1
|
Zhang M, Zhang L, Liu J, Zhao J, Mei J, Zou J, Luo Y, Cai C. Mammary stem cells: molecular cues, orchestrated regulatory mechanisms and its implications in breast cancer. J Genet Genomics 2025:S1673-8527(25)00116-X. [PMID: 40254157 DOI: 10.1016/j.jgg.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/22/2025]
Abstract
Mammary stem cells (MaSCs), endowed with self-renewal and multilineage differentiation capabilities, are crucial for mammary gland development, function, and disease initiation. Recent advances in MaSCs biology research encompass molecular marker identification, regulatory pathway dissection, and microenvironmental crosstalk. This review synthesizes key progress and remaining challenges in MaSC research. Molecular profiling advances have identified key markers recently, such as Procr, Dll1, Bcl11b, and PD-L1. Central to their regulatory logic are evolutionarily conserved pathways, including Wnt, Notch, Hedgehog, and Hippo, which exhibit context-dependent thresholds to balance self-renewal and differentiation. Beyond intrinsic signaling, the dynamic interplay between MaSCs and their microenvironment, such as luminal-derived Wnt4, macrophage-mediated TNF-α signaling, and adrenergic inputs from sympathetic nerves, spatially orchestrates stem cell behavior. In addition, this review also discusses the roles of breast cancer stem cells (BCSCs) in tumorigenesis and therapeutic resistance, focusing on the molecular mechanisms underlying MaSC transformation into BCSCs. Despite progress, challenges remain: human MaSCs functional assays lack standardization, pathway inhibitors risk off-target effects, and delivery systems lack precision. Emerging tools like spatial multi-omics, organoids, and biomimetic scaffolds address these gaps. By integrating MaSCs and BCSCs biology, this review links mechanisms to breast cancer and outlines strategies to target malignancy to accelerate clinical translation.
Collapse
Affiliation(s)
- Mengna Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Lingxian Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Jie Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Jiahui Zhao
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Jiayu Mei
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Jiahua Zou
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Yaogan Luo
- Mengniu Institute of Nutrition Science, Shanghai 200124, China
| | - Cheguo Cai
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China.
| |
Collapse
|
2
|
Chen W, Guo L, Wei W, Cai C, Wu G. Zdhhc1- and Zdhhc2-mediated Gpm6a palmitoylation is essential for maintenance of mammary stem cell activity. Cell Rep 2024; 43:114762. [PMID: 39321020 DOI: 10.1016/j.celrep.2024.114762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/28/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024] Open
Abstract
Adult mammary stem cells (aMaSCs) are vital to tissue expansion and remodeling during the process of postnatal mammary development. The protein C receptor (Procr) is one of the well-identified surface markers of multipotent aMaSCs. However, an understanding of the regulatory mechanisms governing Procr's protein stability remains incomplete. In this study, we identified Glycoprotein m6a (Gpm6a) as a critical protein for aMaSC activity modulation by using the Gpm6a knockout mouse model. Interestingly, we determined that Gpm6a depletion results in a reduction of Procr protein stability. Mechanistically, Gpm6a regulates Procr protein stability by mediating the formation of lipid rafts, a process requiring Zdhhc1 and Zdhhc2 to palmitate Gpm6a at Cys17,18 and Cys246 sites. Our findings highlight an important mechanism involving Zdhhc1- and Zdhhc2-mediated Gpm6a palmitoylation for the regulation of Procr stability, aMaSC activity, and postnatal mammary development.
Collapse
Affiliation(s)
- Weizhen Chen
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan 430071, China
| | - Luyao Guo
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Wei Wei
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Cheguo Cai
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan 430071, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, College of Life Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
3
|
Yang X, Xu H, Yang X, Wang H, Zou L, Yang Q, Qi X, Li L, Duan H, Yan X, Fu NY, Tan J, Hou Z, Jiao B. Mcam inhibits macrophage-mediated development of mammary gland through non-canonical Wnt signaling. Nat Commun 2024; 15:36. [PMID: 38167296 PMCID: PMC10761817 DOI: 10.1038/s41467-023-44338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
While canonical Wnt signaling is well recognized for its crucial regulatory functions in cell fate decisions, the role of non-canonical Wnt signaling in adult stem cells remains elusive and contradictory. Here, we identified Mcam, a potential member of the non-canonical Wnt signaling, as an important negative regulator of mammary gland epithelial cells (MECs) by genome-scale CRISPR-Cas9 knockout (GeCKO) library screening. Loss of Mcam increases the clonogenicity and regenerative capacity of MECs, and promotes the proliferation, differentiation, and ductal morphogenesis of mammary epithelial in knockout mice. Mechanically, Mcam knockout recruits and polarizes macrophages through the Il4-Stat6 axis, thereby promoting secretion of the non-canonical Wnt ligand Wnt5a and its binding to the non-canonical Wnt signaling receptor Ryk to induce the above phenotypes. These findings reveal Mcam roles in mammary gland development by orchestrating communications between MECs and macrophages via a Wnt5a/Ryk axis, providing evidences for non-canonical Wnt signaling in mammary development.
