living Cell Membranes.},
author = {A Lippert and AA Janeczek and A Fürstenberg and A Ponjavic and WE Moerner and R Nusse and J A Helms and ND Evans and SF Lee},
url = {https://linkinghub.elsevier.com/retrieve/pii/S0006-3495(17)31128-1},
doi = {10.1016/j.bpj.2017.08.060},
issn = {0006-3495},
year = {2017},
date = {2017-12-01},
journal = {Biophysical journal},
volume = {113},
pages = {2762-2767},
abstract = {Wnt proteins are secreted, hydrophobic, lipidated proteins found in all animals that play essential roles in development and disease. Lipid modification is thought to facilitate the interaction of the protein with its receptor, Frizzled, but may also regulate the transport of Wnt protein and its localization at the cell membrane. Here, by employing single-molecule fluorescence techniques, we show that Wnt proteins associate with and diffuse on the plasma membranes of living cells in the absence of any receptor binding. We find that labeled Wnt3A transiently and dynamically associates with the membranes of Drosophila Schneider 2 cells, diffuses with Brownian kinetics on flattened membranes and on cellular protrusions, and does not transfer between cells in close contact. In S2 receptor-plus (S2R+) cells, which express Frizzled receptors, membrane diffusion rate is reduced and membrane residency time is increased. These results provide direct evidence of Wnt3A interaction with living cell membranes, and represent, to our knowledge, a new system for investigating the dynamics of Wnt transport.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Wnt proteins are secreted, hydrophobic, lipidated proteins found in all animals that play essential roles in development and disease. Lipid modification is thought to facilitate the interaction of the protein with its receptor, Frizzled, but may also regulate the transport of Wnt protein and its localization at the cell membrane. Here, by employing single-molecule fluorescence techniques, we show that Wnt proteins associate with and diffuse on the plasma membranes of living cells in the absence of any receptor binding. We find that labeled Wnt3A transiently and dynamically associates with the membranes of Drosophila Schneider 2 cells, diffuses with Brownian kinetics on flattened membranes and on cellular protrusions, and does not transfer between cells in close contact. In S2 receptor-plus (S2R+) cells, which express Frizzled receptors, membrane diffusion rate is reduced and membrane residency time is increased. These results provide direct evidence of Wnt3A interaction with living cell membranes, and represent, to our knowledge, a new system for investigating the dynamics of Wnt transport. |
Brown, K; Loh, KM; Nusse, R Live Imaging Reveals that the First Division of Differentiating Human Embryonic Stem Cells Often Yields Asymmetric Fates. Journal Article In: Cell reports, vol. 21, pp. 301-307, 2017. @article{425,
title = {Live Imaging Reveals that the First Division of Differentiating Human Embryonic Stem Cells Often Yields Asymmetric Fates.},
author = {K Brown and KM Loh and R Nusse},
url = {http://linkinghub.elsevier.com/retrieve/pii/S2211-1247(17)31334-7},
doi = {10.1016/j.celrep.2017.09.044},
year = {2017},
date = {2017-10-01},
journal = {Cell reports},
volume = {21},
pages = {301-307},
abstract = {How do stem cells respond to signals to initiate differentiation? Here, we show that, despite uniform exposure to differentiation-inducing extracellular signals, individual human embryonic stem cells (hESCs) respond heterogeneously. To track how hESCs incipiently exit pluripotency, we established a system to differentiate hESCs as single cells and conducted live imaging to track their very first cell division. We followed the fate of their earliest daughters as they remained undifferentiated or differentiated toward the primitive streak (the earliest descendants of pluripotent cells). About 30%-50% of the time, hESCs divided to yield one primitive streak and one undifferentiated daughter. The undifferentiated daughter cell was innately resistant to WNT signaling and could not respond to this primitive-streak-specifying differentiation signal. Hence, the first division of differentiating hESCs sometimes yields daughters with diverging fates, with implications for the efficiency of directed differentiation protocols and the underlying rules of lineage commitment.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
How do stem cells respond to signals to initiate differentiation? Here, we show that, despite uniform exposure to differentiation-inducing extracellular signals, individual human embryonic stem cells (hESCs) respond heterogeneously. To track how hESCs incipiently exit pluripotency, we established a system to differentiate hESCs as single cells and conducted live imaging to track their very first cell division. We followed the fate of their earliest daughters as they remained undifferentiated or differentiated toward the primitive streak (the earliest descendants of pluripotent cells). About 30%-50% of the time, hESCs divided to yield one primitive streak and one undifferentiated daughter. The undifferentiated daughter cell was innately resistant to WNT signaling and could not respond to this primitive-streak-specifying differentiation signal. Hence, the first division of differentiating hESCs sometimes yields daughters with diverging fates, with implications for the efficiency of directed differentiation protocols and the underlying rules of lineage commitment. |
Sigal, M; Logan, C Y; Kapalczynska, M; Mollenkopf, HJ; Berger, H; Wiedenmann, B; Nusse, R; Amieva, MR; Meyer, TF Stromal R-spondin orchestrates gastric epithelial stem cells and gland homeostasis. Journal Article In: Nature, vol. 548, pp. 451-455, 2017, ISSN: 0028-0836. @article{421,
title = {Stromal R-spondin orchestrates gastric epithelial stem cells and gland homeostasis.},
author = {M Sigal and C Y Logan and M Kapalczynska and HJ Mollenkopf and H Berger and B Wiedenmann and R Nusse and MR Amieva and TF Meyer},
url = {http://dx.doi.org/10.1038/nature23642},
doi = {10.1038/nature23642},
issn = {0028-0836},
year = {2017},
date = {2017-08-01},
journal = {Nature},
volume = {548},
pages = {451-455},
abstract = {The constant regeneration of stomach epithelium is driven by long-lived stem cells, but the mechanism that regulates their turnover is not well understood. We have recently found that the gastric pathogen Helicobacter pylori can activate gastric stem cells and increase epithelial turnover, while Wnt signalling is known to be important for stem cell identity and epithelial regeneration in several tissues. Here we find that antral Wnt signalling, marked by the classic Wnt target gene Axin2, is limited to the base and lower isthmus of gastric glands, where the stem cells reside. Axin2 is expressed by Lgr5+ cells, as well as adjacent, highly proliferative Lgr5- cells that are able to repopulate entire glands, including the base, upon depletion of the Lgr5+ population. Expression of both Axin2 and Lgr5 requires stroma-derived R-spondin 3 produced by gastric myofibroblasts proximal to the stem cell compartment. Exogenous R-spondin administration expands and accelerates proliferation of Axin2+/Lgr5- but not Lgr5+ cells. Consistent with these observations, H. pylori infection increases stromal R-spondin 3 expression and expands the Axin2+ cell pool to cause hyperproliferation and gland hyperplasia. The ability of stromal niche cells to control and adapt epithelial stem cell dynamics constitutes a sophisticated mechanism that orchestrates epithelial regeneration and maintenance of tissue integrity.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The constant regeneration of stomach epithelium is driven by long-lived stem cells, but the mechanism that regulates their turnover is not well understood. We have recently found that the gastric pathogen Helicobacter pylori can activate gastric stem cells and increase epithelial turnover, while Wnt signalling is known to be important for stem cell identity and epithelial regeneration in several tissues. Here we find that antral Wnt signalling, marked by the classic Wnt target gene Axin2, is limited to the base and lower isthmus of gastric glands, where the stem cells reside. Axin2 is expressed by Lgr5+ cells, as well as adjacent, highly proliferative Lgr5- cells that are able to repopulate entire glands, including the base, upon depletion of the Lgr5+ population. Expression of both Axin2 and Lgr5 requires stroma-derived R-spondin 3 produced by gastric myofibroblasts proximal to the stem cell compartment. Exogenous R-spondin administration expands and accelerates proliferation of Axin2+/Lgr5- but not Lgr5+ cells. Consistent with these observations, H. pylori infection increases stromal R-spondin 3 expression and expands the Axin2+ cell pool to cause hyperproliferation and gland hyperplasia. The ability of stromal niche cells to control and adapt epithelial stem cell dynamics constitutes a sophisticated mechanism that orchestrates epithelial regeneration and maintenance of tissue integrity. |
Nusse, R; Clevers, H Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Journal Article In: Cell, vol. 169, pp. 985-999, 2017, ISSN: 0092-8674. @article{419,
title = {Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities.},
author = {R Nusse and H Clevers},
url = {https://linkinghub.elsevier.com/retrieve/pii/S0092-8674(17)30547-0},
doi = {10.1016/j.cell.2017.05.016},
issn = {0092-8674},
year = {2017},
date = {2017-06-01},
journal = {Cell},
volume = {169},
pages = {985-999},
abstract = {The WNT signal transduction cascade is a main regulator of development throughout the animal kingdom. Wnts are also key drivers of most types of tissue stem cells in adult mammals. Unsurprisingly, mutated Wnt pathway components are causative to multiple growth-related pathologies and to cancer. Here, we describe the core Wnt/β-catenin signaling pathway, how it controls stem cells, and contributes to disease. Finally, we discuss strategies for Wnt-based therapies.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The WNT signal transduction cascade is a main regulator of development throughout the animal kingdom. Wnts are also key drivers of most types of tissue stem cells in adult mammals. Unsurprisingly, mutated Wnt pathway components are causative to multiple growth-related pathologies and to cancer. Here, we describe the core Wnt/β-catenin signaling pathway, how it controls stem cells, and contributes to disease. Finally, we discuss strategies for Wnt-based therapies. |
2016
|
Loh, KM; Amerongen, R; Nusse, R Generating Cellular Diversity and Spatial Form: Wnt Signaling and the Evolution of Multicellular Animals. Journal Article In: Developmental cell, vol. 38, pp. 643-55, 2016, ISSN: 1534-5807. @article{417,
title = {Generating Cellular Diversity and Spatial Form: Wnt Signaling and the Evolution of Multicellular Animals.},
author = {KM Loh and R Amerongen and R Nusse},
url = {https://linkinghub.elsevier.com/retrieve/pii/S1534-5807(16)30586-X},
doi = {10.1016/j.devcel.2016.08.011},
issn = {1534-5807},
year = {2016},
date = {2016-09-01},
journal = {Developmental cell},
volume = {38},
pages = {643-55},
abstract = {There were multiple prerequisites to the evolution of multicellular animal life, including the generation of multiple cell fates ("cellular diversity") and their patterned spatial arrangement ("spatial form"). Wnt proteins operate as primordial symmetry-breaking signals. By virtue of their short-range nature and their capacity to~activate both lineage-specifying and cell-polarizing intracellular signaling cascades, Wnts can polarize cells at their site of contact, orienting the axis of cell division while simultaneously programming daughter cells to adopt diverging fates in a spatially stereotyped way. By coupling cell fate to position, symmetry-breaking Wnt signals were pivotal in constructing the metazoan body by generating cellular diversity and spatial form.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
