151
|
Abstract
The skin constantly renews itself throughout adult life, and the hair follicle undergoes a perpetual cycle of growth and degeneration. Stem cells (SCs) residing in the epidermis and hair follicle ensure the maintenance of adult skin homeostasis and hair regeneration, but they also participate in the repair of the epidermis after injuries. We summarize here the current knowledge of epidermal SCs of the adult skin. We discuss their fundamental characteristics, the methods recently designed to isolate these cells, the genes preferentially expressed in the multipotent SC niche, and the signaling pathways involved in SC niche formation, SC maintenance, and activation. Finally, we speculate on how the deregulation of these pathways may lead to cancer formation.
Collapse
Affiliation(s)
- Cédric Blanpain
- Howard Hughes Medical Institute, The Rockefeller University, New York, New York 10021, USA
| | | |
Collapse
|
152
|
Abstract
A remarkable interdisciplinary effort has unraveled the WNT (Wingless and INT-1) signal transduction cascade over the last two decades. Wnt genes encode small secreted proteins that are found in all animal genomes. Wnt signaling is involved in virtually every aspect of embryonic development and also controls homeostatic self-renewal in a number of adult tissues. Germline mutations in the Wnt pathway cause several hereditary diseases, and somatic mutations are associated with cancer of the intestine and a variety of other tissues.
Collapse
Affiliation(s)
- Hans Clevers
- Hubrecht Laboratory and Utrecht University, Uppsalalaan 8, 3584CT, Utrecht, the Netherlands.
| |
Collapse
|
153
|
Estrach S, Ambler CA, Lo Celso C, Hozumi K, Watt FM. Jagged 1 is a beta-catenin target gene required for ectopic hair follicle formation in adult epidermis. Development 2006; 133:4427-38. [PMID: 17035290 DOI: 10.1242/dev.02644] [Citation(s) in RCA: 193] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The Wnt and Notch signalling pathways regulate hair follicle maintenance, but how they intersect is unknown. We show that Notch signalling is active in the hair follicle pre-cortex, a region of high Wnt activity, where commitment to hair lineages occurs. Deletion of jagged 1 (Jag1) results in inhibition of the hair growth cycle and conversion of hair follicles into cysts of cells undergoing interfollicular epidermal differentiation. Conversely, activation of Notch in adult epidermis triggers expansion of the base of the hair follicle, sebaceous gland enlargement and abnormal clumping of the follicles. In adult epidermis, the induction of new hair follicle formation by beta-catenin is prevented by blocking Notch signalling pharmacologically or through Jag1 deletion. Conversely, activation of both pathways accelerates growth and differentiation of ectopic follicles. beta-catenin stimulates Notch signalling by inducing Jag1 transcription. We conclude that the Notch pathway acts downstream of the Wnt/beta-catenin pathway to determine epidermal cell fate.
Collapse
Affiliation(s)
- Soline Estrach
- Keratinocyte Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, UK
| | | | | | | | | |
Collapse
|
154
|
Gerdes MJ, Myakishev M, Frost NA, Rishi V, Moitra J, Acharya A, Levy MR, Park SW, Glick A, Yuspa SH, Vinson C. Activator protein-1 activity regulates epithelial tumor cell identity. Cancer Res 2006; 66:7578-88. [PMID: 16885357 DOI: 10.1158/0008-5472.can-06-1247] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To examine the consequences of inhibiting activator protein-1 (AP-1) transcription factors in skin, transgenic mice were generated, which use the tetracycline system to conditionally express A-FOS, a dominant negative that inhibits AP-1 DNA binding. Older mice develop mild alopecia and hyperplasia of sebaceous glands, particularly around the eyes. When A-FOS was expressed during chemical-induced skin carcinogenesis, mice do not develop characteristic benign and malignant squamous lesions but instead develop benign sebaceous adenomas containing a signature mutation in the H-ras proto-oncogene. Inhibiting AP-1 activity after tumor formation caused squamous tumors to transdifferentiate into sebaceous tumors. Furthermore, reactivating AP-1 in sebaceous tumors results in a reciprocal transdifferentiation into squamous tumors. In both cases of transdifferentiation, individual cells express molecular markers for both cell types, indicating individual tumor cells have the capacity to express multiple lineages. Molecular characterization of cultured keratinocytes and tumor material indicates that AP-1 regulates the balance between the wnt/beta-catenin and hedgehog signaling pathways that determine squamous and sebaceous lineages, respectively. Chromatin immunoprecipitation analysis indicates that c-Jun binds several wnt promoters, which are misregulated by A-FOS expression, suggesting that members of the wnt pathway can be a primary targets of AP-1 transcriptional regulation. Thus, AP-1 activity regulates tumor cell lineage and is essential to maintain the squamous tumor cell identity.
Collapse
Affiliation(s)
- Michael J Gerdes
- Laboratory of Cellular Carcinogenesis and Tumor Promotion, National Cancer Institute, Center for Cancer Research, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Wang J, Lu Z, Au JLS. Protection Against Chemotherapy-Induced Alopecia. Pharm Res 2006; 23:2505-14. [PMID: 16972183 DOI: 10.1007/s11095-006-9105-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2006] [Accepted: 06/28/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE The goal is to provide an overview on the advances in protection against chemotherapy-induced alopecia (CIA). MATERIALS AND METHODS The four major parts of this review are (a) overview of the hair follicle biology, (b) characteristics of CIA, (c) state-of-the-art animal models of CIA, and (d) experimental approaches on protection against CIA. RESULTS The hair follicle represents an unintended target of cancer chemotherapy. CIA is a significant side effect that compromises the quality of life of patients. Overcoming CIA represents an area of unmet needs, especially for females and children. Significant progresses have been made in the last decade on the pathobiology of CIA. The pharmacological agents under evaluation include drug-specific antibodies, hair growth cycle modifiers, cytokines and growth factors, antioxidants, cell cycle or proliferation modifiers, and inhibitors of apoptosis. Their potential applications and limitations are discussed. CONCLUSION Multiple classes of agents with different action mechanisms have been evaluated in animal CIA models. Most of these protective agents have activity limited to a single chemotherapeutic agent. In comparison, calcitriol and cyclosporine A have broader spectrum of activity and can prevent against CIA by multiple chemotherapeutic agents. Among the three agents that have been evaluated in humans, AS101 and Minoxidil were able to reduce the severity or shorten the duration of CIA but could not prevent CIA.
Collapse
Affiliation(s)
- Jie Wang
- College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | | | | |
Collapse
|
156
|
Markova NG, Pinkas-Sarafova A, Simon M. A Metabolic Enzyme of the Short-Chain Dehydrogenase/Reductase Superfamily May Moonlight in the Nucleus as a Repressor of Promoter Activity. J Invest Dermatol 2006; 126:2019-31. [PMID: 16691198 DOI: 10.1038/sj.jid.5700347] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Transcriptional repression often depends on the action of recruited co-repressor complexes with intrinsic enzymatic activities. The composition of these complexes depends on the nicotine amide dinucleotide co-factors and is thus directly reflective of the metabolic state of the cells. This study provides evidence that an enzyme, hRoDH-E2, with cytoplasmic phosphorylated and reduced forms of NAD-dependent retinol dehydrogenase activity may function in the nucleus as a transcriptional repressor. By using the promoter of the epidermal late differentiation marker profilaggrin as a model, we show that both in vivo and in vitro the protein is recruited over the promoter. hRoDH-E2 represses profilaggrin promoter activity by altering the function of other activators, such as Sp1. The repressive function is associated with the ability of nuclear hRoDH-E2 to modulate the acetylation/deacetylation activity in the vicinity of transcription initiation site. These findings add hRoDH-E2 to the small group of metabolic enzymes, which, by being recruited over promoter regions, could directly link the cytoplasmic and nuclear functions within the cell.
Collapse
Affiliation(s)
- Nelli G Markova
- Living Skin Bank, Department of Oral Biology and Pathology, School of Dental Medicine, SUNY Stony Brook, New York 11794, USA.
| | | | | |
Collapse
|
157
|
Kuraguchi M, Wang XP, Bronson RT, Rothenberg R, Ohene-Baah NY, Lund JJ, Kucherlapati M, Maas RL, Kucherlapati R. Adenomatous polyposis coli (APC) is required for normal development of skin and thymus. PLoS Genet 2006; 2:e146. [PMID: 17002498 PMCID: PMC1564426 DOI: 10.1371/journal.pgen.0020146] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2006] [Accepted: 07/28/2006] [Indexed: 11/19/2022] Open
Abstract
The tumor suppressor gene Apc (adenomatous polyposis coli) is a member of the Wnt signaling pathway that is involved in development and tumorigenesis. Heterozygous knockout mice for Apc have a tumor predisposition phenotype and homozygosity leads to embryonic lethality. To understand the role of Apc in development we generated a floxed allele. These mice were mated with a strain carrying Cre recombinase under the control of the human Keratin 14 (K14) promoter, which is active in basal cells of epidermis and other stratified epithelia. Mice homozygous for the floxed allele that also carry the K14-cre transgene were viable but had stunted growth and died before weaning. Histological and immunochemical examinations revealed that K14-cre-mediated Apc loss resulted in aberrant growth in many ectodermally derived squamous epithelia, including hair follicles, teeth, and oral and corneal epithelia. In addition, squamous metaplasia was observed in various epithelial-derived tissues, including the thymus. The aberrant growth of hair follicles and other appendages as well as the thymic abnormalities in K14-cre; Apc(CKO/CKO) mice suggest the Apc gene is crucial in embryonic cells to specify epithelial cell fates in organs that require epithelial-mesenchymal interactions for their development.