Collapse
Affiliation(s)
- Xing Yang
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, 650051, China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, 650051, China
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Haibo Xu
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Xu Yang
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Hui Wang
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Li Zou
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Qin Yang
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Xiaopeng Qi
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Li Li
- Research Center of Stem cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Hongxia Duan
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100000, China
| | - Xiyun Yan
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100000, China
| | - Nai Yang Fu
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, 169857, Singapore
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Jing Tan
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, 650051, China.
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, 650051, China.
| | - Zongliu Hou
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, 650051, China.
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, 650051, China.
| | - Baowei Jiao
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650201, China.
| |
Collapse
|
4
|
Jacob JB, Wei KC, Bepler G, Reyes JD, Cani A, Polin L, White K, Kim S, Viola N, McGrath J, Guastella A, Yin C, Mi QS, Kidder BL, Wagner KU, Ratner S, Phillips V, Xiu J, Parajuli P, Wei WZ. Identification of actionable targets for breast cancer intervention using a diversity outbred mouse model. iScience 2023; 26:106320. [PMID: 36968078 PMCID: PMC10034465 DOI: 10.1016/j.isci.2023.106320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/16/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
HER2-targeted therapy has improved breast cancer survival, but treatment resistance and disease prevention remain major challenges. Genes that enable HER2/Neu oncogenesis are the next intervention targets. A bioinformatics discovery platform of HER2/Neu-expressing Diversity Outbred (DO) F1 Mice was established to identify cancer-enabling genes. Quantitative Trait Loci (QTL) associated with onset ages and growth rates of spontaneous mammary tumors were sought. Twenty-six genes in 3 QTL contain sequence variations unique to the genetic backgrounds that are linked to aggressive tumors and 21 genes are associated with human breast cancer survival. Concurrent identification of TSC22D3, a transcription factor, and its target gene LILRB4, a myeloid cell checkpoint receptor, suggests an immune axis for regulation, or intervention, of disease. We also investigated TIEG1 gene that impedes tumor immunity but suppresses tumor growth. Although not an actionable target, TIEG1 study revealed genetic regulation of tumor progression, forming the basis of the genetics-based discovery platform.
Collapse
Affiliation(s)
- Jennifer B. Jacob
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kuang-Chung Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Gerold Bepler
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Joyce D. Reyes
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Andi Cani
- Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lisa Polin
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kathryn White
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Seongho Kim
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Nerissa Viola
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Julie McGrath
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - Anthony Guastella
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - CongCong Yin
- Department of Immunology, Henry Ford Health System, Detroit, MI48202, USA
| | - Qing-Shen Mi
- Department of Immunology, Henry Ford Health System, Detroit, MI48202, USA
| | - Benjamin L. Kidder
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kay-Uwe Wagner
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Stuart Ratner
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Victoria Phillips
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Joanne Xiu
- Clinical and Translational Research, Caris Life Sciences, Irving, TX75039, USA
| | - Prahlad Parajuli
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Wei-Zen Wei
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| |
Collapse
|
5
|
Lin MJ, Lu CPJ. Glandular stem cells in the skin during development, homeostasis, wound repair and regeneration. Exp Dermatol 2021; 30:598-604. [PMID: 33686662 DOI: 10.1111/exd.14319] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023]
Abstract
Glands in the skin are essential for various physiological functions involving exocrine secretion. Like other tissues and organs, they possess the ability to repair injury and self-renew during homeostasis. Progenitor cells in glands are mostly unipotent but include some multipotent stem cells that function when extensive remodelling or regeneration is required. In this review, using two glandular models in skin, mouse sweat gland and mammary gland, we discuss lineage restriction that develops during glandular morphogenesis, as well as the mechanisms regulating cell fate and plasticity during wound repair and regeneration. Understanding the intrinsic and extrinsic factors that control the behaviours of glandular stem cell and maintain glandular functions will provide insight into future prospects for glandular regeneration.