There were multiple prerequisites to the evolution of multicellular animal life, including the generation of multiple cell fates ("cellular diversity") and their patterned spatial arrangement ("spatial form"). Wnt proteins operate as primordial symmetry-breaking signals. By virtue of their short-range nature and their capacity to~activate both lineage-specifying and cell-polarizing intracellular signaling cascades, Wnts can polarize cells at their site of contact, orienting the axis of cell division while simultaneously programming daughter cells to adopt diverging fates in a spatially stereotyped way. By coupling cell fate to position, symmetry-breaking Wnt signals were pivotal in constructing the metazoan body by generating cellular diversity and spatial form. |
Lim, X; Tan, SH; Yu, KL; Lim, SB; Nusse, R Axin2 marks quiescent hair follicle bulge stem cells that are maintained by autocrine Wnt/β-catenin signaling. Journal Article In: Proceedings of the National Academy of Sciences of the United States of America, vol. 113, pp. E1498-505, 2016, ISSN: 0027-8424. @article{411,
title = {Axin2 marks quiescent hair follicle bulge stem cells that are maintained by autocrine Wnt/β-catenin signaling.},
author = {X Lim and SH Tan and KL Yu and SB Lim and R Nusse},
url = {http://www.pnas.org/cgi/pmidlookup?view=long&pmid=26903625},
doi = {10.1073/pnas.1601599113},
issn = {0027-8424},
year = {2016},
date = {2016-03-01},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
volume = {113},
pages = {E1498-505},
abstract = {How stem cells maintain their identity and potency as tissues change during growth is not well understood. In mammalian hair, it is unclear how hair follicle stem cells can enter an extended period of quiescence during the resting phase but retain stem cell potential and be subsequently activated for growth. Here, we use lineage tracing and gene expression mapping to show that the Wnt target gene Axin2 is constantly expressed throughout the hair cycle quiescent phase in outer bulge stem cells that produce their own Wnt signals. Ablating Wnt signaling in the bulge cells causes them to lose their stem cell potency to contribute to hair growth and undergo premature differentiation instead. Bulge cells express secreted Wnt inhibitors, including Dickkopf (Dkk) and secreted frizzled-related protein 1 (Sfrp1). However, the Dickkopf 3 (Dkk3) protein becomes localized to the Wnt-inactive inner bulge that contains differentiated cells. We find that Axin2 expression remains confined to the outer bulge, whereas Dkk3 continues to be localized to the inner bulge during the hair cycle growth phase. Our data suggest that autocrine Wnt signaling in the outer bulge maintains stem cell potency throughout hair cycle quiescence and growth, whereas paracrine Wnt inhibition of inner bulge cells reinforces differentiation.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
How stem cells maintain their identity and potency as tissues change during growth is not well understood. In mammalian hair, it is unclear how hair follicle stem cells can enter an extended period of quiescence during the resting phase but retain stem cell potential and be subsequently activated for growth. Here, we use lineage tracing and gene expression mapping to show that the Wnt target gene Axin2 is constantly expressed throughout the hair cycle quiescent phase in outer bulge stem cells that produce their own Wnt signals. Ablating Wnt signaling in the bulge cells causes them to lose their stem cell potency to contribute to hair growth and undergo premature differentiation instead. Bulge cells express secreted Wnt inhibitors, including Dickkopf (Dkk) and secreted frizzled-related protein 1 (Sfrp1). However, the Dickkopf 3 (Dkk3) protein becomes localized to the Wnt-inactive inner bulge that contains differentiated cells. We find that Axin2 expression remains confined to the outer bulge, whereas Dkk3 continues to be localized to the inner bulge during the hair cycle growth phase. Our data suggest that autocrine Wnt signaling in the outer bulge maintains stem cell potency throughout hair cycle quiescence and growth, whereas paracrine Wnt inhibition of inner bulge cells reinforces differentiation. |
Mizutani, M; Wu, JC; Nusse, R Fibrosis of the Neonatal Mouse Heart After Cryoinjury Is Accompanied by Wnt Signaling Activation and Epicardial-to-Mesenchymal Transition. Journal Article In: Journal of the American Heart Association, vol. 5, pp. e002457, 2016. @article{415,
title = {Fibrosis of the Neonatal Mouse Heart After Cryoinjury Is Accompanied by Wnt Signaling Activation and Epicardial-to-Mesenchymal Transition.},
author = {M Mizutani and JC Wu and R Nusse},
url = {http://jaha.ahajournals.org/cgi/pmidlookup?view=long&pmid=27068625},
doi = {10.1161/JAHA.115.002457},
year = {2016},
date = {2016-03-01},
journal = {Journal of the American Heart Association},
volume = {5},
pages = {e002457},
abstract = {The adult mammalian heart responds to cardiac injury by formation of persistent fibrotic scar that eventually leads to heart failure. In contrast, the neonatal mammalian heart reacts to injury by the development of transient fibrotic tissue that is eventually replaced by regenerated cardiomyocytes. How fibrosis occurs in the neonatal mammalian heart remains unknown. To start elucidating the molecular underpinnings of neonatal cardiac fibrosis, we investigated Wnt signaling in the neonatal heart after cryoinjury.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The adult mammalian heart responds to cardiac injury by formation of persistent fibrotic scar that eventually leads to heart failure. In contrast, the neonatal mammalian heart reacts to injury by the development of transient fibrotic tissue that is eventually replaced by regenerated cardiomyocytes. How fibrosis occurs in the neonatal mammalian heart remains unknown. To start elucidating the molecular underpinnings of neonatal cardiac fibrosis, we investigated Wnt signaling in the neonatal heart after cryoinjury. |
Takase, HM; Nusse, R Paracrine Wnt/β-catenin signaling mediates proliferation of undifferentiated spermatogonia in the adult mouse testis. Journal Article In: Proceedings of the National Academy of Sciences of the United States of America, vol. 113, pp. E1489-97, 2016, ISSN: 0027-8424. @article{413,
title = {Paracrine Wnt/β-catenin signaling mediates proliferation of undifferentiated spermatogonia in the adult mouse testis.},
author = {HM Takase and R Nusse},
url = {http://www.pnas.org/cgi/pmidlookup?view=long&pmid=26929341},
doi = {10.1073/pnas.1601461113},
issn = {0027-8424},
year = {2016},
date = {2016-03-01},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
volume = {113},
pages = {E1489-97},
abstract = {Spermatogonial stem cells (SSCs) fuel the production of male germ cells but the mechanisms behind SSC self-renewal, proliferation, and differentiation are still poorly understood. Using the Wnt target gene Axin2 and genetic lineage-tracing experiments, we found that undifferentiated spermatogonia, comprising SSCs and transit amplifying progenitor cells, respond to Wnt/β-catenin signals. Genetic elimination of β-catenin indicates that Wnt/β-catenin signaling promotes the proliferation of these cells. Signaling is likely initiated by Wnt6, which is uniquely expressed by neighboring Sertoli cells, the only somatic cells in the seminiferous tubule that support germ cells and act as a niche for SSCs. Therefore, unlike other stem cell systems where Wnt/β-catenin signaling is implicated in self-renewal, the Wnt pathway in the testis specifically contributes to the proliferation of SSCs and progenitor cells.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Spermatogonial stem cells (SSCs) fuel the production of male germ cells but the mechanisms behind SSC self-renewal, proliferation, and differentiation are still poorly understood. Using the Wnt target gene Axin2 and genetic lineage-tracing experiments, we found that undifferentiated spermatogonia, comprising SSCs and transit amplifying progenitor cells, respond to Wnt/β-catenin signals. Genetic elimination of β-catenin indicates that Wnt/β-catenin signaling promotes the proliferation of these cells. Signaling is likely initiated by Wnt6, which is uniquely expressed by neighboring Sertoli cells, the only somatic cells in the seminiferous tubule that support germ cells and act as a niche for SSCs. Therefore, unlike other stem cell systems where Wnt/β-catenin signaling is implicated in self-renewal, the Wnt pathway in the testis specifically contributes to the proliferation of SSCs and progenitor cells. |
2015
|
Lee, SH; Johnson, DT; Luong, R; Yu, EJ; Cunha, GR; Nusse, R; Sun, Z Wnt/β-Catenin-Responsive Cells in Prostatic Development and Regeneration. Journal Article In: Stem cells (Dayton, Ohio), vol. 33, pp. 3356-67, 2015, ISSN: 1066-5099. @article{407,
title = {Wnt/β-Catenin-Responsive Cells in Prostatic Development and Regeneration.},
author = {SH Lee and DT Johnson and R Luong and EJ Yu and GR Cunha and R Nusse and Z Sun},
url = {http://dx.doi.org/10.1002/stem.2096},
doi = {10.1002/stem.2096},
issn = {1066-5099},
year = {2015},
date = {2015-11-01},
journal = {Stem cells (Dayton, Ohio)},
volume = {33},
pages = {3356-67},
abstract = {The precise role of Wnt/β-catenin signaling during prostatic development and tumorigenesis is unclear. Axin2 is a direct transcriptional target of β-catenin. Recent studies have shown that Axin2-expressing cells have stem/progenitor cell properties in a variety of mouse tissues. Here, we genetically labeled Axin2-expressing cells at various time points and tracked their cellular behavior at different developmental and mature stages. We found that prostatic Axin2-expressing cells mainly express luminal epithelial cell markers and are able to expand luminal cell lineages during prostatic development and maturation. They can also survive androgen withdrawal and regenerate prostatic luminal epithelial cells following androgen replacement. Deletion of β-catenin or expression of stabilized β-catenin in these Axin2-expressing cells results in abnormal development or oncogenic transformation, respectively. Our study uncovers a critical role of Wnt/β-catenin-responsive cells in prostatic development and regeneration, and that dysregulation of Wnt/β-catenin signaling in these cells contributes to prostatic developmental defects and tumorigenesis. Stem Cells 2015;33:3356-3367.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The precise role of Wnt/β-catenin signaling during prostatic development and tumorigenesis is unclear. Axin2 is a direct transcriptional target of β-catenin. Recent studies have shown that Axin2-expressing cells have stem/progenitor cell properties in a variety of mouse tissues. Here, we genetically labeled Axin2-expressing cells at various time points and tracked their cellular behavior at different developmental and mature stages. We found that prostatic Axin2-expressing cells mainly express luminal epithelial cell markers and are able to expand luminal cell lineages during prostatic development and maturation. They can also survive androgen withdrawal and regenerate prostatic luminal epithelial cells following androgen replacement. Deletion of β-catenin or expression of stabilized β-catenin in these Axin2-expressing cells results in abnormal development or oncogenic transformation, respectively. Our study uncovers a critical role of Wnt/β-catenin-responsive cells in prostatic development and regeneration, and that dysregulation of Wnt/β-catenin signaling in these cells contributes to prostatic developmental defects and tumorigenesis. Stem Cells 2015;33:3356-3367. |
Wang, B; Zhao, L; Fish, M; Logan, C Y; Nusse, R Self-renewing diploid Axin2(+) cells fuel homeostatic renewal of the liver. Journal Article In: Nature, vol. 524, pp. 180-5, 2015, ISSN: 0028-0836. @article{409,
title = {Self-renewing diploid Axin2(+) cells fuel homeostatic renewal of the liver.},
author = {B Wang and L Zhao and M Fish and C Y Logan and R Nusse},