Collapse
Affiliation(s)
- Mari Kuraguchi
- Harvard-Partners Center for Genetics and Genomics, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Xiu-Ping Wang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Roderick T Bronson
- Rodent Histopathology Core, Dana-Farber Harvard Cancer Center, Boston, Massachusetts, United States of America
| | - Rebecca Rothenberg
- Harvard-Partners Center for Genetics and Genomics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nana Yaw Ohene-Baah
- Harvard-Partners Center for Genetics and Genomics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jennifer J Lund
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Melanie Kucherlapati
- Harvard-Partners Center for Genetics and Genomics, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Richard L Maas
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Raju Kucherlapati
- Harvard-Partners Center for Genetics and Genomics, Harvard Medical School, Boston, Massachusetts, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
158
|
Lotem J, Sachs L. Epigenetics and the plasticity of differentiation in normal and cancer stem cells. Oncogene 2006; 25:7663-72. [PMID: 16847453 DOI: 10.1038/sj.onc.1209816] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Embryonic stem cells are characterized by their differentiation to all cell types during embryogenesis. In adult life, different tissues also have somatic stem cells, called adult stem cells, which in specific niches can undergo multipotent differentiation. The use of these adult stem cells has considerable therapeutic potential for the regeneration of damaged tissues. In both embryonic and adult stem cells, differentiation is controlled by epigenetic mechanisms, and the plasticity of differentiation in these cells is associated with transcription accessibility for genes expressed in different normal tissues. Abnormalities in genetic and/or epigenetic controls can lead to development of cancer, which is maintained by self-renewing cancer stem cells. Although the genetic abnormalities produce defects in growth and differentiation in cancer stem cells, these cells have not always lost the ability to undergo differentiation through epigenetic changes that by-pass the genomic abnormalities, thus creating the basis for differentiation therapy. Like normal stem cells, cancer stem cells can show plasticity for differentiation. This plasticity of cancer stem cells is also associated with transcription accessibility for genes that are normally expressed in different tissues, including tissues other than those from which the cancers originated. This broad transcription accessibility can also contribute to the behavior of cancer cells by overexpressing genes that promote cell viability, growth and metastasis.
Collapse
Affiliation(s)
- J Lotem
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
159
|
Abstract
The Keystone Symposium on `Wnt and β-catenin signaling in development and disease' was held recently in Snowbird, UT, USA. Organized by Mariann Bienz and Hans Clevers, this meeting covered a wide range of topics, including Wnt protein biogenesis, Wnt receptors and signaling pathways,β-catenin/Tcf complexes and gene expression, Wnt signaling in development, cancer, stem cell biology and regeneration, and therapeutics that target the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Xi He
- Neurobiology Program, Children's Hospital and Harvard Medical School, Boston, MA 02115-5724, USA.
| | | |
Collapse
|
160
|
Abstract
Skin and bone - what comes to mind at hearing this phrase? While certainly a metaphor for disease, it also defines two very different tissues, one a flexible and contiguous outer covering, the other a morphologically diverse hard tissue distributed at over 200 sites in the body. As the accompanying series of Reviews highlights, these tissues are indeed diverse, but there are also surprising similarities. Skin is the interface between the internal organs and the environment, and as such plays a crucial role in the body's defense mechanism. The skin and its many appendages are responsible for functions as diverse as epidermal barrier and defense, immune surveillance, UV protection, thermoregulation, sweating, lubrication, pigmentation, the sensations of pain and touch, and, importantly, the protection of various stem cell niches in the skin. Bone serves a number of purposes: it provides protection for vital organs, a lever for locomotion, a reservoir for calcium, and the site of adult hematopoiesis. The tissue is composed of osteoblasts, osteoclasts, and their individual precursors plus a complex mixture of mesenchymal, myeloid, and lymphoid cells in the marrow space. Finally, the endothelial microenvironment provides nutrition and is a conduit for the influx and emigration of cells that impact bone biology in several important ways. This Review series guides the reader through these various facets of 2 diverse, yet interdependent, tissues.
Collapse
Affiliation(s)
- F. Patrick Ross
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
Departments of Dermatology and Genetics and Development, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Angela M. Christiano
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
Departments of Dermatology and Genetics and Development, Columbia University College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
161
|
Cho HH, Kim YJ, Kim SJ, Kim JH, Bae YC, Ba B, Jung JS. Endogenous Wnt signaling promotes proliferation and suppresses osteogenic differentiation in human adipose derived stromal cells. ACTA ACUST UNITED AC 2006; 12:111-21. [PMID: 16499448 DOI: 10.1089/ten.2006.12.111] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Multipotential adult mesenchymal stem cells (MSC) are able to differentiate along several known lineages, and lineage commitment is tightly regulated through specific cellular mediators and interactions. Human adipose tissues contain cell populations that have similar characteristics to bone marrow stromal cells. Wnt proteins have been reported to be involved in proliferation and differentiation of stem cells. RNA interference (RNAi) has recently emerged as a specific and efficient method to silence gene expression in mammalian cells. To analyze the role of beta-catenin signaling in human adipose stromal cells (hADSC), the effects of beta-catenin short hairpin RNAs (shRNA) expression and Wnt3a conditioned media on the growth and differentiation properties of hADSC were examined. Expression of an RNAi molecule to beta-catenin from a lentivirus vector decreased beta-catenin expression in hADSC, as indicated by Western blot and immunohistochemistry. Cells transduced with sibeta-catenin lentivirus had decreased CFU and lower numbers of cells per colony than transduced control cells, but this outcome did not result from altered attachment efficiency of hADSC. The inhibition of beta-catenin signal by RNAi expression increased osteogenic differentiation. The treatment of Wnt3a conditioned media increased cellular beta-catenin levels and the rate of cellular proliferation, but inhibited osteogenic differentiation. Transduction of beta-catenin RNAi lentivirus blocked the effect of Wnt3a on proliferation of hADSC. Taken together, these findings indicate that endogenous Wnt3a plays an important role in the regulation of proliferation and differentiation of hADSC.
Collapse
Affiliation(s)
- Hyun Hwa Cho
- Department of Physiology, College of Medicine, Pusan National University, Pusan, Korea
| | | | | | | | | | | | | |
Collapse
|
162
|
Crosnier C, Stamataki D, Lewis J. Organizing cell renewal in the intestine: stem cells, signals and combinatorial control. Nat Rev Genet 2006; 7:349-59. [PMID: 16619050 DOI: 10.1038/nrg1840] [Citation(s) in RCA: 537] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The lining of the intestine is renewed at an extraordinary rate, outpacing all other tissues in the vertebrate body. The renewal process is neatly organized in space, so that the whole production line, from the ever-youthful stem cells to their dying, terminally differentiated progeny, is laid out to view in histological sections. A flurry of recent papers has clarified the key regulatory signals and brought us to the point where we can begin to give a coherent account, for at least one tissue, of how these signals collaborate to organize the architecture and behaviour of a stem-cell system.
Collapse
Affiliation(s)
- Cécile Crosnier
- Vertebrate Development Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, UK
| | | | | |
Collapse
|
163
|
Stewart R, Stojkovic M, Lako M. Mechanisms of self-renewal in human embryonic stem cells. Eur J Cancer 2006; 42:1257-72. [PMID: 16630716 DOI: 10.1016/j.ejca.2006.01.033] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Accepted: 01/23/2006] [Indexed: 01/07/2023]
Abstract
Embryonic stem cells (ESCs) are the pluripotent cell population derived from the inner cell mass of pre-implantation embryos and are characterised by prolonged self-renewal and the potential to differentiate into cells representing all three germ layers both in vitro and in vivo. Preservation of the undifferentiated status of the ESC population requires the maintenance of self-renewal whilst inhibiting differentiation and regulating senescence and apoptosis. In this review, we discuss the intrinsic and extrinsic factors associated with self-renewal process, together with possible signalling pathway interactions and mechanisms of regulation.
Collapse
Affiliation(s)
- Rebecca Stewart
- Centre for Stem Cell Biology and Developmental Genetics, Institute of Human Genetics, Newcastle University, International Centre for Life, Central Parkway, Newcastle-Upon-Tyne NE1 3BZ, UK.
| | | | | |
Collapse
|
164
|
Wu X, Quondamatteo F, Lefever T, Czuchra A, Meyer H, Chrostek A, Paus R, Langbein L, Brakebusch C. Cdc42 controls progenitor cell differentiation and beta-catenin turnover in skin. Genes Dev 2006; 20:571-85. [PMID: 16510873 PMCID: PMC1410804 DOI: 10.1101/gad.361406] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Differentiation of skin stem cells into hair follicles (HFs) requires the inhibition of beta-catenin degradation, which is controlled by a complex containing axin and the protein kinase GSK3beta. Using conditional gene targeting in mice, we show now that the small GTPase Cdc42 is crucial for differentiation of skin progenitor cells into HF lineage and that it regulates the turnover of beta-catenin. In the absence of Cdc42, degradation of beta-catenin was increased corresponding to a decreased phosphorylation of GSK3beta at Ser 9 and an increased phosphorylation of axin, which is known to be required for binding of beta-catenin to the degradation machinery. Cdc42-mediated regulation of beta-catenin turnover was completely dependent on PKCzeta, which associated with Cdc42, Par6, and Par3. These data suggest that Cdc42 regulation of beta-catenin turnover is important for terminal differentiation of HF progenitor cells in vivo.