Collapse
Affiliation(s)
- Meng-Ju Lin
- The Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, NY, USA
| | - Catherine Pei-Ju Lu
- The Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, NY, USA.,Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
6
|
Kuehn J, Espinoza-Sanchez NA, Teixeira FCOB, Pavão MSG, Kiesel L, Győrffy B, Greve B, Götte M. Prognostic significance of hedgehog signaling network-related gene expression in breast cancer patients. J Cell Biochem 2021; 122:577-597. [PMID: 33417295 DOI: 10.1002/jcb.29886] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/19/2020] [Accepted: 12/22/2020] [Indexed: 12/25/2022]
Abstract
Breast cancer continues to be a serious public health problem. The role of the hedgehog pathway in normal development of the mammary gland as well as in carcinogenesis and progression of breast cancer is the subject of intense investigation, revealing functional interactions with cell surface heparan sulfate. Nevertheless, its influence on breast cancer prognosis, and its relation to specific sulfation motifs in heparan sulfate have only been poorly studied in large patient cohorts. Using the public database KMplotter that includes gene expression and survival data of 3951 patients, we found that the higher expression of SHH, HHAT, PTCH1, GLI1, GLI2, and GLI3 positively influences breast cancer prognosis. Stratifying patients according to the expression of hormone receptors, histological grade, lymph node metastasis, and systemic therapy, we observed that GLI1, GLI2, and GLI3 expression, as well as co-expression of SHH and ELP1 were associated with worse relapse-free survival in patients with HER2-positive tumors. Moreover, GLI1 expression in progesterone receptor-negative tumors and GLI3 expression in grade 3 tumors correlated with poor prognosis. SHH, in a panel of cell lines representing different breast cancer subtypes, and HHAT, PTCH1, GLI1, GLI2, and GLI3 were mostly expressed in cell lines classified as HER2-positive and basal-like. Expression of SHH, HHAT, GLI2, and GLI3 was differentially affected by overexpression of the heparan sulfate sulfotransferases HS2ST1 and HS3ST2 in vitro. Although high HS2ST1 expression was associated with poor prognosis in KMplotter analysis, high levels of HS3ST2 were associated with a good prognosis, except for ER-positive breast cancer. We suggest the GLI transcription factors as possible markers for the diagnosis, treatment, and prognosis of breast cancer especially in HER2-positive tumors, but also in progesterone receptor-negative and grade-3 tumors. The pathway interaction and prognostic impact of specific heparan sulfate sulfotransferases provide novel perspectives regarding a therapeutical targeting of the hedgehog pathway in breast cancer.
Collapse
Affiliation(s)
- Julia Kuehn
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Nancy Adriana Espinoza-Sanchez
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.,Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - Felipe C O B Teixeira
- Instituto de Bioquímica Médica Leopoldo de Meis, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mauro S G Pavão
- Instituto de Bioquímica Médica Leopoldo de Meis, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, and Semmelweis University 2nd Department of Pediatrics, TTK Momentum Cancer Biomarker Research Group, Budapest, Hungary
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| |
Collapse
|
7
|
Oliphant MUJ, Kong D, Zhou H, Lewis MT, Ford HL. Two Sides of the Same Coin: The Role of Developmental pathways and pluripotency factors in normal mammary stem cells and breast cancer metastasis. J Mammary Gland Biol Neoplasia 2020; 25:85-102. [PMID: 32323111 PMCID: PMC7395869 DOI: 10.1007/s10911-020-09449-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Breast cancer initiation and progression are often observed as the result of dysregulation of normal developmental processes and pathways. Studies focused on normal mammary stem/progenitor cell activity have led to an understanding of how breast cancer cells acquire stemness-associated properties including tumor initiation, survival and multi-lineage differentiation into heterogeneous tumors that become difficult to target therapeutically. Importantly, more recent investigations have provided valuable insight into how key developmental regulators can impact multiple phases of metastasis, where they are repurposed to not only promote metastatic phenotypes such as migration, invasion and EMT at the primary site, but also to regulate the survival, initiation and maintenance of metastatic lesions at secondary organs. Herein, we discuss findings that have led to a better understanding of how embryonic and pluripotency factors contribute not only to normal mammary development, but also to metastatic progression. We further examine the therapeutic potential of targeting these developmental pathways, and discuss how a better understanding of compensatory mechanisms, crosstalk between pathways, and novel experimental models could provide critical insight into how we might exploit embryonic and pluripotency regulators to inhibit tumor progression and metastasis.