url = {http://dx.doi.org/10.1038/nature14863},
doi = {10.1038/nature14863},
issn = {0028-0836},
year = {2015},
date = {2015-08-01},
journal = {Nature},
volume = {524},
pages = {180-5},
abstract = {The source of new hepatocytes in the uninjured liver has remained an open question. By lineage tracing using the Wnt-responsive gene Axin2 in mice, we identify a population of proliferating and self-renewing cells adjacent to the central vein in the liver lobule. These pericentral cells express the early liver progenitor marker Tbx3, are diploid, and thereby differ from mature hepatocytes, which are mostly polyploid. The descendants of pericentral cells differentiate into Tbx3-negative, polyploid hepatocytes, and can replace all hepatocytes along the liver lobule during homeostatic renewal. Adjacent central vein endothelial cells provide Wnt signals that maintain the pericentral cells, thereby constituting the niche. Thus, we identify a cell population in the liver that subserves homeostatic hepatocyte renewal, characterize its anatomical niche, and identify molecular signals that regulate its activity.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The source of new hepatocytes in the uninjured liver has remained an open question. By lineage tracing using the Wnt-responsive gene Axin2 in mice, we identify a population of proliferating and self-renewing cells adjacent to the central vein in the liver lobule. These pericentral cells express the early liver progenitor marker Tbx3, are diploid, and thereby differ from mature hepatocytes, which are mostly polyploid. The descendants of pericentral cells differentiate into Tbx3-negative, polyploid hepatocytes, and can replace all hepatocytes along the liver lobule during homeostatic renewal. Adjacent central vein endothelial cells provide Wnt signals that maintain the pericentral cells, thereby constituting the niche. Thus, we identify a cell population in the liver that subserves homeostatic hepatocyte renewal, characterize its anatomical niche, and identify molecular signals that regulate its activity. |
Guenther, CA; Wang, Z; Li, E; Tran, MC; Logan, C Y; Nusse, R; Pantalena-Filho, L; Yang, GP; Kingsley, DM A distinct regulatory region of the Bmp5 locus activates gene expression following adult bone fracture or soft tissue injury. Journal Article In: Bone, vol. 77, pp. 31-41, 2015, ISSN: 8756-3282. @article{405,
title = {A distinct regulatory region of the Bmp5 locus activates gene expression following adult bone fracture or soft tissue injury.},
author = {CA Guenther and Z Wang and E Li and MC Tran and C Y Logan and R Nusse and L Pantalena-Filho and GP Yang and DM Kingsley},
url = {http://linkinghub.elsevier.com/retrieve/pii/S8756-3282(15)00123-4},
doi = {10.1016/j.bone.2015.04.010},
issn = {8756-3282},
year = {2015},
date = {2015-08-01},
journal = {Bone},
volume = {77},
pages = {31-41},
abstract = {Bone morphogenetic proteins (BMPs) are key signaling molecules required for normal development of bones and other tissues. Previous studies have shown that null mutations in the mouse Bmp5 gene alter the size, shape and number of multiple bone and cartilage structures during development. Bmp5 mutations also delay healing of rib fractures in adult mutants, suggesting that the same signals used to pattern embryonic bone and cartilage are also reused during skeletal regeneration and repair. Despite intense interest in BMPs as agents for stimulating bone formation in clinical applications, little is known about the regulatory elements that control developmental or injury-induced BMP expression. To compare the DNA sequences that activate gene expression during embryonic bone formation and following acute injuries in adult animals, we assayed regions surrounding the Bmp5 gene for their ability to stimulate lacZ reporter gene expression in transgenic mice. Multiple genomic fragments, distributed across the Bmp5 locus, collectively coordinate expression in discrete anatomic domains during normal development, including in embryonic ribs. In contrast, a distinct regulatory region activated expression following rib fracture in adult animals. The same injury control region triggered gene expression in mesenchymal cells following tibia fracture, in migrating keratinocytes following dorsal skin wounding, and in regenerating epithelial cells following lung injury. The Bmp5 gene thus contains an "injury response" control region that is distinct from embryonic enhancers, and that is activated by multiple types of injury in adult animals.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Bone morphogenetic proteins (BMPs) are key signaling molecules required for normal development of bones and other tissues. Previous studies have shown that null mutations in the mouse Bmp5 gene alter the size, shape and number of multiple bone and cartilage structures during development. Bmp5 mutations also delay healing of rib fractures in adult mutants, suggesting that the same signals used to pattern embryonic bone and cartilage are also reused during skeletal regeneration and repair. Despite intense interest in BMPs as agents for stimulating bone formation in clinical applications, little is known about the regulatory elements that control developmental or injury-induced BMP expression. To compare the DNA sequences that activate gene expression during embryonic bone formation and following acute injuries in adult animals, we assayed regions surrounding the Bmp5 gene for their ability to stimulate lacZ reporter gene expression in transgenic mice. Multiple genomic fragments, distributed across the Bmp5 locus, collectively coordinate expression in discrete anatomic domains during normal development, including in embryonic ribs. In contrast, a distinct regulatory region activated expression following rib fracture in adult animals. The same injury control region triggered gene expression in mesenchymal cells following tibia fracture, in migrating keratinocytes following dorsal skin wounding, and in regenerating epithelial cells following lung injury. The Bmp5 gene thus contains an "injury response" control region that is distinct from embryonic enhancers, and that is activated by multiple types of injury in adult animals. |
Sigal, M; Rothenberg, ME; Logan, C Y; Lee, JY; Honaker, RW; Cooper, RL; Passarelli, B; Camorlinga, M; Bouley, DM; Alvarez, G; Nusse, R; Torres, J; Amieva, MR Helicobacter pylori Activates and Expands Lgr5(+) Stem Cells Through Direct Colonization of the Gastric Glands. Journal Article In: Gastroenterology, vol. 148, pp. 1392-1404.e21, 2015, ISSN: 0016-5085. @article{401,
title = {Helicobacter pylori Activates and Expands Lgr5(+) Stem Cells Through Direct Colonization of the Gastric Glands.},
author = {M Sigal and ME Rothenberg and C Y Logan and JY Lee and RW Honaker and RL Cooper and B Passarelli and M Camorlinga and DM Bouley and G Alvarez and R Nusse and J Torres and MR Amieva},
url = {http://linkinghub.elsevier.com/retrieve/pii/S0016-5085(15)00271-1},
doi = {10.1053/j.gastro.2015.02.049},
issn = {0016-5085},
year = {2015},
date = {2015-06-01},
journal = {Gastroenterology},
volume = {148},
pages = {1392-1404.e21},
abstract = {Helicobacter pylori infection is the main risk factor for gastric cancer. We characterized the interactions of H pylori with gastric epithelial progenitor and stem cells in humans and mice and investigated how these interactions contribute to H pylori-induced pathology.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Helicobacter pylori infection is the main risk factor for gastric cancer. We characterized the interactions of H pylori with gastric epithelial progenitor and stem cells in humans and mice and investigated how these interactions contribute to H pylori-induced pathology. |
Nusse, R Cell signalling: Disarming Wnt. Journal Article In: Nature, vol. 519, pp. 163-4, 2015, ISSN: 0028-0836. @article{403,
title = {Cell signalling: Disarming Wnt.},
author = {R Nusse},
url = {http://dx.doi.org/10.1038/nature14208},
doi = {10.1038/nature14208},
issn = {0028-0836},
year = {2015},
date = {2015-03-01},
journal = {Nature},
volume = {519},
pages = {163-4},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
|
2014
|
Tan, SH; Senarath-Yapa, K; Chung, MT; Longaker, MT; Wu, JY; Nusse, R Wnts produced by Osterix-expressing osteolineage cells regulate their proliferation and differentiation. Journal Article In: Proceedings of the National Academy of Sciences of the United States of America, vol. 111, pp. E5262-71, 2014, ISSN: 0027-8424. @article{399,
title = {Wnts produced by Osterix-expressing osteolineage cells regulate their proliferation and differentiation.},
author = {SH Tan and K Senarath-Yapa and MT Chung and MT Longaker and JY Wu and R Nusse},
url = {http://www.pnas.org/cgi/pmidlookup?view=long&pmid=25422448},
doi = {10.1073/pnas.1420463111},
issn = {0027-8424},
year = {2014},
date = {2014-12-01},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
volume = {111},
pages = {E5262-71},
abstract = {Wnt signaling is a critical regulator of bone development, but the identity and role of the Wnt-producing cells are still unclear. We addressed these questions through in situ hybridization, lineage tracing, and genetic experiments. First, we surveyed the expression of all 19 Wnt genes and Wnt target gene Axin2 in the neonatal mouse bone by in situ hybridization, and demonstrated--to our knowledge for the first time--that Osterix-expressing cells coexpress Wnt and Axin2. To track the behavior and cell fate of Axin2-expressing osteolineage cells, we performed lineage tracing and showed that they sustain bone formation over the long term. Finally, to examine the role of Wnts produced by Osterix-expressing cells, we inhibited Wnt secretion in vivo, and observed inappropriate differentiation, impaired proliferation, and diminished Wnt signaling response. Therefore, Osterix-expressing cells produce their own Wnts that in turn induce Wnt signaling response, thereby regulating their proliferation and differentiation.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Wnt signaling is a critical regulator of bone development, but the identity and role of the Wnt-producing cells are still unclear. We addressed these questions through in situ hybridization, lineage tracing, and genetic experiments. First, we surveyed the expression of all 19 Wnt genes and Wnt target gene Axin2 in the neonatal mouse bone by in situ hybridization, and demonstrated--to our knowledge for the first time--that Osterix-expressing cells coexpress Wnt and Axin2. To track the behavior and cell fate of Axin2-expressing osteolineage cells, we performed lineage tracing and showed that they sustain bone formation over the long term. Finally, to examine the role of Wnts produced by Osterix-expressing cells, we inhibited Wnt secretion in vivo, and observed inappropriate differentiation, impaired proliferation, and diminished Wnt signaling response. Therefore, Osterix-expressing cells produce their own Wnts that in turn induce Wnt signaling response, thereby regulating their proliferation and differentiation. |
Clevers, H; Loh, KM; Nusse, R Stem cell signaling. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control. Journal Article In: Science (New York, N.Y.), vol. 346, pp. 1248012, 2014, ISSN: 0036-8075. @article{397,
title = {Stem cell signaling. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control.},
author = {H Clevers and KM Loh and R Nusse},
url = {http://www.sciencemag.org/cgi/pmidlookup?view=long&pmid=25278615},
doi = {10.1126/science.1248012},
issn = {0036-8075},
year = {2014},
date = {2014-10-01},
journal = {Science (New York, N.Y.)},
volume = {346},
pages = {1248012},