Collapse
Affiliation(s)
- Xunwei Wu
- Max Planck Institute of Biochemistry, Heisenberg Group Regulation of Cytoskeletal Organization, Department of Molecular Medicine, 82152 Martinsried, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
165
|
Takeda H, Lyle S, Lazar AJF, Zouboulis CC, Smyth I, Watt FM. Human sebaceous tumors harbor inactivating mutations in LEF1. Nat Med 2006; 12:395-7. [PMID: 16565724 DOI: 10.1038/nm1386] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2006] [Accepted: 03/01/2006] [Indexed: 11/09/2022]
Abstract
We found that one-third of human sebaceous tumors examined had double-nucleotide substitutions in the same LEF1 allele, irrespective of DNA mismatch repair status. The mutations impaired both LEF1 binding to beta-catenin and transcriptional activation, and are the first tumor-associated mutations that inactivate Wnt signaling. Mutant LEF1 not only inhibited expression of beta-catenin target genes but also stimulated expression of sebocyte markers, suggesting that it may determine the differentiated characteristics of sebaceous tumors.
Collapse
Affiliation(s)
- Hikaru Takeda
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, UK
| | | | | | | | | | | |
Collapse
|
166
|
Wildwater M, Campilho A, Perez-Perez JM, Heidstra R, Blilou I, Korthout H, Chatterjee J, Mariconti L, Gruissem W, Scheres B. The RETINOBLASTOMA-RELATED gene regulates stem cell maintenance in Arabidopsis roots. Cell 2006; 123:1337-49. [PMID: 16377572 DOI: 10.1016/j.cell.2005.09.042] [Citation(s) in RCA: 258] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2005] [Revised: 08/19/2005] [Accepted: 09/20/2005] [Indexed: 12/16/2022]
Abstract
The maintenance of stem cells in defined locations is crucial for all multicellular organisms. Although intrinsic factors and signals for stem cell fate have been identified in several species, it has remained unclear how these connect to the ability to reenter the cell cycle that is one of the defining properties of stem cells. We show that local reduction of expression of the RETINOBLASTOMA-RELATED (RBR) gene in Arabidopsis roots increases the amount of stem cells without affecting cell cycle duration in mitotically active cells. Conversely, induced RBR overexpression dissipates stem cells prior to arresting other mitotic cells. Overexpression of D cyclins, KIP-related proteins, and E2F factors also affects root stem cell pool size, and genetic interactions suggest that these factors function in a canonical RBR pathway to regulate somatic stem cells. Expression analysis and genetic interactions position RBR-mediated regulation of the stem cell state downstream of the patterning gene SCARECROW.
Collapse
Affiliation(s)
- Marjolein Wildwater
- Department of Molecular Cell Biology, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Ouji Y, Yoshikawa M, Shiroi A, Ishizaka S. Wnt-10b promotes differentiation of skin epithelial cells in vitro. Biochem Biophys Res Commun 2006; 342:28-35. [PMID: 16469292 DOI: 10.1016/j.bbrc.2006.01.104] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Accepted: 01/20/2006] [Indexed: 10/25/2022]
Abstract
To evaluate the role of Wnt-10b in epithelial differentiation, we investigated the effects of Wnt-10b on adult mouse-derived primary skin epithelial cells (MPSEC). Recombinant Wnt-10b protein (rWnt-10b) was prepared using a gene engineering technique and MPSEC were cultured in its presence, which resulted in morphological changes from cuboidal to spindle-shaped and inhibited their proliferation. Further, involvement of the canonical Wnt signal pathway was also observed. MPSEC treated with rWnt-10b showed characteristics of the hair shaft and inner root sheath of the hair follicle, in results of Ayoub Shklar staining and immunocytochemistry. Further, the cells expressed mRNA for differentiated epithelial cells, including keratin 1, keratin 2, loricrin, mHa5, and mHb5, in association with a decreased expression of the basal cell marker keratin 5. These results suggest that Wnt-10b promotes the differentiation of MPSEC.
Collapse
Affiliation(s)
- Yukiteru Ouji
- Program in Tissue Engineering and Department of Parasitology, Nara Medical University, Kashihara, Nara, Japan.
| | | | | | | |
Collapse
|
168
|
Ouji Y, Yoshikawa M, Shiroi A, Ishizaka S. Wnt-10b secreted from lymphocytes promotes differentiation of skin epithelial cells. Biochem Biophys Res Commun 2006; 342:1063-9. [PMID: 16510119 DOI: 10.1016/j.bbrc.2006.02.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Accepted: 02/04/2006] [Indexed: 11/18/2022]
Abstract
Wnt-10b was originally isolated from lymphoid tissue and is known to be involved in a wide range of biological actions, while recently it was found to be expressed early in the development of hair follicles. However, few studies have been conducted concerning the role of Wnt-10b with the differentiation of skin epithelial cells. To evaluate its role in epithelial differentiation, we purified Wnt-10b from the supernatant of a concanavalin A-stimulated lymphocyte culture using an affinity column and investigated its effects on the differentiation of adult mouse-derived primary skin epithelial cells (MPSEC). MPSEC cultured with Wnt-10b showed morphological changes from cuboidal to spindle-shaped with inhibited proliferation, and also obtained characteristics of the hair shaft and inner root sheath of the hair follicle, represented by red-colored Ayoub Shklar staining, and reactions to AE-13 and AE-15 as seen with immunocytology. Further, RT-PCR analysis demonstrated the expression of mRNA for keratin 1, keratin 2, loricrin, mHa5, and mHb5, in association with a decreased expression of the basal cell marker keratin 5, in Wnt-10b-treated MPSEC. In addition, involvement of the canonical Wnt signal pathway was demonstrated by a TCF reporter (pTOPFLASH) assay. These results suggest that Wnt-10b promotes the differentiation of MPSEC and may play an important role in hair follicle development by promoting differentiation of epithelial cells.
Collapse
Affiliation(s)
- Yukiteru Ouji
- Program in Tissue Engineering and Department of Parasitology, Nara Medical University, Kashihara, Nara, Japan.
| | | | | | | |
Collapse
|
169
|
Abstract
Microchimerism, the stable presence of foreign cells in an individual, may result from trafficking during pregnancy or from organ or hematopoietic transplantation, and has been hypothesized to cause autoimmunity and certain skin diseases. Yet microchimeric cells are found in normal individuals and may be important to tissue repair. Thus microchimerism may be common, and finding microchimeric cells in diseased as well as normal tissue may be a "true-true unrelated" situation.
Collapse
Affiliation(s)
- Anita C Gilliam
- Department of Dermatology, Case/University Hospitals of Cleveland, Cleveland, Ohio 44106-5028, USA.
| |
Collapse
|
170
|
Fathke C, Wilson L, Shah K, Kim B, Hocking A, Moon R, Isik F. Wnt signaling induces epithelial differentiation during cutaneous wound healing. BMC Cell Biol 2006; 7:4. [PMID: 16426441 PMCID: PMC1388211 DOI: 10.1186/1471-2121-7-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Accepted: 01/20/2006] [Indexed: 12/23/2022] Open
Abstract
Background Cutaneous wound repair in adult mammals does not regenerate the original epithelial architecture and results in altered skin function. We propose that lack of regeneration may be due to the absence of appropriate molecular signals to promote regeneration. In this study, we investigated the regulation of Wnt signaling during cutaneous wound healing and the consequence of activating either the beta-catenin-dependent or beta-catenin-independent Wnt signaling on epidermal architecture during wound repair. Results We determined that the expression of Wnt ligands that typically signal via the beta-catenin-independent pathway is up-regulated in the wound while the beta-catenin-dependent Wnt signaling is activated in the hair follicles adjacent to the wound edge. Ectopic activation of beta-catenin-dependent Wnt signaling with lithium chloride in the wound resulted in epithelial cysts and occasional rudimentary hair follicle structures within the epidermis. In contrast, forced expression of Wnt-5a in the deeper wound induced changes in the interfollicular epithelium mimicking regeneration, including formation of epithelia-lined cysts in the wound dermis, rudimentary hair follicles and sebaceous glands, without formation of tumors. Conclusion These findings suggest that adult interfollicular epithelium is capable of responding to Wnt morphogenic signals necessary for restoring epithelial tissue patterning in the skin during wound repair.
Collapse
Affiliation(s)
- Carrie Fathke
- Department of Surgery, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Lynne Wilson
- Department of Surgery, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Kavita Shah
- Howard Hughes Medical Institute/Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Brian Kim
- Department of Surgery, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Anne Hocking
- Department of Surgery, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Randall Moon
- Howard Hughes Medical Institute/Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Frank Isik
- Department of Surgery, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
171
|
Kirilly D, Spana EP, Perrimon N, Padgett RW, Xie T. BMP signaling is required for controlling somatic stem cell self-renewal in the Drosophila ovary. Dev Cell 2006; 9:651-62. [PMID: 16256740 DOI: 10.1016/j.devcel.2005.09.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2004] [Revised: 08/12/2005] [Accepted: 09/20/2005] [Indexed: 01/22/2023]
Abstract
BMP signaling is essential for promoting self-renewal of mouse embryonic stem cells and Drosophila germline stem cells and for repressing stem cell proliferation in the mouse intestine and skin. However, it remains unknown whether BMP signaling can promote self-renewal of adult somatic stem cells. In this study, we show that BMP signaling is necessary and sufficient for promoting self-renewal and proliferation of somatic stem cells (SSCs) in the Drosophila ovary. BMP signaling is required in SSCs to directly control their maintenance and division, but is dispensable for proliferation of their differentiated progeny. Furthermore, BMP signaling is required to control SSC self-renewal, but not survival. Moreover, constitutive BMP signaling prolongs the SSC lifespan. Therefore, our study clearly demonstrates that BMP signaling directly promotes SSC self-renewal and proliferation in the Drosophila ovary. Our work further suggests that BMP signaling could promote self-renewal of adult stem cells in other systems.