Collapse
Affiliation(s)
- M U J Oliphant
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Department of Cell Biology and Ludwig Center at Harvard, Harvard Medical School, 240 Longwood Avenue, Building C1, Room 513B, Boston, MA, 02115, USA
| | - Deguang Kong
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
| | - Hengbo Zhou
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
| | - M T Lewis
- Departments of Molecular and Cellular Biology and Radiology. Lester and Sue Smith Breast Center, Baylor College of Medicine. One Baylor Plaza BCM600, Room N1210, Houston, TX, 77030, USA
| | - H L Ford
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
| |
Collapse
|
8
|
Landua JD, Moraes R, Carpenter EM, Lewis MT. Hoxd10 Is Required Systemically for Secretory Activation in Lactation and Interacts Genetically with Hoxd9. J Mammary Gland Biol Neoplasia 2020; 25:145-162. [PMID: 32705545 PMCID: PMC7392944 DOI: 10.1007/s10911-020-09454-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/01/2020] [Indexed: 11/24/2022] Open
Abstract
Targeted disruption of the murine Hoxd10 gene (ΔHoxd10) leads to a high frequency of localized (gland-to-gland or regionally within a gland) lactation impairment in homozygous mutant mice as a single gene mutation. The effect of Hoxd10 disruption was enhanced by simultaneous disruption of Hoxd9 (ΔHoxd9/d10), a mutation shown previously to have no effect on mammary function as a single gene alteration. Mammary glands of homozygous ΔHoxd10 and ΔHoxd9/d10 females were indistinguishable from those of wild type littermate and age-matched control mice in late pregnancy. However, in lactation, 47% of homozygous ΔHoxd10 females, and 100% of homozygous ΔHoxd9/d10 females, showed localized or complete failure of two or more glands to undergo lactation-associated morphological changes and to secrete milk. Affected regions of ΔHoxd10 and ΔHoxd9/d10 mutants showed reduced prolactin receptor expression, reduced signal transducer and activator transcription protein 5 (STAT5) phosphorylation, reduced expression of downstream milk proteins, mislocalized glucose transporter 1 (GLUT1), increased STAT3 expression and phosphorylation, recruitment of leukocytes, altered cell cycle status, and increased apoptosis relative to unaffected regions and wild type control glands. Despite these local effects on alveolar function, transplantation results and hormone analysis indicate that Hoxd10 primarily has systemic functions that confer attenuated STAT5 phosphorylation on both wild type and ΔHoxd10 transplants when placed in ΔHoxd10 hosts, thereby exacerbating an underlying propensity for lactation failure in C57Bl/6 mice.
Collapse
Affiliation(s)
- John D Landua
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dan L Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Room N1210; BCM600, Houston, TX, 77030, USA
| | - Ricardo Moraes
- Center for Cell and Gene Therapy, Texas Children's Feigin Center, Baylor College of Medicine, 1102 Bates Avenue, Houston, TX, 77030, USA
| | - Ellen M Carpenter
- Division of Undergraduate Education, National Science Foundation, 2415 Eisenhower Avenue, Alexandria, VA, 22314, USA
| | - Michael T Lewis
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dan L Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Room N1210; BCM600, Houston, TX, 77030, USA.