abstract = {Stem cells fuel tissue development, renewal, and regeneration, and these activities are controlled by the local stem cell microenvironment, the "niche." Wnt signals emanating from the niche can act as self-renewal factors for stem cells in multiple mammalian tissues. Wnt proteins are lipid-modified, which constrains them to act as short-range cellular signals. The locality of Wnt signaling dictates that stem cells exiting the Wnt signaling domain differentiate, spatially delimiting the niche in certain tissues. In some instances, stem cells may act as or generate their own niche, enabling the self-organization of patterned tissues. In this Review, we discuss the various ways by which Wnt operates in stem cell control and, in doing so, identify an integral program for tissue renewal and regeneration.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Stem cells fuel tissue development, renewal, and regeneration, and these activities are controlled by the local stem cell microenvironment, the "niche." Wnt signals emanating from the niche can act as self-renewal factors for stem cells in multiple mammalian tissues. Wnt proteins are lipid-modified, which constrains them to act as short-range cellular signals. The locality of Wnt signaling dictates that stem cells exiting the Wnt signaling domain differentiate, spatially delimiting the niche in certain tissues. In some instances, stem cells may act as or generate their own niche, enabling the self-organization of patterned tissues. In this Review, we discuss the various ways by which Wnt operates in stem cell control and, in doing so, identify an integral program for tissue renewal and regeneration. |
Rinkevich, Y; Montoro, DT; Contreras-Trujillo, H; Harari-Steinberg, O; Newman, AM; Tsai, JM; Lim, X; Van-Amerongen, R; Bowman, A; Januszyk, M; Pleniceanu, O; Nusse, R; Longaker, MT; Weissman, IL; Dekel, B In vivo clonal analysis reveals lineage-restricted progenitor characteristics in mammalian kidney development, maintenance, and regeneration. Journal Article In: Cell reports, vol. 7, pp. 1270-83, 2014. @article{395,
title = {In vivo clonal analysis reveals lineage-restricted progenitor characteristics in mammalian kidney development, maintenance, and regeneration.},
author = {Y Rinkevich and DT Montoro and H Contreras-Trujillo and O Harari-Steinberg and AM Newman and JM Tsai and X Lim and R Van-Amerongen and A Bowman and M Januszyk and O Pleniceanu and R Nusse and MT Longaker and IL Weissman and B Dekel},
url = {http://linkinghub.elsevier.com/retrieve/pii/S2211-1247(14)00305-2},
doi = {10.1016/j.celrep.2014.04.018},
year = {2014},
date = {2014-05-01},
journal = {Cell reports},
volume = {7},
pages = {1270-83},
abstract = {The mechanism and magnitude by which the mammalian kidney generates and maintains its proximal tubules, distal tubules, and collecting ducts remain controversial. Here, we use long-term in vivo genetic lineage tracing and clonal analysis of individual cells from kidneys undergoing development, maintenance, and regeneration. We show that the adult mammalian kidney undergoes continuous tubulogenesis via expansions of fate-restricted clones. Kidneys recovering from damage undergo tubulogenesis through expansions of clones with segment-specific borders, and renal spheres developing in vitro from individual cells maintain distinct, segment-specific fates. Analysis of mice derived by transfer of color-marked embryonic stem cells (ESCs) into uncolored blastocysts demonstrates that nephrons are polyclonal, developing from expansions of singly fated clones. Finally, we show that adult renal clones are derived from Wnt-responsive precursors, and their tracing in vivo generates tubules that are segment specific. Collectively, these analyses demonstrate that fate-restricted precursors functioning as unipotent progenitors continuously maintain and self-preserve the mouse kidney throughout life.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The mechanism and magnitude by which the mammalian kidney generates and maintains its proximal tubules, distal tubules, and collecting ducts remain controversial. Here, we use long-term in vivo genetic lineage tracing and clonal analysis of individual cells from kidneys undergoing development, maintenance, and regeneration. We show that the adult mammalian kidney undergoes continuous tubulogenesis via expansions of fate-restricted clones. Kidneys recovering from damage undergo tubulogenesis through expansions of clones with segment-specific borders, and renal spheres developing in vitro from individual cells maintain distinct, segment-specific fates. Analysis of mice derived by transfer of color-marked embryonic stem cells (ESCs) into uncolored blastocysts demonstrates that nephrons are polyclonal, developing from expansions of singly fated clones. Finally, we show that adult renal clones are derived from Wnt-responsive precursors, and their tracing in vivo generates tubules that are segment specific. Collectively, these analyses demonstrate that fate-restricted precursors functioning as unipotent progenitors continuously maintain and self-preserve the mouse kidney throughout life. |
Green, J; Nusse, R; Amerongen, R The role of Ryk and Ror receptor tyrosine kinases in Wnt signal transduction. Journal Article In: Cold Spring Harbor perspectives in biology, vol. 6, 2014. @article{393,
title = {The role of Ryk and Ror receptor tyrosine kinases in Wnt signal transduction.},
author = {J Green and R Nusse and R Amerongen},
url = {http://cshperspectives.cshlp.org/cgi/pmidlookup?view=long&pmid=24370848},
doi = {10.1101/cshperspect.a009175},
year = {2014},
date = {2014-02-01},
journal = {Cold Spring Harbor perspectives in biology},
volume = {6},
abstract = {Receptor tyrosine kinases of the Ryk and Ror families were initially classified as orphan receptors because their ligands were unknown. They are now known to contain functional extracellular Wnt-binding domains and are implicated in Wnt-signal transduction in multiple species. Although their signaling mechanisms still remain to be resolved in detail, both Ryk and Ror control important developmental processes in different tissues. However, whereas many other Wnt-signaling responses affect cell proliferation and differentiation, Ryk and Ror are mostly associated with controlling processes that rely on the polarized migration of cells. Here we discuss what is currently known about the involvement of this exciting class of receptors in development and disease.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Receptor tyrosine kinases of the Ryk and Ror families were initially classified as orphan receptors because their ligands were unknown. They are now known to contain functional extracellular Wnt-binding domains and are implicated in Wnt-signal transduction in multiple species. Although their signaling mechanisms still remain to be resolved in detail, both Ryk and Ror control important developmental processes in different tissues. However, whereas many other Wnt-signaling responses affect cell proliferation and differentiation, Ryk and Ror are mostly associated with controlling processes that rely on the polarized migration of cells. Here we discuss what is currently known about the involvement of this exciting class of receptors in development and disease. |
2013
|
Lim, X; Tan, SH; Koh, WL; Chau, RM; Yan, KS; Kuo, CJ; Amerongen, R; Klein, AM; Nusse, R Interfollicular epidermal stem cells self-renew via autocrine Wnt signaling. Journal Article In: Science (New York, N.Y.), vol. 342, pp. 1226-30, 2013, ISSN: 0036-8075. @article{391,
title = {Interfollicular epidermal stem cells self-renew via autocrine Wnt signaling.},
author = {X Lim and SH Tan and WL Koh and RM Chau and KS Yan and CJ Kuo and R Amerongen and AM Klein and R Nusse},
url = {http://www.sciencemag.org/cgi/pmidlookup?view=long&pmid=24311688},
doi = {10.1126/science.1239730},
issn = {0036-8075},
year = {2013},
date = {2013-12-01},
journal = {Science (New York, N.Y.)},
volume = {342},
pages = {1226-30},
abstract = {The skin is a classical example of a tissue maintained by stem cells. However, the identity of the stem cells that maintain the interfollicular epidermis and the source of the signals that control their activity remain unclear. Using mouse lineage tracing and quantitative clonal analyses, we showed that the Wnt target gene Axin2 marks interfollicular epidermal stem cells. These Axin2-expressing cells constitute the majority of the basal epidermal layer, compete neutrally, and require Wnt/β-catenin signaling to proliferate. The same cells contribute robustly to wound healing, with no requirement for a quiescent stem cell subpopulation. By means of double-labeling RNA in situ hybridization in mice, we showed that the Axin2-expressing cells themselves produce Wnt signals as well as long-range secreted Wnt inhibitors, suggesting an autocrine mechanism of stem cell self-renewal.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The skin is a classical example of a tissue maintained by stem cells. However, the identity of the stem cells that maintain the interfollicular epidermis and the source of the signals that control their activity remain unclear. Using mouse lineage tracing and quantitative clonal analyses, we showed that the Wnt target gene Axin2 marks interfollicular epidermal stem cells. These Axin2-expressing cells constitute the majority of the basal epidermal layer, compete neutrally, and require Wnt/β-catenin signaling to proliferate. The same cells contribute robustly to wound healing, with no requirement for a quiescent stem cell subpopulation. By means of double-labeling RNA in situ hybridization in mice, we showed that the Axin2-expressing cells themselves produce Wnt signals as well as long-range secreted Wnt inhibitors, suggesting an autocrine mechanism of stem cell self-renewal. |
Sugiyama, T; Benitez, CM; Ghodasara, A; Liu, L; McLean, GW; Lee, J; Blauwkamp, TA; Nusse, R; Wright, CV; Gu, G; Kim, SK Reconstituting pancreas development from purified progenitor cells reveals genes essential for islet differentiation. Journal Article In: Proceedings of the National Academy of Sciences of the United States of America, vol. 110, pp. 12691-6, 2013, ISSN: 0027-8424. @article{389,
title = {Reconstituting pancreas development from purified progenitor cells reveals genes essential for islet differentiation.},
author = {T Sugiyama and CM Benitez and A Ghodasara and L Liu and GW McLean and J Lee and TA Blauwkamp and R Nusse and CV Wright and G Gu and SK Kim},
url = {http://www.pnas.org/cgi/pmidlookup?view=long&pmid=23852729},
doi = {10.1073/pnas.1304507110},
issn = {0027-8424},
year = {2013},
date = {2013-07-01},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
volume = {110},
pages = {12691-6},
abstract = {Developmental biology is challenged to reveal the function of numerous candidate genes implicated by recent genome-scale studies as regulators of organ development and diseases. Recapitulating organogenesis from purified progenitor cells that can be genetically manipulated would provide powerful opportunities to dissect such gene functions. Here we describe systems for reconstructing pancreas development, including islet β-cell and α-cell differentiation, from single fetal progenitor cells. A strict requirement for native genetic regulators of in vivo pancreas development, such as Ngn3, Arx, and Pax4, revealed the authenticity of differentiation programs in vitro. Efficient genetic screens permitted by this system revealed that Prdm16 is required for pancreatic islet development in vivo. Discovering the function of genes regulating pancreas development with our system should enrich strategies for regenerating islets for treating diabetes mellitus.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Developmental biology is challenged to reveal the function of numerous candidate genes implicated by recent genome-scale studies as regulators of organ development and diseases. Recapitulating organogenesis from purified progenitor cells that can be genetically manipulated would provide powerful opportunities to dissect such gene functions. Here we describe systems for reconstructing pancreas development, including islet β-cell and α-cell differentiation, from single fetal progenitor cells. A strict requirement for native genetic regulators of in vivo pancreas development, such as Ngn3, Arx, and Pax4, revealed the authenticity of differentiation programs in vitro. Efficient genetic screens permitted by this system revealed that Prdm16 is required for pancreatic islet development in vivo. Discovering the function of genes regulating pancreas development with our system should enrich strategies for regenerating islets for treating diabetes mellitus. |