Collapse
Affiliation(s)
- Daniel Kirilly
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, Missouri 64110, USA
| | | | | | | | | |
Collapse
|
172
|
Abstract
Despite intensive research, many longstanding questions of experimental hematology remain unsolved. One major reason is the fact that hematopoiesis is usually followed by analyzing populations of cells rather than individual cells, at few points in time during an experiment and without knowing (or quickly loosing) the cells' individual identities. The static picture yielded by this approach makes it impossible to appreciate the dynamic developmental processes leading to the generation of the full hematopoietic system from individual hematopoietic stem cells (HSCs). Real-time tracking of individual cells in culture, tissues, or whole organisms would be an extremely powerful approach to fully understand the developmental complexity of hematopoiesis. To this end, a computer-aided culture and bioimaging system is being developed to follow the fate of individual cells over long periods of time. This system is used to follow the development of multilineage cobblestone colonies from adult HSCs in stroma cocultures at the single cell level over many generations. To facilitate noninvasive detection of lineage commitment in these cultures, new subcellular forms of optimized fluorescent proteins have been developed to allow simultaneous marking of multiple hematopoietic lineages within the same animal.
Collapse
Affiliation(s)
- Timm Schroeder
- Institute of Stem Cell Research, GSF-National Research Center for Environment and Health, Ingolstaedter Landstr. 1, D-85764 Neuherberg, Germany.
| |
Collapse
|
173
|
Das AV, Zhao X, James J, Kim M, Cowan KH, Ahmad I. Neural stem cells in the adult ciliary epithelium express GFAP and are regulated by Wnt signaling. Biochem Biophys Res Commun 2006; 339:708-16. [PMID: 16332461 DOI: 10.1016/j.bbrc.2005.11.064] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2005] [Accepted: 11/09/2005] [Indexed: 12/28/2022]
Abstract
The identification of neural stem cells with retinal potential in the ciliary epithelium (CE) of the adult mammals is of considerable interest because of their potential for replacing or rescuing degenerating retinal neurons in disease or injury. The evaluation of such a potential requires characterization of these cells with regard to their phenotypic properties, potential, and regulatory mechanisms. Here, we demonstrate that rat CE stem cells/progenitors in neurosphere culture display astrocytic nature in terms of expressing glial intermediate neurofilament protein, GFAP. The GFAP-expressing CE stem cells/progenitors form neurospheres in proliferating conditions and generate neurons when shifted to differentiating conditions. These cells express components of the canonical Wnt pathway and its activation promotes their proliferation. Furthermore, we demonstrate that the activation of the canonical Wnt pathway influences neuronal differentiation of CE stem cells/progenitors in a context dependent manner. Our observations suggest that CE stem cells/progenitors share phenotypic properties and regulatory mechanism(s) with neural stem cells elsewhere in the adult CNS.
Collapse
Affiliation(s)
- Ani V Das
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198-5840, USA
| | | | | | | | | | | |
Collapse
|
174
|
Itami S, Inui S. Role of androgen in mesenchymal epithelial interactions in human hair follicle. J Investig Dermatol Symp Proc 2005; 10:209-11. [PMID: 16382666 DOI: 10.1111/j.1087-0024.2005.10107.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Human hair follicles, which are distributed in various and specific sites of the body, appear to have an inherited susceptibility for androgen-dependent growth. Beard, axillary, and frontal scalp dermal papilla cells (DPC) were recently shown to possess the characteristics of androgen target cells. These DPC show strong expression of androgen receptors, and the expression of type II 5alpha reductase is restricted to beard and frontal scalp DPC. These findings suggest that DPC mediate the signals of androgen to follicular epithelial cells in a paracrine fashion. We developed an in vitro co-culture system using DPC and keratinocytes (KC) to characterize the mode of androgen action in human hair follicles. Androgen significantly stimulated the proliferation of KC co-cultured with beard DPC, indicating that beard DPC produce androgen-dependent diffusible growth factors. Insulin-like growth factor-I was identified as one of the androgen-dependent paracrine growth factors produced by beard DPC. We also identified the inhibitory role of androgen on the growth of KC co-cultured with DPC from androgenetic alopecia (AGA) when the DPC were transfected with an expression vector encoding the androgen receptor. This growth suppression of KC was mediated by transforming growth factor-beta1 (TGF-beta1) derived from DPC of AGA, suggesting that TGF-beta1 is a paracrine mediator for AGA.
Collapse
Affiliation(s)
- Satoshi Itami
- Department of Dermatology, Course of Molecular Medicine, Graduate School of Medicine, Osaka University, Japan.
| | | |
Collapse
|
175
|
Wagner RA, Tabibiazar R, Liao A, Quertermous T. Genome-wide expression dynamics during mouse embryonic development reveal similarities to Drosophila development. Dev Biol 2005; 288:595-611. [PMID: 16310180 DOI: 10.1016/j.ydbio.2005.09.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2005] [Revised: 07/16/2005] [Accepted: 09/01/2005] [Indexed: 11/16/2022]
Abstract
Gene transcription mediates many vital aspects of mammalian embryonic development. A comprehensive characterization and analysis of the dynamics of gene transcription in the embryo is therefore likely to provide significant insights into the basic mechanisms of this process. We used microarrays to map transcription in the mouse embryo in the important period from embryonic day 8 (e8.0) to postnatal day 1 (p1) during which the bulk of the differentiation and development of organ systems takes place. Analysis of these expression profiles revealed distinct patterns of gene expression which correlate with the differentiation of organs including the nervous system, liver, skin, lungs, and digestive system, among others. Statistical analysis of the data based on Gene Ontology (GO) group annotation showed that specific temporal sequence patterns in gene class utilization across development are very similar to patterns seen during the embryonic development of Drosophila, suggesting conservation of the temporal progression of these processes across 550 million years of evolution. The temporal profiles of gene expression and activation of processes revealed here provide intriguing insights into the mechanisms of mammalian development, embryogenesis, and organogenesis, as well as into the evolution of developmental processes.
Collapse
Affiliation(s)
- Roger A Wagner
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Falk CVRC, 300 Pasteur Drive, Stanford, CA 94305, USA.
| | | | | | | |
Collapse
|
176
|
Das AV, Edakkot S, Thoreson WB, James J, Bhattacharya S, Ahmad I. Membrane properties of retinal stem cells/progenitors. Prog Retin Eye Res 2005; 24:663-81. [PMID: 15939659 DOI: 10.1016/j.preteyeres.2005.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The membrane properties of cells help integrate extrinsic information relayed through growth factors, chemokines, extracellular matrix, gap junctions and neurotransmitters towards modulating cell-intrinsic properties, which in turn determine whether cells remain quiescent, proliferate, differentiate, establish contact with other cells or remove themselves by activating programmed cell death. This review highlights some of the membrane properties of early and late retinal stem cells/progenitors, which are likely to be helpful in the identification and enrichment of these cells and in understanding mechanisms underlying their maintenance and differentiation. Understanding of membrane properties of retinal stem cells/progenitors is essential for the successful formulation of approaches to treat retinal degeneration and diseases by cell therapy.
Collapse
Affiliation(s)
- Ani V Das
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198-7691, USA
| | | | | | | | | | | |
Collapse
|
177
|
Abstract
The identification of adult epidermal stem cells that are capable of self-renewal and can reconstitute not only the epidermis but also the cutaneous appendages opens new perspectives for the treatment of a variety of human skin disorders including severe burns, cutaneous cancers, alopecia and acne. However, the implementation and improvement of these novel treatment strategies require a better understanding of the biology of stem cells, in particular regarding their isolation and the maintenance of their unique characteristics in culture. In this review, we summarize the main features of epidermal stem cells and we present the most recent advances in our understanding of the development and maintenance of these cells. In addition, we discuss some of the challenges and the potential clinical applications of epidermal stem cell technology.