| |
Collapse
|
9
|
Jeng KS, Chang CF, Lin SS. Sonic Hedgehog Signaling in Organogenesis, Tumors, and Tumor Microenvironments. Int J Mol Sci 2020; 21:ijms21030758. [PMID: 31979397 PMCID: PMC7037908 DOI: 10.3390/ijms21030758] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/20/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023] Open
Abstract
During mammalian embryonic development, primary cilia transduce and regulate several signaling pathways. Among the various pathways, Sonic hedgehog (SHH) is one of the most significant. SHH signaling remains quiescent in adult mammalian tissues. However, in multiple adult tissues, it becomes active during differentiation, proliferation, and maintenance. Moreover, aberrant activation of SHH signaling occurs in cancers of the skin, brain, liver, gallbladder, pancreas, stomach, colon, breast, lung, prostate, and hematological malignancies. Recent studies have shown that the tumor microenvironment or stroma could affect tumor development and metastasis. One hypothesis has been proposed, claiming that the pancreatic epithelia secretes SHH that is essential in establishing and regulating the pancreatic tumor microenvironment in promoting cancer progression. The SHH signaling pathway is also activated in the cancer stem cells (CSC) of several neoplasms. The self-renewal of CSC is regulated by the SHH/Smoothened receptor (SMO)/Glioma-associated oncogene homolog I (GLI) signaling pathway. Combined use of SHH signaling inhibitors and chemotherapy/radiation therapy/immunotherapy is therefore key in targeting CSCs.
Collapse
|
10
|
Fu NY, Nolan E, Lindeman GJ, Visvader JE. Stem Cells and the Differentiation Hierarchy in Mammary Gland Development. Physiol Rev 2019; 100:489-523. [PMID: 31539305 DOI: 10.1152/physrev.00040.2018] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mammary gland is a highly dynamic organ that undergoes profound changes within its epithelium during puberty and the reproductive cycle. These changes are fueled by dedicated stem and progenitor cells. Both short- and long-lived lineage-restricted progenitors have been identified in adult tissue as well as a small pool of multipotent mammary stem cells (MaSCs), reflecting intrinsic complexity within the epithelial hierarchy. While unipotent progenitor cells predominantly execute day-to-day homeostasis and postnatal morphogenesis during puberty and pregnancy, multipotent MaSCs have been implicated in coordinating alveologenesis and long-term ductal maintenance. Nonetheless, the multipotency of stem cells in the adult remains controversial. The advent of large-scale single-cell molecular profiling has revealed striking changes in the gene expression landscape through ontogeny and the presence of transient intermediate populations. An increasing number of lineage cell-fate determination factors and potential niche regulators have now been mapped along the hierarchy, with many implicated in breast carcinogenesis. The emerging diversity among stem and progenitor populations of the mammary epithelium is likely to underpin the heterogeneity that characterizes breast cancer.
Collapse
Affiliation(s)
- Nai Yang Fu
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Emma Nolan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Geoffrey J Lindeman
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jane E Visvader
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
11
|
Role of Hedgehog Signaling in Breast Cancer: Pathogenesis and Therapeutics. Cells 2019; 8:cells8040375. [PMID: 31027259 PMCID: PMC6523618 DOI: 10.3390/cells8040375] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is the leading cause of cancer-related mortality in women, only followed by lung cancer. Given the importance of BC in public health, it is essential to identify biomarkers to predict prognosis, predetermine drug resistance and provide treatment guidelines that include personalized targeted therapies. The Hedgehog (Hh) signaling pathway plays an essential role in embryonic development, tissue regeneration, and stem cell renewal. Several lines of evidence endorse the important role of canonical and non-canonical Hh signaling in BC. In this comprehensive review we discuss the role of Hh signaling in breast development and homeostasis and its contribution to tumorigenesis and progression of different subtypes of BC. We also examine the efficacy of agents targeting different components of the Hh pathway both in preclinical models and in clinical trials. The contribution of the Hh pathway in BC tumorigenesis and progression, its prognostic role, and its value as a therapeutic target vary according to the molecular, clinical, and histopathological characteristics of the BC patients. The evidence presented here highlights the relevance of the Hh signaling in BC, and suggest that this pathway is key for BC progression and metastasis.
Collapse
|
12
|
Gli Proteins: Regulation in Development and Cancer. Cells 2019; 8:cells8020147. [PMID: 30754706 PMCID: PMC6406693 DOI: 10.3390/cells8020147] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/29/2019] [Accepted: 02/02/2019] [Indexed: 12/18/2022] Open
Abstract
Gli proteins are transcriptional effectors of the Hedgehog signaling pathway. They play key roles in the development of many organs and tissues, and are deregulated in birth defects and cancer. We review the molecular mechanisms of Gli protein regulation in mammals, with special emphasis on posttranslational modifications and intracellular transport. We also discuss how Gli proteins interact with co-activators and co-repressors to fine-tune the expression of Hedgehog target genes. Finally, we provide an overview of the regulation of developmental processes and tissue regeneration by Gli proteins and discuss how these proteins are involved in cancer progression, both through canonical regulation via the Hedgehog pathway and through cross-talk with other signaling pathways.