Chu, ML; Ahn, VE; Choi, HJ; Daniels, DL; Nusse, R; Weis, WI structural Studies of Wnts and identification of an LRP6 binding site. Journal Article In: Structure (London, England : 1993), vol. 21, pp. 1235-42, 2013, ISSN: 0969-2126. @article{387,
title = {structural Studies of Wnts and identification of an LRP6 binding site.},
author = {ML Chu and VE Ahn and HJ Choi and DL Daniels and R Nusse and WI Weis},
url = {http://linkinghub.elsevier.com/retrieve/pii/S0969-2126(13)00164-0},
doi = {10.1016/j.str.2013.05.006},
issn = {0969-2126},
year = {2013},
date = {2013-07-01},
journal = {Structure (London, England : 1993)},
volume = {21},
pages = {1235-42},
abstract = {Wnts are secreted growth factors that have critical roles in cell fate determination and stem cell renewal. The Wnt/β-catenin pathway is initiated by binding of a Wnt protein to a Frizzled (Fzd) receptor and a coreceptor, LDL receptor-related protein 5 or 6 (LRP5/6). We report the 2.1~r A resolution crystal structure of a Drosophila WntD fragment encompassing the N-terminal domain and the linker that connects it to the C-terminal domain. Differences in the structures of WntD and Xenopus Wnt8, including the positions of a receptor-binding β hairpin and a large solvent-filled cavity in the helical core, indicate conformational plasticity in the N-terminal domain that may be important for Wnt-Frizzled specificity. Structure-based mutational analysis of mouse Wnt3a shows that the linker between the N- and C-terminal domains is required for LRP6 binding. These findings provide important insights into Wnt function and evolution.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Wnts are secreted growth factors that have critical roles in cell fate determination and stem cell renewal. The Wnt/β-catenin pathway is initiated by binding of a Wnt protein to a Frizzled (Fzd) receptor and a coreceptor, LDL receptor-related protein 5 or 6 (LRP5/6). We report the 2.1~r A resolution crystal structure of a Drosophila WntD fragment encompassing the N-terminal domain and the linker that connects it to the C-terminal domain. Differences in the structures of WntD and Xenopus Wnt8, including the positions of a receptor-binding β hairpin and a large solvent-filled cavity in the helical core, indicate conformational plasticity in the N-terminal domain that may be important for Wnt-Frizzled specificity. Structure-based mutational analysis of mouse Wnt3a shows that the linker between the N- and C-terminal domains is required for LRP6 binding. These findings provide important insights into Wnt function and evolution. |
Bowman, AN; Amerongen, R; Palmer, TD; Nusse, R Lineage tracing with Axin2 reveals distinct developmental and adult populations of Wnt/β-catenin-responsive neural stem cells. Journal Article In: Proceedings of the National Academy of Sciences of the United States of America, vol. 110, pp. 7324-9, 2013, ISSN: 0027-8424. @article{381,
title = {Lineage tracing with Axin2 reveals distinct developmental and adult populations of Wnt/β-catenin-responsive neural stem cells.},
author = {AN Bowman and R Amerongen and TD Palmer and R Nusse},
url = {http://www.pnas.org/cgi/pmidlookup?view=long&pmid=23589866},
doi = {10.1073/pnas.1305411110},
issn = {0027-8424},
year = {2013},
date = {2013-04-01},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
volume = {110},
pages = {7324-9},
abstract = {Since the discovery of neural stem cells in the mammalian brain, there has been significant interest in understanding their contribution to tissue homeostasis at both the cellular and molecular level. Wnt/β-catenin signaling is crucial for development of the central nervous system and has been implicated in stem cell maintenance in multiple tissues. Based on this, we hypothesized that the Wnt pathway likely controls neural stem cell maintenance and differentiation along the entire developmental continuum. To test this, we performed lineage tracing experiments using the recently developed tamoxifen-inducible Cre at Axin2 mouse strain to follow the developmental fate of Wnt/β-catenin-responsive cells in both the embryonic and postnatal mouse brain. From as early as embryonic day 8.5 onwards, Axin2(+) cells can give rise to spatially and functionally restricted populations of adult neural stem cells in the subventricular zone. Similarly, progeny from Axin2(+) cells labeled from E12.5 contribute to both the subventricular zone and the dentate gyrus of the hippocampus. Labeling in the postnatal brain, in turn, demonstrates the persistence of long-lived, Wnt/β-catenin-responsive stem cells in both of these sites. These results demonstrate the continued importance of Wnt/β-catenin signaling for neural stem and progenitor cell formation and function throughout developmental time.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Since the discovery of neural stem cells in the mammalian brain, there has been significant interest in understanding their contribution to tissue homeostasis at both the cellular and molecular level. Wnt/β-catenin signaling is crucial for development of the central nervous system and has been implicated in stem cell maintenance in multiple tissues. Based on this, we hypothesized that the Wnt pathway likely controls neural stem cell maintenance and differentiation along the entire developmental continuum. To test this, we performed lineage tracing experiments using the recently developed tamoxifen-inducible Cre at Axin2 mouse strain to follow the developmental fate of Wnt/β-catenin-responsive cells in both the embryonic and postnatal mouse brain. From as early as embryonic day 8.5 onwards, Axin2(+) cells can give rise to spatially and functionally restricted populations of adult neural stem cells in the subventricular zone. Similarly, progeny from Axin2(+) cells labeled from E12.5 contribute to both the subventricular zone and the dentate gyrus of the hippocampus. Labeling in the postnatal brain, in turn, demonstrates the persistence of long-lived, Wnt/β-catenin-responsive stem cells in both of these sites. These results demonstrate the continued importance of Wnt/β-catenin signaling for neural stem and progenitor cell formation and function throughout developmental time. |
Green, JL; La, J; Yum, KW; Desai, P; Rodewald, LW; Zhang, X; Leblanc, M; Nusse, R; Lewis, MT; Wahl, GM Paracrine Wnt signaling both promotes and inhibits human breast tumor growth. Journal Article In: Proceedings of the National Academy of Sciences of the United States of America, vol. 110, pp. 6991-6, 2013, ISSN: 0027-8424. @article{377,
title = {Paracrine Wnt signaling both promotes and inhibits human breast tumor growth.},
author = {JL Green and J La and KW Yum and P Desai and LW Rodewald and X Zhang and M Leblanc and R Nusse and MT Lewis and GM Wahl},
url = {http://www.pnas.org/cgi/pmidlookup?view=long&pmid=23559372},
doi = {10.1073/pnas.1303671110},
issn = {0027-8424},
year = {2013},
date = {2013-04-01},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
volume = {110},
pages = {6991-6},
abstract = {Wnt signaling in mouse mammary development and tumorigenesis has been heavily studied and characterized, but its role in human breast cancer remains elusive. Although Wnt inhibitors are in early clinical development, it is unclear whether they will be of therapeutic benefit to breast cancer patients, and subsequently, to which ones. To address this, we generated a panel of Wnt reporting human breast cancer cell lines and identified a previously unrecognized enrichment for the ability to respond to Wnt in the basal B or claudin-low subtype, which has a poor prognosis and no available targeted therapies. By co-injecting Wnt3A expressing human mammary fibroblasts with human breast cancer cell lines into mouse mammary fat pads, we showed that elevated paracrine Wnt signaling was correlated with accelerated tumor growth. Using this heterotypic system and a dual lentiviral reporter system that enables simultaneous real-time measurement of both Wnt-responsive cells and bulk tumor cells, we analyzed the outcome of elevated Wnt signaling in patient-derived xenograft (PDX) models. Interestingly, the PDX models exhibited responses not observed in the cell lines analyzed. Exogenous WNT3A promoted tumor growth in one human epidermal growth factor receptor 2-overexpressing PDX line but inhibited growth in a second PDX line obtained from a patient with triple-negative breast cancer. Tumor suppression was associated with squamous differentiation in the latter. Thus, our work suggests that paracrine Wnt signaling can either fuel or repress the growth of human breast cancers depending on yet to be determined aspects of the molecular pathways they express.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Wnt signaling in mouse mammary development and tumorigenesis has been heavily studied and characterized, but its role in human breast cancer remains elusive. Although Wnt inhibitors are in early clinical development, it is unclear whether they will be of therapeutic benefit to breast cancer patients, and subsequently, to which ones. To address this, we generated a panel of Wnt reporting human breast cancer cell lines and identified a previously unrecognized enrichment for the ability to respond to Wnt in the basal B or claudin-low subtype, which has a poor prognosis and no available targeted therapies. By co-injecting Wnt3A expressing human mammary fibroblasts with human breast cancer cell lines into mouse mammary fat pads, we showed that elevated paracrine Wnt signaling was correlated with accelerated tumor growth. Using this heterotypic system and a dual lentiviral reporter system that enables simultaneous real-time measurement of both Wnt-responsive cells and bulk tumor cells, we analyzed the outcome of elevated Wnt signaling in patient-derived xenograft (PDX) models. Interestingly, the PDX models exhibited responses not observed in the cell lines analyzed. Exogenous WNT3A promoted tumor growth in one human epidermal growth factor receptor 2-overexpressing PDX line but inhibited growth in a second PDX line obtained from a patient with triple-negative breast cancer. Tumor suppression was associated with squamous differentiation in the latter. Thus, our work suggests that paracrine Wnt signaling can either fuel or repress the growth of human breast cancers depending on yet to be determined aspects of the molecular pathways they express. |
Habib, SJ; Chen, BC; Tsai, FC; Anastassiadis, K; Meyer, T; Betzig, E; Nusse, R A localized Wnt signal orients asymmetric stem cell division in vitro. Journal Article In: Science (New York, N.Y.), vol. 339, pp. 1445-8, 2013, ISSN: 0036-8075. @article{375,
title = {A localized Wnt signal orients asymmetric stem cell division in vitro.},
author = {SJ Habib and BC Chen and FC Tsai and K Anastassiadis and T Meyer and E Betzig and R Nusse},
url = {http://www.sciencemag.org/cgi/pmidlookup?view=long&pmid=23520113},
doi = {10.1126/science.1231077},
issn = {0036-8075},
year = {2013},
date = {2013-03-01},
journal = {Science (New York, N.Y.)},
volume = {339},
pages = {1445-8},