Collapse
Affiliation(s)
- R Barthel
- INSERM U634 Faculté de Médecine, Avenue Valombrose, 06107 Nice cedex, France
| | | |
Collapse
|
178
|
Selleri S, Arnaboldi F, Palazzo M, Hussein U, Balsari A, Rumio C. Caveolin-1 is expressed on multipotent cells of hair follicles and might be involved in their resistance to chemotherapy. Br J Dermatol 2005; 153:506-13. [PMID: 16120134 DOI: 10.1111/j.1365-2133.2005.06746.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Caveolin-1 is the principal protein that composes caveolae, which are vesicular invaginations present on the plasma membrane of different cell types. Caveolae are involved in a variety of cellular functions including regulation of proliferation rate and resistance to chemotherapeutic drugs. Chemotherapy frequently induces alopecia which is reversible most probably due to the low proliferative rate of hair follicle stem cells and due to the expression of proteins which confer resistance. OBJECTIVES Using a specific animal model and immunohistochemistry, we analysed the expression of both caveolin-1 and the cell proliferation marker beta-catenin, at different stages of the hair follicle cycle, both before and after doxorubicin (DXR) -induced alopecia. METHODS Seven-week-old C57BL/6 mice were depilated in order to synchronize hair follicle cycle in the anagen phase. Chemotherapy with DXR 15 mg kg(-1) was used to induce alopecia. Control and treated mice were then sacrificed at precise time points and caveolin-1 expression in hairs at different stages of the cycle were analysed by immunohistochemistry. By double immunofluorescence, colocalization of caveolin-1 and cytokeratin-15 was confirmed in the bulge region. The state of proliferation of cells composing hair follicle was assessed by beta-catenin immunohistochemistry. RESULTS Caveolin-1 was expressed by the cells of the bulge area, the multipotent compartment of the hair follicle, during all phases of growth (anagen), regression (catagen) and resting (telogen). During the anagen phases, nuclear beta-catenin labelling was not observed in bulge cells, but rather in the deeper portion of the follicle. Damaged hair follicles from DXR-treated mice presented bulge cells which still expressed caveolin-1, suggesting that this protein might play a role in their drug resistance. As expected, no beta-catenin nuclear staining was detectable in DXR-treated hair follicles, indicating the complete lack of proliferative processes. The differential localization of caveolin-1 and beta-catenin suggests that the mutually exclusive expression of these proteins is useful for correct hair regrowth, whether during the physiological cycle or after chemotherapy-induced alopecia. CONCLUSIONS Expression of caveolin-1 within the multipotent cell compartment of the hair follicle can explain the resistance of bulge cells to many chemotherapeutics, suggested by the reversibility of chemotherapy-induced alopecia.
Collapse
Affiliation(s)
- S Selleri
- Department of Human Morphology, National Cancer Institute, Università degli Studi di Milano, Via Mangiagalli, 31, 20133 Milan, Italy
| | | | | | | | | | | |
Collapse
|
179
|
Miller SJ, Lavker RM, Sun TT. Interpreting epithelial cancer biology in the context of stem cells: tumor properties and therapeutic implications. Biochim Biophys Acta Rev Cancer 2005; 1756:25-52. [PMID: 16139432 DOI: 10.1016/j.bbcan.2005.07.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2005] [Revised: 07/12/2005] [Accepted: 07/15/2005] [Indexed: 12/17/2022]
Abstract
Over 90% of all human neoplasia is derived from epithelia. Significant progress has been made in the identification of stem cells of many epithelia. In general, epithelial stem cells lack differentiation markers, have superior in vivo and in vitro proliferative potential, form clusters in association with a specialized mesenchymal environment (the 'niche'), are located in well-protected and nourished sites, and are slow-cycling and thus can be experimentally identified as 'label-retaining cells'. Stem cells may divide symmetrically giving rise to two identical stem cell progeny. Any stem cells in the niche, which defines the size of the stem cell pool, may be randomly expelled from the niche due to population pressure (the stochastic model). Alternatively, a stem cell may divide asymmetrically yielding one stem cell and one non-stem cell that is destined to exit from the stem cell niche (asymmetric division model). Stem cells separated from their niche lose their stemness, although such a loss may be reversible, becoming 'transit-amplifying cells' that are rapidly proliferating but have a more limited proliferative potential, and can give rise to terminally differentiated cells. The identification of the stem cell subpopulation in a normal epithelium leads to a better understanding of many previously enigmatic properties of an epithelium including the preferential sites of carcinoma formation, as exemplified by the almost exclusive association of corneal epithelial carcinoma with the limbus, the corneal epithelial stem cell zone. Being long-term residents in an epithelium, stem cells are uniquely susceptible to the accumulation of multiple, oncogenic changes giving rise to tumors. The application of the stem cell concept can explain many important carcinoma features including the clonal origin and heterogeneity of tumors, the occasional formation of tumors from the transit amplifying cells or progenitor cells, the formation of precancerous 'patches' and 'fields', the mesenchymal influence on carcinoma formation and behavior, and the plasticity of tumor cells. While the concept of cancer stem cells is extremely useful and it is generally assumed that such cells are derived from normal stem cells, more work is needed to identify and characterize epithelial cancer stem cells, to address their precise relationship with normal stem cells, to study their markers and their proliferative and differentiation properties and to design new therapies that can overcome their unusual resistance to chemotherapy and other conventional tumor modalities.
Collapse
Affiliation(s)
- Stanley J Miller
- Department of Dermatology, Johns Hopkins Hospital, Baltimore, MD 21287, USA.
| | | | | |
Collapse
|
180
|
Swain RK, Katoh M, Medina A, Steinbeisser H. Xenopus frizzled-4S, a splicing variant of Xfz4 is a context-dependent activator and inhibitor of Wnt/beta-catenin signaling. Cell Commun Signal 2005; 3:12. [PMID: 16236168 PMCID: PMC1291380 DOI: 10.1186/1478-811x-3-12] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2005] [Accepted: 10/19/2005] [Indexed: 11/10/2022] Open
Abstract
Background Secreted Frizzled related proteins (SFRPs) are extracellular regulators of Wnt signaling. These proteins contain an N-terminal cysteine rich domain (CRD) highly similar to the CRDs of the Frizzled family of seven-transmembrane proteins that act as Wnt receptors. SFRPs can bind to Wnts and prevent their interaction with the Frizzled receptor. Recently it has been reported that a splice variant of human Frizzled-4 (FZD4S) lacking the transmembrane and the cytoplasmic domains of Frizzled-4 can activate rather than inhibit Wnt-8 activity in Xenopus embryos. This indicates that secreted CRD containing proteins such as Frizzled ecto-domains and SFRPs may not always act as Wnt inhibitors. It is not known how FZD4S can activate Wnt/β-catenin signaling and what biological role this molecule plays in vivo. Results Here we report that the Xenopus frizzled-4 is alternatively spliced to give rise to a putative secreted protein that lacks the seven-transmembrane and the cytoplasmic domains. We performed functional experiments in Xenopus embryos to investigate how this novel splicing variant, Xfz4S, can modulate the Wnt/β-catenin pathway. We show that Xfz4S as well as the extracellular domain of Xfz8 (ECD8) can act as both activators and inhibitors of Wnt/β-catenin signaling dependent on the Wnt ligand presented. The positive regulation of Wnt/β-catenin signaling by the extracellular domains of Frizzled receptors is mediated by the members of low density lipoprotein receptor-related protein (LRP-5/6) that act as Wnt coreceptors. Conclusion This work provides evidence that the secreted extracellular domains of Frizzled receptors may act as both inhibitors and activators of Wnt signaling dependent on the Wnt ligand presented.
Collapse
Affiliation(s)
- Rajeeb Kumar Swain
- Institute of Human Genetics, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Masaru Katoh
- Genetics and Cell Biology Section, Genetics Division, National Cancer Center Research Institute, Tsukiji 5-chome, Chuo-ku, Tokyo, 104-0045, Japan
| | - Araceli Medina
- Deptartment of Cell Biology, Max Planck Institute for Developmental Biology, Spemann Str. 35, 72076 Tübingen, Germany
| | - Herbert Steinbeisser
- Institute of Human Genetics, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| |
Collapse
|
181
|
Zhao H, Cui Y, Dupont J, Sun H, Hennighausen L, Yakar S. Overexpression of the tumor suppressor gene phosphatase and tensin homologue partially inhibits wnt-1-induced mammary tumorigenesis. Cancer Res 2005; 65:6864-73. [PMID: 16061670 DOI: 10.1158/0008-5472.can-05-0181] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The tumor suppressor phosphatase and tensin homologue (PTEN) is involved in cell proliferation, adhesion, and apoptosis. PTEN overexpression in mammary epithelium leads to reduced cell number and impaired differentiation and secretion. In contrast, overexpression of the proto-oncogene Wnt-1 in mammary epithelium leads to mammary hyperplasia and subsequently focal mammary tumors. To explore the possibility that PTEN intersects with Wnt-induced tumorigenesis, mice that ectopically express PTEN and Wnt-1 in mammary epithelium were generated. PTEN overexpression resulted in an 11% reduction of Wnt-1-induced tumors within a 12-month period and the onset of tumors was delayed from an average of 5.9 to 7.7 months. The rate of tumor growth, measured from 0.5 cm diameter until the tumors reached 1.0 cm diameter, was increased from 8.4 days in Wnt-1 mice to 17.7 days in Wnt-1 mice overexpressing PTEN. Here we show for the first time in vivo that overexpression of PTEN in the Wnt-1 transgenic mice resulted in a marked decrease in the insulin-like growth factor (IGF)-I receptor levels leading to a reduced IGF-I-mediated mitogenesis. Moreover, the percentage of BrdUrd-positive epithelial nuclei was decreased by 48%. beta-Catenin immunoreactivity was significantly decreased and the percentage of signal transducer and activator of transcription 5a (stat5a)-positive mammary epithelial cells was increased by 2-fold in Wnt-1 mice overexpressing PTEN. The present study shows that PTEN can partially inhibit the Wnt-1-induced mammary tumorigenesis in early neoplastic stages by blocking the AKT pathway and by reducing the IGF-I receptor levels in mammary gland. This study identifies the PTEN as a therapeutic target for the treatment of mammary cancer and presumably other types of cancer.