abstract = {Developmental signals such as Wnts are often presented to cells in an oriented manner. To examine the consequences of local Wnt signaling, we immobilized Wnt proteins on beads and introduced them to embryonic stem cells in culture. At the single-cell level, the Wnt-bead induced asymmetric distribution of Wnt-β-catenin signaling components, oriented the plane of mitotic division, and directed asymmetric inheritance of centrosomes. Before cytokinesis was completed, the Wnt-proximal daughter cell expressed high levels of nuclear β-catenin and pluripotency genes, whereas the distal daughter cell acquired hallmarks of differentiation. We suggest that a spatially restricted Wnt signal induces an oriented cell division that generates distinct cell fates at predictable positions relative to the Wnt source.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Developmental signals such as Wnts are often presented to cells in an oriented manner. To examine the consequences of local Wnt signaling, we immobilized Wnt proteins on beads and introduced them to embryonic stem cells in culture. At the single-cell level, the Wnt-bead induced asymmetric distribution of Wnt-β-catenin signaling components, oriented the plane of mitotic division, and directed asymmetric inheritance of centrosomes. Before cytokinesis was completed, the Wnt-proximal daughter cell expressed high levels of nuclear β-catenin and pluripotency genes, whereas the distal daughter cell acquired hallmarks of differentiation. We suggest that a spatially restricted Wnt signal induces an oriented cell division that generates distinct cell fates at predictable positions relative to the Wnt source. |
Jan, TA; Chai, R; Sayyid, ZN; Amerongen, R; Xia, A; Wang, T; Sinkkonen, ST; Zeng, YA; Levin, JR; Heller, S; Nusse, R; Cheng, AG Tympanic border cells are Wnt-responsive and can act as progenitors for postnatal mouse cochlear cells. Journal Article In: Development (Cambridge, England), vol. 140, pp. 1196-206, 2013, ISSN: 0950-1991. @article{373,
title = {Tympanic border cells are Wnt-responsive and can act as progenitors for postnatal mouse cochlear cells.},
author = {TA Jan and R Chai and ZN Sayyid and R Amerongen and A Xia and T Wang and ST Sinkkonen and YA Zeng and JR Levin and S Heller and R Nusse and AG Cheng},
url = {http://dev.biologists.org/cgi/pmidlookup?view=long&pmid=23444352},
doi = {10.1242/dev.087528},
issn = {0950-1991},
year = {2013},
date = {2013-03-01},
journal = {Development (Cambridge, England)},
volume = {140},
pages = {1196-206},
abstract = {Permanent hearing loss is caused by the irreversible damage of cochlear sensory hair cells and nonsensory supporting cells. In the postnatal cochlea, the sensory epithelium is terminally differentiated, whereas tympanic border cells (TBCs) beneath the sensory epithelium are proliferative. The functions of TBCs are poorly characterized. Using an Axin2(lacZ) Wnt reporter mouse, we found transient but robust Wnt signaling and proliferation in TBCs during the first 3 postnatal weeks, when the number of TBCs decreases. In vivo lineage tracing shows that a subset of hair cells and supporting cells is derived postnatally from Axin2-expressing TBCs. In cochlear explants, Wnt agonists stimulated the proliferation of TBCs, whereas Wnt inhibitors suppressed it. In addition, purified Axin2(lacZ) cells were clonogenic and self-renewing in culture in a Wnt-dependent manner, and were able to differentiate into hair cell-like and supporting cell-like cells. Taken together, our data indicate that Axin2-positive TBCs are Wnt responsive and can act as precursors to sensory epithelial cells in the postnatal cochlea.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Permanent hearing loss is caused by the irreversible damage of cochlear sensory hair cells and nonsensory supporting cells. In the postnatal cochlea, the sensory epithelium is terminally differentiated, whereas tympanic border cells (TBCs) beneath the sensory epithelium are proliferative. The functions of TBCs are poorly characterized. Using an Axin2(lacZ) Wnt reporter mouse, we found transient but robust Wnt signaling and proliferation in TBCs during the first 3 postnatal weeks, when the number of TBCs decreases. In vivo lineage tracing shows that a subset of hair cells and supporting cells is derived postnatally from Axin2-expressing TBCs. In cochlear explants, Wnt agonists stimulated the proliferation of TBCs, whereas Wnt inhibitors suppressed it. In addition, purified Axin2(lacZ) cells were clonogenic and self-renewing in culture in a Wnt-dependent manner, and were able to differentiate into hair cell-like and supporting cell-like cells. Taken together, our data indicate that Axin2-positive TBCs are Wnt responsive and can act as precursors to sensory epithelial cells in the postnatal cochlea. |
Ardehali, R; Ali, SR; Inlay, MA; Abilez, OJ; Chen, MQ; Blauwkamp, TA; Yazawa, M; Gong, Y; Nusse, R; Drukker, M; Weissman, IL Prospective isolation of human embryonic stem cell-derived cardiovascular progenitors that integrate into human fetal heart tissue. Journal Article In: Proceedings of the National Academy of Sciences of the United States of America, vol. 110, pp. 3405-10, 2013, ISSN: 0027-8424. @article{371,
title = {Prospective isolation of human embryonic stem cell-derived cardiovascular progenitors that integrate into human fetal heart tissue.},
author = {R Ardehali and SR Ali and MA Inlay and OJ Abilez and MQ Chen and TA Blauwkamp and M Yazawa and Y Gong and R Nusse and M Drukker and IL Weissman},
url = {http://www.pnas.org/cgi/pmidlookup?view=long&pmid=23391730},
doi = {10.1073/pnas.1220832110},
issn = {0027-8424},
year = {2013},
date = {2013-02-01},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
volume = {110},
pages = {3405-10},
abstract = {A goal of regenerative medicine is to identify cardiovascular progenitors from human ES cells (hESCs) that can functionally integrate into the human heart. Previous studies to evaluate the developmental potential of candidate hESC-derived progenitors have delivered these cells into murine and porcine cardiac tissue, with inconclusive evidence regarding the capacity of these human cells to physiologically engraft in xenotransplantation assays. Further, the potential of hESC-derived cardiovascular lineage cells to functionally couple to human myocardium remains untested and unknown. Here, we have prospectively identified a population of hESC-derived ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells that give rise to cardiomyocytes, endothelial cells, and vascular smooth muscle cells in vitro at a clonal level. We observed rare clusters of ROR2(+) cells and diffuse expression of KDR and PDGFRα in first-trimester human fetal hearts. We then developed an in vivo transplantation model by transplanting second-trimester human fetal heart tissues s.c. into the ear pinna of a SCID mouse. ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells were delivered into these functioning fetal heart tissues: in contrast to traditional murine heart models for cell transplantation, we show structural and functional integration of hESC-derived cardiovascular progenitors into human heart.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
A goal of regenerative medicine is to identify cardiovascular progenitors from human ES cells (hESCs) that can functionally integrate into the human heart. Previous studies to evaluate the developmental potential of candidate hESC-derived progenitors have delivered these cells into murine and porcine cardiac tissue, with inconclusive evidence regarding the capacity of these human cells to physiologically engraft in xenotransplantation assays. Further, the potential of hESC-derived cardiovascular lineage cells to functionally couple to human myocardium remains untested and unknown. Here, we have prospectively identified a population of hESC-derived ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells that give rise to cardiomyocytes, endothelial cells, and vascular smooth muscle cells in vitro at a clonal level. We observed rare clusters of ROR2(+) cells and diffuse expression of KDR and PDGFRα in first-trimester human fetal hearts. We then developed an in vivo transplantation model by transplanting second-trimester human fetal heart tissues s.c. into the ear pinna of a SCID mouse. ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells were delivered into these functioning fetal heart tissues: in contrast to traditional murine heart models for cell transplantation, we show structural and functional integration of hESC-derived cardiovascular progenitors into human heart. |
Lim, X; Nusse, R Wnt signaling in skin development, homeostasis, and disease. Journal Article In: Cold Spring Harbor perspectives in biology, vol. 5, 2013. @article{369,
title = {Wnt signaling in skin development, homeostasis, and disease.},
author = {X Lim and R Nusse},
url = {http://cshperspectives.cshlp.org/cgi/pmidlookup?view=long&pmid=23209129},
doi = {10.1101/cshperspect.a008029},
year = {2013},
date = {2013-02-01},
journal = {Cold Spring Harbor perspectives in biology},
volume = {5},
abstract = {The skin and its appendages constitute the largest organ of the body. Its stratified epithelia offer protection from environmental stresses such as dehydration, irradiation, mechanical trauma, and pathogenic infection, whereas its appendages, like hair and sebaceous glands, help regulate body temperature as well as influence animal interaction and social behavior through camouflage and sexual signaling. To respond to and function effectively in a dynamic external environment, the skin and its appendages possess a remarkable ability to regenerate in a carefully controlled fashion. When this finely tuned homeostatic process is disrupted, skin diseases such as cancers may result. At present, the molecular signals that orchestrate cell proliferation, differentiation, and patterning in the skin remain incompletely understood. It is increasingly apparent that many morphogenetic pathways with key roles in development are also important in regulating skin biology. Of these, Wnt signaling has emerged as the dominant pathway controlling the patterning of skin and influencing the decisions of embryonic and adult stem cells to adopt the various cell lineages of the skin and its appendages, as well as subsequently controlling the function of differentiated skin cells. Here we will review established concepts and present recent advances in our understanding of the diverse roles that Wnt signaling plays in skin development, homeostasis, and disease.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The skin and its appendages constitute the largest organ of the body. Its stratified epithelia offer protection from environmental stresses such as dehydration, irradiation, mechanical trauma, and pathogenic infection, whereas its appendages, like hair and sebaceous glands, help regulate body temperature as well as influence animal interaction and social behavior through camouflage and sexual signaling. To respond to and function effectively in a dynamic external environment, the skin and its appendages possess a remarkable ability to regenerate in a carefully controlled fashion. When this finely tuned homeostatic process is disrupted, skin diseases such as cancers may result. At present, the molecular signals that orchestrate cell proliferation, differentiation, and patterning in the skin remain incompletely understood. It is increasingly apparent that many morphogenetic pathways with key roles in development are also important in regulating skin biology. Of these, Wnt signaling has emerged as the dominant pathway controlling the patterning of skin and influencing the decisions of embryonic and adult stem cells to adopt the various cell lineages of the skin and its appendages, as well as subsequently controlling the function of differentiated skin cells. Here we will review established concepts and present recent advances in our understanding of the diverse roles that Wnt signaling plays in skin development, homeostasis, and disease. |
Blauwkamp, TA; Nigam, S; Ardehali, R; Weissman, IL; Nusse, R Endogenous Wnt signalling in human embryonic stem cells generates an equilibrium of distinct lineage-specified progenitors. Journal Article In: Nature communications, vol. 3, pp. 1070, 2013. @article{367,
title = {Endogenous Wnt signalling in human embryonic stem cells generates an equilibrium of distinct lineage-specified progenitors.},
author = {TA Blauwkamp and S Nigam and R Ardehali and IL Weissman and R Nusse},
url = {http://dx.doi.org/10.1038/ncomms2064},
doi = {10.1038/ncomms2064},
year = {2013},
date = {2013-00-01},
journal = {Nature communications},
volume = {3},
pages = {1070},
abstract = {The pluripotent nature of human embryonic stem cells (hESCs) makes them convenient for deriving therapeutically relevant cells. Here we show using Wnt reporter hESC lines that the cells are heterogeneous with respect to endogenous Wnt signalling activity. Moreover, the level of Wnt signalling activity in individual cells correlates with differences in clonogenic potential and lineage-specific differentiation propensity. The addition of Wnt protein or, conversely, a small-molecule Wnt inhibitor (IWP2) reduces heterogeneity, allowing stable expansion of Wnt(high) or Wnt(low) hESC populations, respectively. On differentiation, the Wnt(high) hESCs predominantly form endodermal and cardiac cells, whereas the Wnt(low) hESCs generate primarily neuroectodermal cells. Thus, heterogeneity with respect to endogenous Wnt signalling underlies much of the inefficiency in directing hESCs towards specific cell types. The relatively uniform differentiation potential of the Wnt(high) and Wnt(low) hESCs leads to faster and more efficient derivation of targeted cell types from these populations.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The pluripotent nature of human embryonic stem cells (hESCs) makes them convenient for deriving therapeutically relevant cells. Here we show using Wnt reporter hESC lines that the cells are heterogeneous with respect to endogenous Wnt signalling activity. Moreover, the level of Wnt signalling activity in individual cells correlates with differences in clonogenic potential and lineage-specific differentiation propensity. The addition of Wnt protein or, conversely, a small-molecule Wnt inhibitor (IWP2) reduces heterogeneity, allowing stable expansion of Wnt(high) or Wnt(low) hESC populations, respectively. On differentiation, the Wnt(high) hESCs predominantly form endodermal and cardiac cells, whereas the Wnt(low) hESCs generate primarily neuroectodermal cells. Thus, heterogeneity with respect to endogenous Wnt signalling underlies much of the inefficiency in directing hESCs towards specific cell types. The relatively uniform differentiation potential of the Wnt(high) and Wnt(low) hESCs leads to faster and more efficient derivation of targeted cell types from these populations. |
2012
|
Amerongen, R; Bowman, AN; Nusse, R Developmental stage and time dictate the fate of Wnt/β-catenin-responsive stem cells in the mammary gland. Journal Article In: Cell stem cell, vol. 11, pp. 387-400, 2012, ISSN: 1934-5909. @article{363,
title = {Developmental stage and time dictate the fate of Wnt/β-catenin-responsive stem cells in the mammary gland.},
author = {R Amerongen and AN Bowman and R Nusse},
url = {http://linkinghub.elsevier.com/retrieve/pii/S1934-5909(12)00342-6},
doi = {10.1016/j.stem.2012.05.023},
issn = {1934-5909},
year = {2012},
date = {2012-09-01},
journal = {Cell stem cell},
volume = {11},
pages = {387-400},
abstract = {The mammary epithelium undergoes extensive growth and remodeling during pregnancy, suggesting a role for stem cells. Yet their origin, identity, and behavior in the intact tissue remain unknown. Using an Axin2(CreERT2) allele, we labeled and traced Wnt/β-catenin-responsive cells throughout mammary gland development. This reveals a switch in Wnt/β-catenin signaling around birth and shows that, depending on the developmental stage, Axin2(+) cells contribute differently to basal and luminal epithelial cell lineages of the mammary epithelium. Moreover, an important difference exists between the developmental potential tested in transplantation assays and that displayed by the same cell population in situ. Finally, Axin2(+) cells in the adult build alveolar structures during multiple pregnancies, demonstrating the existence of a Wnt/β-catenin-responsive adult stem cell. Our study uncovers dynamic changes in Wnt/β-catenin signaling in the mammary epithelium and offers insights into the developmental fate of mammary gland stem and progenitor cells.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The mammary epithelium undergoes extensive growth and remodeling during pregnancy, suggesting a role for stem cells. Yet their origin, identity, and behavior in the intact tissue remain unknown. Using an Axin2(CreERT2) allele, we labeled and traced Wnt/β-catenin-responsive cells throughout mammary gland development. This reveals a switch in Wnt/β-catenin signaling around birth and shows that, depending on the developmental stage, Axin2(+) cells contribute differently to basal and luminal epithelial cell lineages of the mammary epithelium. Moreover, an important difference exists between the developmental potential tested in transplantation assays and that displayed by the same cell population in situ. Finally, Axin2(+) cells in the adult build alveolar structures during multiple pregnancies, demonstrating the existence of a Wnt/β-catenin-responsive adult stem cell. Our study uncovers dynamic changes in Wnt/β-catenin signaling in the mammary epithelium and offers insights into the developmental fate of mammary gland stem and progenitor cells. |
Willert, K; Nusse, R Wnt proteins. Journal Article In: Cold Spring Harbor perspectives in biology, vol. 4, pp. a007864, 2012. @article{365,
title = {Wnt proteins.},
author = {K Willert and R Nusse},
url = {http://cshperspectives.cshlp.org/cgi/pmidlookup?view=long&pmid=22952392},
doi = {10.1101/cshperspect.a007864},
year = {2012},
date = {2012-09-01},
journal = {Cold Spring Harbor perspectives in biology},
volume = {4},
pages = {a007864},
abstract = {Wnt proteins comprise a major family of signaling molecules that orchestrate and influence a myriad of cell biological and developmental processes. Although our understanding of the role of Wnt signaling in regulating development and affecting disease, such as cancer, has been ever increasing, the study of the Wnt proteins themselves has been painstaking and slow moving. Despite advances in the biochemical characterization of Wnt proteins, many mysteries remain unsolved. In contrast to other developmental signaling molecules, such as fibroblast growth factors (FGF), transforming growth factors (TGFβ), and Sonic hedgehog (Shh), Wnt proteins have not conformed to many standard methods of protein production, such as bacterial overexpression, and analysis, such as ligand-receptor binding assays. The reasons for their recalcitrant nature are likely a consequence of the complex set of posttranslational modifications involving several highly specialized and poorly characterized processing enzymes. With the recent description of the first Wnt protein structure, the time is ripe to uncover and possibly resolve many of the remaining issues surrounding Wnt proteins and their interactions. Here we describe the process of maturation of Wnt from its initial translation to its eventual release from a cell and interactions in the extracellular environment.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Wnt proteins comprise a major family of signaling molecules that orchestrate and influence a myriad of cell biological and developmental processes. Although our understanding of the role of Wnt signaling in regulating development and affecting disease, such as cancer, has been ever increasing, the study of the Wnt proteins themselves has been painstaking and slow moving. Despite advances in the biochemical characterization of Wnt proteins, many mysteries remain unsolved. In contrast to other developmental signaling molecules, such as fibroblast growth factors (FGF), transforming growth factors (TGFβ), and Sonic hedgehog (Shh), Wnt proteins have not conformed to many standard methods of protein production, such as bacterial overexpression, and analysis, such as ligand-receptor binding assays. The reasons for their recalcitrant nature are likely a consequence of the complex set of posttranslational modifications involving several highly specialized and poorly characterized processing enzymes. With the recent description of the first Wnt protein structure, the time is ripe to uncover and possibly resolve many of the remaining issues surrounding Wnt proteins and their interactions. Here we describe the process of maturation of Wnt from its initial translation to its eventual release from a cell and interactions in the extracellular environment. |
Amerongen, R; Fuerer, C; Mizutani, M; Nusse, R Wnt5a can both activate and repress Wnt/β-catenin signaling during mouse embryonic development. Journal Article In: Developmental biology, vol. 369, pp. 101-14, 2012, ISSN: 0012-1606. @article{361,
title = {Wnt5a can both activate and repress Wnt/β-catenin signaling during mouse embryonic development.},
author = {R Amerongen and C Fuerer and M Mizutani and R Nusse},
url = {http://linkinghub.elsevier.com/retrieve/pii/S0012-1606(12)00349-1},
doi = {10.1016/j.ydbio.2012.06.020},
issn = {0012-1606},
year = {2012},
date = {2012-09-01},
journal = {Developmental biology},
volume = {369},
pages = {101-14},
abstract = {Embryonic development is controlled by a small set of signal transduction pathways, with vastly different phenotypic outcomes depending on the time and place of their recruitment. How the same molecular machinery can elicit such specific and distinct responses, remains one of the outstanding questions in developmental biology. Part of the answer may lie in the high inherent genetic complexity of these signaling cascades, as observed for the Wnt-pathway. The mammalian genome encodes multiple Wnt proteins and receptors, each of which show dynamic and tightly controlled expression patterns in the embryo. Yet how these components interact in the context of the whole organism remains unknown. Here we report the generation of a novel, inducible transgenic mouse model that allows spatiotemporal control over the expression of Wnt5a, a protein implicated in many developmental processes and multiple Wnt-signaling responses. We show that ectopic Wnt5a expression from E10.5 onwards results in a variety of developmental defects, including loss of hair follicles and reduced bone formation in the skull. Moreover, we find that Wnt5a can have dual signaling activities during mouse embryonic development. Specifically, Wnt5a is capable of both inducing and repressing β-catenin/TCF signaling in vivo, depending on the time and site of expression and the receptors expressed by receiving cells. These experiments show for the first time that a single mammalian Wnt protein can have multiple signaling activities in vivo, thereby furthering our understanding of how signaling specificity is achieved in a complex developmental context.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Embryonic development is controlled by a small set of signal transduction pathways, with vastly different phenotypic outcomes depending on the time and place of their recruitment. How the same molecular machinery can elicit such specific and distinct responses, remains one of the outstanding questions in developmental biology. Part of the answer may lie in the high inherent genetic complexity of these signaling cascades, as observed for the Wnt-pathway. The mammalian genome encodes multiple Wnt proteins and receptors, each of which show dynamic and tightly controlled expression patterns in the embryo. Yet how these components interact in the context of the whole organism remains unknown. Here we report the generation of a novel, inducible transgenic mouse model that allows spatiotemporal control over the expression of Wnt5a, a protein implicated in many developmental processes and multiple Wnt-signaling responses. We show that ectopic Wnt5a expression from E10.5 onwards results in a variety of developmental defects, including loss of hair follicles and reduced bone formation in the skull. Moreover, we find that Wnt5a can have dual signaling activities during mouse embryonic development. Specifically, Wnt5a is capable of both inducing and repressing β-catenin/TCF signaling in vivo, depending on the time and site of expression and the receptors expressed by receiving cells. These experiments show for the first time that a single mammalian Wnt protein can have multiple signaling activities in vivo, thereby furthering our understanding of how signaling specificity is achieved in a complex developmental context. |