Collapse
MESH Headings
- Animals
- Cadherins/metabolism
- Cell Growth Processes/physiology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cyclin D1/biosynthesis
- Cytoskeletal Proteins/metabolism
- DNA-Binding Proteins/biosynthesis
- Down-Regulation
- Female
- Hyperplasia
- Intercellular Signaling Peptides and Proteins/biosynthesis
- Intercellular Signaling Peptides and Proteins/genetics
- Male
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/prevention & control
- Matrix Metalloproteinase 9/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Milk Proteins/biosynthesis
- PTEN Phosphohydrolase
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 1
- Protein Tyrosine Phosphatases/biosynthesis
- Protein Tyrosine Phosphatases/genetics
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt
- Receptor, IGF Type 1/metabolism
- STAT5 Transcription Factor
- Signal Transduction
- Trans-Activators/biosynthesis
- Trans-Activators/metabolism
- Transgenes
- Tumor Suppressor Proteins/biosynthesis
- Tumor Suppressor Proteins/genetics
- Wnt Proteins
- Wnt1 Protein
- beta Catenin
Collapse
Affiliation(s)
- Hong Zhao
- Diabetes Branch and Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland 20892-1758, USA
| | | | | | | | | | | |
Collapse
|
182
|
Wang Q, Liu Y, Han C. The molecular mechanism of embryonic stem cell pluripotency maintenance. CHINESE SCIENCE BULLETIN-CHINESE 2005. [DOI: 10.1007/bf03182658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
183
|
Beaudoin GMJ, Sisk JM, Coulombe PA, Thompson CC. Hairless triggers reactivation of hair growth by promoting Wnt signaling. Proc Natl Acad Sci U S A 2005; 102:14653-8. [PMID: 16195376 PMCID: PMC1253615 DOI: 10.1073/pnas.0507609102] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The mammalian hair cycle involves periodic regeneration of a tiny organ, the hair follicle, through a stem-cell-mediated process. The Hairless (Hr) gene encodes a nuclear receptor corepressor (HR) that is essential for hair follicle regeneration, but its role in this process is unknown. Here, we demonstrate that transgenic expression of HR in progenitor keratinocytes rescues follicle regeneration in Hr(-/-) mice. We show that expression of Wise, a modulator of Wnt signaling, is repressed by HR in these cells, coincident with the timing of follicle regeneration. This work links HR and Wnt function, providing a model in which HR regulates the precise timing of Wnt signaling required for hair follicle regeneration.
Collapse
Affiliation(s)
- Gerard M J Beaudoin
- Kennedy Krieger Research Institute and Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
184
|
Sarin KY, Cheung P, Gilison D, Lee E, Tennen RI, Wang E, Artandi MK, Oro AE, Artandi SE. Conditional telomerase induction causes proliferation of hair follicle stem cells. Nature 2005; 436:1048-52. [PMID: 16107853 PMCID: PMC1361120 DOI: 10.1038/nature03836] [Citation(s) in RCA: 325] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2005] [Accepted: 05/06/2005] [Indexed: 12/17/2022]
Abstract
TERT, the protein component of telomerase, serves to maintain telomere function through the de novo addition of telomere repeats to chromosome ends, and is reactivated in 90% of human cancers. In normal tissues, TERT is expressed in stem cells and in progenitor cells, but its role in these compartments is not fully understood. Here we show that conditional transgenic induction of TERT in mouse skin epithelium causes a rapid transition from telogen (the resting phase of the hair follicle cycle) to anagen (the active phase), thereby facilitating robust hair growth. TERT overexpression promotes this developmental transition by causing proliferation of quiescent, multipotent stem cells in the hair follicle bulge region. This new function for TERT does not require the telomerase RNA component, which encodes the template for telomere addition, and therefore operates through a mechanism independent of its activity in synthesizing telomere repeats. These data indicate that, in addition to its established role in extending telomeres, TERT can promote proliferation of resting stem cells through a non-canonical pathway.
Collapse
Affiliation(s)
- Kavita Y. Sarin
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, CA 94305
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
| | - Peggie Cheung
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, CA 94305
| | - Daniel Gilison
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, CA 94305
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
| | - Eunice Lee
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, CA 94305
| | - Ruth I. Tennen
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, CA 94305
- Cancer Biology Program, Stanford School of Medicine, Stanford, CA 94305
| | - Estee Wang
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, CA 94305
| | - Maja K. Artandi
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, CA 94305
| | - Anthony E. Oro
- Cancer Biology Program, Stanford School of Medicine, Stanford, CA 94305
- Department of Dermatology, Stanford School of Medicine, Stanford, CA 94305
| | - Steven E. Artandi
- Department of Medicine, Division of Hematology, Stanford School of Medicine, Stanford, CA 94305
- Cancer Biology Program, Stanford School of Medicine, Stanford, CA 94305
- Correspondence and requests should be addressed to S.E.A. ()
| |
Collapse
|
185
|
Okuse T, Chiba T, Katsuumi I, Imai K. Differential expression and localization of WNTs in an animal model of skin wound healing. Wound Repair Regen 2005; 13:491-7. [PMID: 16176457 DOI: 10.1111/j.1067-1927.2005.00069.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Wound healing is a dynamic process, and a variety of growth factors have a significant impact on the process. Although the WNT family has a multitude of effects on the state of various physiological pathways, the expression and role of WNT in wounded tissue have remained an enigma. The aim of this study was to assess the expression and localization of WNTs in a murine model of wound healing. RNA isolated from full-thickness cutaneous wounds from day 1 to day 21 postwounding were subjected to reverse transcription-polymerase chain reaction, and expression of WNT3, 4, 5a, and 10b were observed. Immunohistochemistry localized WNT10b to regenerating epithelial cells on day 1 and 3, and WNT4 on day 3 and 5. WNT4 also reacted with fibroblast-like cells beneath the epithelium. The cytoplasmic staining of beta-catenin, a WNT signaling molecule, in the epithelial cells indicates an activation of the WNT signaling pathway. Among target genes downstream of the pathway, matrix metalloproteinases (MMPs) degrade and remodel the extracellular matrix during wound healing. Gelatin zymography showed that MMP9 was expressed from day 1 to day 5. MMP-2 was continuously expressed, but maximally up-regulated at day 5. Activation of MMP-2 coincided with expression of membrane-type 1 MMP, suggesting an involvement of WNTs in this proteolytic cascade. Therefore, WNTs may contribute to the process of wound healing in a spatiotemporal manner.
Collapse
Affiliation(s)
- Toshiyuki Okuse
- The Department of Endodontics and Operative Dentistry, School of Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan
| | | | | | | |
Collapse
|
186
|
Stojadinovic O, Brem H, Vouthounis C, Lee B, Fallon J, Stallcup M, Merchant A, Galiano RD, Tomic-Canic M. Molecular pathogenesis of chronic wounds: the role of beta-catenin and c-myc in the inhibition of epithelialization and wound healing. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 167:59-69. [PMID: 15972952 PMCID: PMC1603435 DOI: 10.1016/s0002-9440(10)62953-7] [Citation(s) in RCA: 259] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Lack of understanding of the molecular mechanisms and pathogenesis of impaired healing in chronic ulcers is a serious health issue that contributes to excessive limb amputations and mortality. Here we show that beta-catenin and its downstream targets in keratinocytes, c-myc, and keratins K6 and K16, play important roles in the development of chronic wounds. In contrast to normal epidermis, we observed a significant nuclear presence of beta-catenin and elevated c-myc expression at the nonhealing wound edge of chronic ulcers from 10 patients. In vitro studies indicated that stabilization of nuclear beta-catenin inhibited wound healing and keratinocyte migration by blocking epidermal growth factor response, inducing c-myc and repressing the K6/K16 keratins (cytoskeletal components important for migration). The molecular mechanism of K6/K16 repression involved beta-catenin and arginine methyltransferase (CARM-1) acting as co-repressors of glucocorticoid receptor monomers. We conclude that activation of the beta-catenin/c-myc pathway(s) contributes to impaired healing by inhibiting keratinocyte migration and altering their differentiation. The presence of activated beta-catenin and c-myc in the epidermis of chronic wounds may serve as a molecular marker of impaired healing and may provide future targets for therapeutic intervention.
Collapse
Affiliation(s)
- Olivera Stojadinovic
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, 550 First Ave., TH100, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Legué E, Nicolas JF. Hair follicle renewal: organization of stem cells in the matrix and the role of stereotyped lineages and behaviors. Development 2005; 132:4143-54. [PMID: 16107474 DOI: 10.1242/dev.01975] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hair follicles (HFs) are renewed via multipotent stem cells located in a reservoir (the bulge); however, little is known about how they generate multi-tissue HFs from a proliferative zone (the matrix). To address this issue, we temporally induced clonal labeling during HF growth. Challenging the prevailing hypothesis, we found that the matrix contains restricted self-renewing stem cells for each inner structure. These cells are located around the dermal papilla forming a germinative layer. They occupy different proximodistal sectors and produce differentiated cells along the matrix radial axis via stereotyped lineages and cell behavior. By contrast, the outer layer of HFs displays a mode of growth involving apoptosis that coordinates the development of outer and inner structures. HF morphology is therefore determined by the organization of cell fates along the proximodistal axis and by cell behavior along the radial (lateral) axis in the matrix. Thus, our studies suggest that fate and behavior are organized by two systems (uncoupled), and this uncoupling may represent a fundamental way to simplify morphogenesis.