Clevers, H; Nusse, R Wnt/β-catenin signaling and disease. Journal Article In: Cell, vol. 149, pp. 1192-205, 2012, ISSN: 0092-8674. @article{359,
title = {Wnt/β-catenin signaling and disease.},
author = {H Clevers and R Nusse},
url = {http://linkinghub.elsevier.com/retrieve/pii/S0092-8674(12)00586-7},
doi = {10.1016/j.cell.2012.05.012},
issn = {0092-8674},
year = {2012},
date = {2012-06-01},
journal = {Cell},
volume = {149},
pages = {1192-205},
abstract = {The WNT signal transduction cascade controls myriad biological phenomena throughout development and adult life of all animals. In parallel, aberrant Wnt signaling underlies a wide range of pathologies in humans. In this Review, we provide an update of the core Wnt/β-catenin signaling pathway, discuss how its various components contribute to disease, and pose outstanding questions to be addressed in the future.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
The WNT signal transduction cascade controls myriad biological phenomena throughout development and adult life of all animals. In parallel, aberrant Wnt signaling underlies a wide range of pathologies in humans. In this Review, we provide an update of the core Wnt/β-catenin signaling pathway, discuss how its various components contribute to disease, and pose outstanding questions to be addressed in the future. |
Edris, B; Espinosa, I; Mühlenberg, T; Mikels, A; Lee, CH; Steigen, SE; Zhu, S; Montgomery, KD; Lazar, AJ; Lev, D; Fletcher, JA; Beck, AH; West, RB; Nusse, R; Rijn, M ROR2 is a novel prognostic biomarker and a potential therapeutic target in leiomyosarcoma and gastrointestinal stromal tumour. Journal Article In: The Journal of pathology, vol. 227, pp. 223-33, 2012, ISSN: 0022-3417. @article{351,
title = {ROR2 is a novel prognostic biomarker and a potential therapeutic target in leiomyosarcoma and gastrointestinal stromal tumour.},
author = {B Edris and I Espinosa and T Mühlenberg and A Mikels and CH Lee and SE Steigen and S Zhu and KD Montgomery and AJ Lazar and D Lev and JA Fletcher and AH Beck and RB West and R Nusse and M Rijn},
url = {http://dx.doi.org/10.1002/path.3986},
doi = {10.1002/path.3986},
issn = {0022-3417},
year = {2012},
date = {2012-06-01},
journal = {The Journal of pathology},
volume = {227},
pages = {223-33},
abstract = {Soft-tissue sarcomas are a group of malignant tumours whose clinical management is complicated by morphological heterogeneity, inadequate molecular markers and limited therapeutic options. Receptor tyrosine kinases (RTKs) have been shown to play important roles in cancer, both as therapeutic targets and as prognostic biomarkers. An initial screen of gene expression data for 48 RTKs in 148 sarcomas showed that ROR2 was expressed in a subset of leiomyosarcoma (LMS), gastrointestinal stromal tumour (GIST) and desmoid-type fibromatosis (DTF). This was further confirmed by immunohistochemistry (IHC) on 573 tissue samples from 59 sarcoma tumour types. Here we provide evidence that ROR2 expression plays a role in the invasive abilities of LMS and GIST cells in vitro. We also show that knockdown of ROR2 significantly reduces tumour mass in vivo using a xenotransplantation model of LMS. Lastly, we show that ROR2 expression, as measured by IHC, predicts poor clinical outcome in patients with LMS and GIST, although it was not independent of other clinico-pathological features in a multivariate analysis, and that ROR2 expression is maintained between primary tumours and their metastases. Together, these results show that ROR2 is a useful prognostic indicator in the clinical management of these soft-tissue sarcomas and may represent a novel therapeutic target. Copyright textcopyright 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Soft-tissue sarcomas are a group of malignant tumours whose clinical management is complicated by morphological heterogeneity, inadequate molecular markers and limited therapeutic options. Receptor tyrosine kinases (RTKs) have been shown to play important roles in cancer, both as therapeutic targets and as prognostic biomarkers. An initial screen of gene expression data for 48 RTKs in 148 sarcomas showed that ROR2 was expressed in a subset of leiomyosarcoma (LMS), gastrointestinal stromal tumour (GIST) and desmoid-type fibromatosis (DTF). This was further confirmed by immunohistochemistry (IHC) on 573 tissue samples from 59 sarcoma tumour types. Here we provide evidence that ROR2 expression plays a role in the invasive abilities of LMS and GIST cells in vitro. We also show that knockdown of ROR2 significantly reduces tumour mass in vivo using a xenotransplantation model of LMS. Lastly, we show that ROR2 expression, as measured by IHC, predicts poor clinical outcome in patients with LMS and GIST, although it was not independent of other clinico-pathological features in a multivariate analysis, and that ROR2 expression is maintained between primary tumours and their metastases. Together, these results show that ROR2 is a useful prognostic indicator in the clinical management of these soft-tissue sarcomas and may represent a novel therapeutic target. Copyright textcopyright 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd. |
Nusse, R; Varmus, H Three decades of Wnts: a personal perspective on how a scientific field developed. Journal Article In: The EMBO journal, vol. 31, pp. 2670-84, 2012, ISSN: 0261-4189. @article{357,
title = {Three decades of Wnts: a personal perspective on how a scientific field developed.},
author = {R Nusse and H Varmus},
url = {http://dx.doi.org/10.1038/emboj.2012.146},
doi = {10.1038/emboj.2012.146},
issn = {0261-4189},
year = {2012},
date = {2012-05-01},
journal = {The EMBO journal},
volume = {31},
pages = {2670-84},
abstract = {Wnt genes and components of Wnt signalling pathways have been implicated in a wide spectrum of important biological phenomena, ranging from early organismal development to cell behaviours to several diseases, especially cancers. Emergence of the field of Wnt signalling can be largely traced back to the discovery of the first mammalian Wnt gene in 1982. In this essay, we mark the thirtieth anniversary of that discovery by describing some of the critical scientific developments that led to the flowering of this field of research.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Wnt genes and components of Wnt signalling pathways have been implicated in a wide spectrum of important biological phenomena, ranging from early organismal development to cell behaviours to several diseases, especially cancers. Emergence of the field of Wnt signalling can be largely traced back to the discovery of the first mammalian Wnt gene in 1982. In this essay, we mark the thirtieth anniversary of that discovery by describing some of the critical scientific developments that led to the flowering of this field of research. |
Chai, R; Kuo, B; Wang, T; Liaw, EJ; Xia, A; Jan, TA; Liu, Z; Taketo, MM; Oghalai, JS; Nusse, R; Zuo, J; Cheng, AG Wnt signaling induces proliferation of sensory precursors in the postnatal mouse cochlea. Journal Article In: Proceedings of the National Academy of Sciences of the United States of America, vol. 109, pp. 8167-72, 2012, ISSN: 0027-8424. @article{355,
title = {Wnt signaling induces proliferation of sensory precursors in the postnatal mouse cochlea.},
author = {R Chai and B Kuo and T Wang and EJ Liaw and A Xia and TA Jan and Z Liu and MM Taketo and JS Oghalai and R Nusse and J Zuo and AG Cheng},
url = {http://www.pnas.org/cgi/pmidlookup?view=long&pmid=22562792},
doi = {10.1073/pnas.1202774109},
issn = {0027-8424},
year = {2012},
date = {2012-05-01},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
volume = {109},
pages = {8167-72},
abstract = {Inner ear hair cells are specialized sensory cells essential for auditory function. Previous studies have shown that the sensory epithelium is postmitotic, but it harbors cells that can behave as progenitor cells in vitro, including the ability to form new hair cells. Lgr5, a Wnt target gene, marks distinct supporting cell types in the neonatal cochlea. Here, we tested the hypothesis that Lgr5(+) cells are Wnt-responsive sensory precursor cells. In contrast to their quiescent in vivo behavior, Lgr5(+) cells isolated by flow cytometry from neonatal Lgr5(EGFP-CreERT2/+) mice proliferated and formed clonal colonies. After 10 d in culture, new sensory cells formed and displayed specific hair cell markers (myo7a, calretinin, parvalbumin, myo6) and stereocilia-like structures expressing F-actin and espin. In comparison with other supporting cells, Lgr5(+) cells were enriched precursors to myo7a(+) cells, most of which formed without mitotic division. Treatment with Wnt agonists increased proliferation and colony-formation capacity. Conversely, small-molecule inhibitors of Wnt signaling suppressed proliferation without compromising the myo7a(+) cells formed by direct differentiation. In vivo lineage tracing supported the idea that Lgr5(+) cells give rise to myo7a(+) hair cells in the neonatal Lgr5(EGFP-CreERT2/+) cochlea. In addition, overexpression of β-catenin initiated proliferation and led to transient expansion of Lgr5(+) cells within the cochlear sensory epithelium. These results suggest that Lgr5 marks sensory precursors and that Wnt signaling can promote their proliferation and provide mechanistic insights into Wnt-responsive progenitor cells during sensory organ development.},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
Inner ear hair cells are specialized sensory cells essential for auditory function. Previous studies have shown that the sensory epithelium is postmitotic, but it harbors cells that can behave as progenitor cells in vitro, including the ability to form new hair cells. Lgr5, a Wnt target gene, marks distinct supporting cell types in the neonatal cochlea. Here, we tested the hypothesis that Lgr5(+) cells are Wnt-responsive sensory precursor cells. In contrast to their quiescent in vivo behavior, Lgr5(+) cells isolated by flow cytometry from neonatal Lgr5(EGFP-CreERT2/+) mice proliferated and formed clonal colonies. After 10 d in culture, new sensory cells formed and displayed specific hair cell markers (myo7a, calretinin, parvalbumin, myo6) and stereocilia-like structures expressing F-actin and espin. In comparison with other supporting cells, Lgr5(+) cells were enriched precursors to myo7a(+) cells, most of which formed without mitotic division. Treatment with Wnt agonists increased proliferation and colony-formation capacity. Conversely, small-molecule inhibitors of Wnt signaling suppressed proliferation without compromising the myo7a(+) cells formed by direct differentiation. In vivo lineage tracing supported the idea that Lgr5(+) cells give rise to myo7a(+) hair cells in the neonatal Lgr5(EGFP-CreERT2/+) cochlea. In addition, overexpression of β-catenin initiated proliferation and led to transient expansion of Lgr5(+) cells within the cochlear sensory epithelium. These results suggest that Lgr5 marks sensory precursors and that Wnt signaling can promote their proliferation and provide mechanistic insights into Wnt-responsive progenitor cells during sensory organ development. |
Nusse, R Wnt signaling. Journal Article In: Cold Spring Harbor perspectives in biology, vol. 4, 2012. @article{353,
title = {Wnt signaling.},
author = {R Nusse},
url = {http://cshperspectives.cshlp.org/cgi/pmidlookup?view=long&pmid=22550232},
doi = {10.1101/cshperspect.a011163},
year = {2012},
date = {2012-05-01},
journal = {Cold Spring Harbor perspectives in biology},
volume = {4},
keywords = {},
pubstate = {published},
tppubtype = {article}
}
|