Collapse
Affiliation(s)
- Emilie Legué
- Unité de Biologie moléculaire du Développement, Institut Pasteur, 25 rue du Docteur Roux, 75724 Paris Cedex 15, France
| | | |
Collapse
|
188
|
Murphy MJ, Wilson A, Trumpp A. More than just proliferation: Myc function in stem cells. Trends Cell Biol 2005; 15:128-37. [PMID: 15752976 DOI: 10.1016/j.tcb.2005.01.008] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Adult stem cells are essential to maintain regenerative tissues such as skin epidermis, gastrointestinal mucosa or the hematopoietic system. Recent studies in mice suggest that the transcription factor and oncoprotein c-Myc has unexpected functions during both self-renewal and the differentiation of stem and early progenitor cells, particularly in interactions between stem cells and the local microenvironment or "niche". By incorporating recent findings on Myc and hematopoietic stem cells we propose a model in which "resting" hematopoietic stem cells are "activated" to self-renew and to differentiate at the interface between the niche and non-niche microenvironments.
Collapse
Affiliation(s)
- Mark J Murphy
- Genetics and Stem Cell Laboratory; Swiss Institute for Experimental Cancer Research (ISREC), Chemin des Boveresses 155, CH-1066 Epalinges, Switzerland
| | | | | |
Collapse
|
189
|
Dravid G, Ye Z, Hammond H, Chen G, Pyle A, Donovan P, Yu X, Cheng L. Defining the role of Wnt/beta-catenin signaling in the survival, proliferation, and self-renewal of human embryonic stem cells. Stem Cells 2005; 23:1489-501. [PMID: 16002782 DOI: 10.1634/stemcells.2005-0034] [Citation(s) in RCA: 257] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We used a panel of human and mouse fibroblasts with various abilities for supporting the prolonged growth of human embryonic stem cells (hESCs) to elucidate growth factors required for hESC survival, proliferation, and maintenance of the undifferentiated and pluripotent state (self-renewal). We found that supportive feeder cells secrete growth factors required for both hESC survival/proliferation and blocking hESC spontaneous differentiation to achieve self-renewal. The antidifferentiation soluble factor is neither leukemia inhibitory factor nor Wnt, based on blocking experiments using their antagonists. Because Wnt/beta-catenin signaling has been implicated in cell-fate determination and stem cell expansion, we further examined the effects of blocking or adding recombinant Wnt proteins on undifferentiated hESCs. In the absence of feeder cell-derived factors, hESCs cultured under a feeder-free condition survived/proliferated poorly and gradually differentiated. Adding recombinant Wnt3a stimulated hESC proliferation but also differentiation. After 4-5 days of Wnt3a treatment, hESCs that survived maintained the undifferentiated phenotype but few could form undifferentiated hESC colonies subsequently. Using a functional reporter assay, we found that the beta-catenin-mediated transcriptional activation in the canonical Wnt pathway was minimal in undifferentiated hESCs, but greatly upregulated during differentiation induced by the Wnt treatment and several other methods. Thus, Wnt/beta-catenin activation does not suffice to maintain the undifferentiated and pluripotent state of hESCs. We propose a new model for the role of Wnt/beta-catenin signaling in undifferentiated hESCs.
Collapse
Affiliation(s)
- Gautam Dravid
- The Institute for Cell Engineering, Department of Gynecology & Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | |
Collapse
|
190
|
Faraldo MM, Teulière J, Deugnier MA, Taddei-De La Hosseraye I, Thiery JP, Glukhova MA. Myoepithelial cells in the control of mammary development and tumorigenesis: data from genetically modified mice. J Mammary Gland Biol Neoplasia 2005; 10:211-9. [PMID: 16807801 DOI: 10.1007/s10911-005-9582-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Until recently, myoepithelial cells-the second major cell population in the mammary epithelium-were not considered to play an important role in the morphogenetic events during gland development. Mouse mutants with changes in the gene expression pattern characteristic of the basal myoepithelial cell layer have been generated and used to show that these cells influence the proliferation, survival and differentiation of luminal cells, modulate stromal-epithelial interactions and actively participate in mammary morphogenesis. Various cellular and molecular mechanisms may underlie the observed phenotypes. These include an unbalanced expression of matrix degrading metalloproteinases (MMPs) and their inhibitors, leading to changes in the composition and organization of the (extracellular matrix) ECM, the production of soluble growth factors affecting stromal and epithelial cell growth and differentiation and direct signaling through cell-cell contacts between the myoepithelial and luminal cell layers.
Collapse
Affiliation(s)
- Marisa M Faraldo
- Institut Curie, CNRS UMR144, 26 rue d'Ulm, 75248, Paris Cedex 05, France
| | | | | | | | | | | |
Collapse
|
191
|
Zhang X, Podsypanina K, Huang S, Mohsin SK, Chamness GC, Hatsell S, Cowin P, Schiff R, Li Y. Estrogen receptor positivity in mammary tumors of Wnt-1 transgenic mice is influenced by collaborating oncogenic mutations. Oncogene 2005; 24:4220-31. [PMID: 15824740 DOI: 10.1038/sj.onc.1208597] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The majority (75%) of human breast cancers express estrogen receptor (ER). Although ER-positive tumors usually respond to antiestrogen therapies, 30% of them do not. It is not known what controls the ER status of breast cancers or their responsiveness to antihormone interventions. In this report, we document that transgenic (TG) expression of Wnt-1 in mice induces ER-positive tumors. Loss of Pten or gain of Ras mutations during the evolution of tumors in Wnt-1 TG mice has no effect on the expression of ER, but overexpression of Neu or loss of p53 leads to ER-negative tumors. Thus, our results provide compelling evidence that expression of ER in breast cancer may be influenced by specific genetic changes that promote cancer progression. These findings constitute a first step to explore the molecular mechanisms leading to ER-positive or ER-negative mammary tumors. In addition, we find that ER-positive tumors arising in Wnt-1 TG mice are refractory to both ovariectomy and the ER antagonist tamoxifen, but lose ER expression with tamoxifen, suggesting that antiestrogen selects for ER-negative tumor cells and that the ER-positive cell fraction is dispensable for growth of these tumors. This is a first report of a mouse model of antiestrogen-resistant ER-positive breast cancers, and could provide a powerful tool to study the molecular mechanisms that control antiestrogen resistance.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Hormonal/pharmacology
- Breast Neoplasms/genetics
- Disease Models, Animal
- Drug Resistance, Neoplasm
- Gene Expression Regulation, Neoplastic
- Genes, p53
- Humans
- Immunohistochemistry
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/pharmacology
- Mammary Neoplasms, Animal/genetics
- Mice
- Mice, Transgenic
- Mitogens
- Ovariectomy/veterinary
- PTEN Phosphohydrolase
- Phosphoric Monoester Hydrolases/genetics
- Protein-Tyrosine Kinases
- Receptor, ErbB-2/genetics
- Receptors, Estrogen/biosynthesis
- Receptors, Estrogen/physiology
- Signal Transduction
- Tamoxifen/pharmacology
- Tumor Suppressor Proteins/genetics
- Wnt Proteins
- Wnt1 Protein
Collapse
Affiliation(s)
- Xiaomei Zhang
- Breast Center, Baylor College of Medicine, One Baylor Plaza, N1210.03, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Byun T, Karimi M, Marsh JL, Milovanovic T, Lin F, Holcombe RF. Expression of secreted Wnt antagonists in gastrointestinal tissues: potential role in stem cell homeostasis. J Clin Pathol 2005; 58:515-9. [PMID: 15858124 PMCID: PMC1770654 DOI: 10.1136/jcp.2004.018598] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Wnt signalling dysregulation has been implicated in cancer, including colon and gastric cancer. Initiation of Wnt signalling is modulated by soluble Wnt antagonists (sWAs), including soluble frizzled related proteins, dickkopf (Dkk) proteins, and Wnt inhibitory factor-1 (Wif1). AIMS To evaluate the role of sWAs in upper (gastric) and lower (colon) gastrointestinal tract tumorigenesis. METHODS Dkk1-3, Wif1, and FrzB expression was evaluated by in situ RNA hybridisation on normal and malignant human gastric and colon tissues. Expression was graded semiquantitatively. RESULTS Wif1, Dkk1, and Dkk2 were not expressed in normal gastric tissue. Dkk3 was expressed in some samples, with stronger expression in deep gastric glands. FrzB was expressed in several normal gastric samples, but not in matched tumour specimens. In contrast, Dkk1 and FrzB were not expressed in normal colon. Wif1 was expressed in most colon samples, with stronger expression at crypt bases. Dkk3 and Dkk2 expression was also concentrated at crypt bases. There were no differences between sWA expression in malignant colon and matched normal tissue. CONCLUSIONS sWA expression differed between upper and lower gastrointestinal tract. The loss of FrzB in gastric cancer suggests that it acts as a tumour suppressor. The graded expression of Dkk3 in gastric tissue, and Dkk2, Dkk3, and Wif1 in colon tissue, with increased expression in the deep gastric glands/colonic crypt bases, where gastrointestinal stem cells reside, suggests that sWAs may be crucial Wnt signalling regulators in these tissues, and may contribute to stem cell pool maintenance. sWAs are important components of the gastrointestinal proliferative regulatory network.
Collapse
Affiliation(s)
- T Byun
- Division of Hematology/Oncology, University of California, Irvine Medical Center, 101 The City Drive, Bld 23, Rm 244, Orange, CA 92868, USA
| | | | | | | | | | | |
Collapse
|
193
|
Morasso MI, Tomic-Canic M. Epidermal stem cells: the cradle of epidermal determination, differentiation and wound healing. Biol Cell 2005; 97:173-83. [PMID: 15715523 PMCID: PMC1283090 DOI: 10.1042/bc20040098] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The field of epidermal stem cells has dramatically advanced in the last decade, leading to a better understanding of the molecular factors, signalling pathways and cellular events that identify and characterize stem cells, thus revealing their immense potential for therapeutic use. Furthermore, multipotent epidermal stem cells present the major advantage of easy accessibility with the discovery of their specific location within the bulge of the hair follicle. This review focuses on the most recent findings on epidermal stem cells, and their potential role in initial epidermal commitment, differentiation and wound healing processes in the skin.
Collapse
Affiliation(s)
- Maria I Morasso
- Developmental Skin Biology Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
194
|
Abstract
Wnt and Hedgehog (Hh) signalling regulate stem-cell self-renewal and differentiation in a range of epithelia and the inappropriate activation of these pathways contributes to epithelial cancers. Recently, it was reported that Indian Hedgehog (Ihh) antagonises Wnt signalling in colonic epithelium. This observation contrasts with other reports of positive synergy between the pathways and challenges the view that systemically administered Hedgehog antagonists could be beneficial for the treatment of intestinal tumours. The work is discussed in the broader context of Ihh expression and function in epithelia and the different ways in which the Hh and Wnt pathways interact.
Collapse
Affiliation(s)
- Fiona M Watt
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, UK.
| |
Collapse
|
195
|
Ciani L, Salinas PC. WNTs in the vertebrate nervous system: from patterning to neuronal connectivity. Nat Rev Neurosci 2005; 6:351-62. [PMID: 15832199 DOI: 10.1038/nrn1665] [Citation(s) in RCA: 488] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
WNT signalling has a key role in early embryonic patterning through the regulation of cell fate decisions, tissue polarity and cell movements. In the nervous system, WNT signalling also regulates neuronal connectivity by controlling axon pathfinding, axon remodelling, dendrite morphogenesis and synapse formation. Studies, from invertebrates to mammals, have led to a considerable understanding of WNT signal transduction pathways. This knowledge provides a framework for the study of the mechanisms by which WNTs regulate diverse neuronal functions. Manipulation of the WNT pathways could provide new strategies for nerve regeneration and neuronal circuit modulation.
Collapse
Affiliation(s)
- Lorenza Ciani
- Department of Anatomy and Developmental Biology, University College London, London WC1E 6BT, UK
| | | |
Collapse
|
196
|
Abstract
Canonical Wnt signaling plays a crucial role in controlling cell expansion in many types of stem cells. Recent studies, however, demonstrated that Wnt is not only a general stem cell growth factor but can also influence cell lineage decisions in certain stem cell types by promoting specific fates at the expense of others. Thus, Wnt signaling elicits multiple functions in stem cells. Wnt activity appears to depend on cell-intrinsic properties that might change with time during development, thereby altering the cellular response to Wnt. Moreover, the spatial context of a stem cell also determines how the cell interprets Wnt signal activity, in that synergistic or antagonistic signaling pathways can modulate Wnt signaling. How a stem cell integrates Wnt and other signals and how such signaling networks regulate stem cell function on the molecular level remains to be elucidated.
Collapse
Affiliation(s)
- Maurice Kléber
- Institute of Cell Biology, Department of Biology, Swiss Federal Institute of Technology, ETH Hönggerberg, CH-8093 Zurich, Switzerland.
| | | |
Collapse
|
197
|
Abstract
To what extent are the pathways that regulate self-renewal conserved between stem cells at different stages of development and in different tissues? Some pathways play a strikingly conserved role in regulating the self-renewal of diverse stem cells, whereas other pathways are specific to stem cells in certain tissues or at certain stages of development. Recent studies have highlighted differences between the self-renewal of embryonic, fetal and adult stem cells. By understanding these similarities and differences we may come to a molecular understanding of how stem cells replicate themselves and why aspects of this process differ between stem cells.
Collapse
Affiliation(s)
- Anna V Molofsky
- Howard Hughes Medical Institute, and Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, 3215 CCGC, Ann Arbor, Michigan 48109-0934, USA
| | | | | |
Collapse
|
198
|
Boland GM, Perkins G, Hall DJ, Tuan RS. Wnt 3a promotes proliferation and suppresses osteogenic differentiation of adult human mesenchymal stem cells. J Cell Biochem 2005; 93:1210-30. [PMID: 15486964 DOI: 10.1002/jcb.20284] [Citation(s) in RCA: 440] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multipotential adult mesenchymal stem cells (MSCs) are able to differentiate along several known lineages, and lineage commitment is tightly regulated through specific cellular mediators and interactions. Recent observations of a low/high bone-mass phenotype in patients expressing a loss-/gain-of-function mutation in LRP5, a coreceptor of the Wnt family of signaling molecules, suggest the importance of Wnt signaling in bone formation, possibly involving MSCs. To analyze the role of Wnt signaling in mesenchymal osteogenesis, we have profiled the expression of WNTs and their receptors, FRIZZLEDs (FZDs), and several secreted Wnt inhibitors, such as SFRPs, and examined the effect of Wnt 3a, as a representative canonical Wnt member, during MSC osteogenesis in vitro. WNT11, FZD6, SFRP2, and SFRP3 are upregulated during MSC osteogenesis, while WNT9A and FZD7 are downregulated. MSCs also respond to exogenous Wnt 3a, based on increased beta-catenin nuclearization and activation of a Wnt-responsive promoter, and the magnitude of this response depends on the MSC differentiation state. Wnt 3a exposure inhibits MSC osteogenic differentiation, with decreased matrix mineralization and reduced alkaline phosphatase mRNA and activity. Wnt 3a treatment of fully osteogenically differentiated MSCs also suppresses osteoblastic marker gene expression. The Wnt 3a effect is accompanied by increased cell number, resulting from both increased proliferation and decreased apoptosis, particularly during expansion of undifferentiated MSCs. The osteo-suppressive effects of Wnt 3a are fully reversible, i.e., treatment prior to osteogenic induction does not compromise subsequent MSC osteogenesis. The results also showed that sFRP3 treatment attenuates some of the observed Wnt 3a effects on MSCs, and that inhibition of canonical Wnt signaling using a dominant negative TCF1 enhances MSC osteogenesis. Interestingly, expression of Wnt 5a, a non-canonical Wnt member, appeared to promote osteogenesis. Taken together, these findings suggest that canonical Wnt signaling functions in maintaining an undifferentiated, proliferating progenitor MSC population, whereas non-canonical Wnts facilitate osteogenic differentiation. Release from canonical Wnt regulation is a prerequisite for MSC differentiation. Thus, loss-/gain-of-function mutations of LRP5 would perturb Wnt signaling and depress/promote bone formation by affecting the progenitor cell pool. Elucidating Wnt regulation of MSC differentiation is important for their potential application in tissue regeneration.
Collapse
Affiliation(s)
- Genevieve M Boland
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892-8022, USA
| | | | | | | |
Collapse
|
199
|
Li B, Mackay DR, Ma J, Dai X. Cloning and developmental expression of mouse pygopus 2, a putative Wnt signaling component. Genomics 2005; 84:398-405. [PMID: 15234002 PMCID: PMC2893388 DOI: 10.1016/j.ygeno.2004.04.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2003] [Accepted: 04/14/2004] [Indexed: 11/29/2022]
Abstract
Recent studies in Drosophila identified pygopus, which encodes a PHD finger protein, as an additional nuclear component of the canonical Wingless(Wg)/Wnt signaling pathway. In this study, we describe the molecular cloning and expression analysis of a mouse pygopus gene, mpygo2. mpygo2 transcripts were detected in almost all adult mouse tissues examined, whereas transcripts of another mouse pygopus gene, mpygo1, were detected only in heart tissue. Abundant mpygo2 transcripts were observed during embryogenesis in multiple developmental sites. Consistent with the demonstrated role of the Wnt-beta-catenin-LEF/TCF signaling pathway in mammalian skin development, mpygo2 expression was detected in the developing epidermis and hair follicles, which suggests that mpygo2 might mediate the effect of this signaling pathway in mouse skin.
Collapse
Affiliation(s)
| | | | | | - Xing Dai
- Corresponding author. Department of Biological Chemistry, College of Medicine, D234 Med Sci I, University of California, Irvine, CA 92697-1700; Fax: 949-824-2688. (X. Dai)
| |
Collapse
|
200
|
Bamberger C, Hafner A, Schmale H, Werner S. Expression of different p63 variants in healing skin wounds suggests a role of p63 in reepithelialization and muscle repair. Wound Repair Regen 2005; 13:41-50. [PMID: 15659035 DOI: 10.1111/j.1067-1927.2005.130106.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Healing of skin wounds in mammals involves partial reconstruction of the dermis and coverage of the injured site by keratinocytes. The latter process is achieved by extensive migration and hyperproliferation of keratinocytes at the wound rim. Because the p53 protein family member p63 is expressed in human hyperproliferative epidermis, this study determined whether enhanced keratinocyte proliferation correlates with the expression of p63. Therefore, we investigated the temporal and spatial distribution of four major variants of the p63 transcription factor-TAp63alpha, TAp63gamma, DeltaNp63alpha and DeltaNp63gamma-during normal skin wound healing in mice. Transcripts encoding amino-terminally truncated DeltaNp63 variants were found at high levels in basal and suprabasal keratinocytes of the hyperproliferative wound epithelium. Interestingly, TAp63 variants, which include the conserved transactivation domain TA at their amino-terminus, were also expressed in wound keratinocytes as well as at the edge of the injured subcutaneous muscle panniculus carnosus. These findings suggest splice-variant specific functions of p63 in reepithelialization and muscle repair.
Collapse
Affiliation(s)
- Casimir Bamberger
- Institute of Cell Biology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| | | | | | | |
Collapse
|