Collapse
|
13
|
Cazet AS, Hui MN, Elsworth BL, Wu SZ, Roden D, Chan CL, Skhinas JN, Collot R, Yang J, Harvey K, Johan MZ, Cooper C, Nair R, Herrmann D, McFarland A, Deng N, Ruiz-Borrego M, Rojo F, Trigo JM, Bezares S, Caballero R, Lim E, Timpson P, O'Toole S, Watkins DN, Cox TR, Samuel MS, Martín M, Swarbrick A. Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer. Nat Commun 2018. [PMID: 30042390 DOI: 10.1038/s41467-018-05220-6.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The cellular and molecular basis of stromal cell recruitment, activation and crosstalk in carcinomas is poorly understood, limiting the development of targeted anti-stromal therapies. In mouse models of triple negative breast cancer (TNBC), Hedgehog ligand produced by neoplastic cells reprograms cancer-associated fibroblasts (CAFs) to provide a supportive niche for the acquisition of a chemo-resistant, cancer stem cell (CSC) phenotype via FGF5 expression and production of fibrillar collagen. Stromal treatment of patient-derived xenografts with smoothened inhibitors (SMOi) downregulates CSC markers expression and sensitizes tumors to docetaxel, leading to markedly improved survival and reduced metastatic burden. In the phase I clinical trial EDALINE, 3 of 12 patients with metastatic TNBC derived clinical benefit from combination therapy with the SMOi Sonidegib and docetaxel chemotherapy, with one patient experiencing a complete response. These studies identify Hedgehog signaling to CAFs as a novel mediator of CSC plasticity and an exciting new therapeutic target in TNBC.
Collapse
Affiliation(s)
- Aurélie S Cazet
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Mun N Hui
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,The Chris O' Brien Lifehouse, Camperdown, NSW, 2050, Australia.,Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Benjamin L Elsworth
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Bristol, BS8 2BN, UK
| | - Sunny Z Wu
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Daniel Roden
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Chia-Ling Chan
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - Joanna N Skhinas
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - Raphaël Collot
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - Jessica Yang
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - Kate Harvey
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - M Zahied Johan
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, 5000, Australia.,Faculty of Health Sciences, School of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Caroline Cooper
- Pathology Queensland and School of Medicine, University of Queensland, St Lucia, QLD, 4006, Australia
| | - Radhika Nair
- Rajiv Gandhi Centre for Biotechnology, Thycaud Post, Poojappura, Thiruvananthapuram, Kerala, 695014, India
| | - David Herrmann
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Andrea McFarland
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - Niantao Deng
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Manuel Ruiz-Borrego
- Department of Medical Oncology, Hospital Universitario Virgen del Rocío, 41013, Sevilla, Spain
| | - Federico Rojo
- Department of Pathology, Hospital Universitario Fundación Jiménez Díaz, 28040, Madrid, Spain
| | - José M Trigo
- Department of Medical Oncology, Hospital Clínico Universitario Virgen de la Victoria, IBIMA, 29010, Málaga, Spain
| | - Susana Bezares
- GEICAM, Spanish Breast Cancer Group, Madrid, 28703, Spain
| | | | - Elgene Lim
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia.,St Vincent's Hospital, 2010, Darlinghurst, NSW, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Sandra O'Toole
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - D Neil Watkins
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia.,St Vincent's Hospital, 2010, Darlinghurst, NSW, Australia
| | - Thomas R Cox
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Michael S Samuel
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, 5000, Australia.,Faculty of Health Sciences, School of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Miguel Martín
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, 28040, Madrid, Spain
| | - Alexander Swarbrick
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia. .,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia. .,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia.
| |
Collapse
|
14
|
Cazet AS, Hui MN, Elsworth BL, Wu SZ, Roden D, Chan CL, Skhinas JN, Collot R, Yang J, Harvey K, Johan MZ, Cooper C, Nair R, Herrmann D, McFarland A, Deng N, Ruiz-Borrego M, Rojo F, Trigo JM, Bezares S, Caballero R, Lim E, Timpson P, O'Toole S, Watkins DN, Cox TR, Samuel MS, Martín M, Swarbrick A. Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer. Nat Commun 2018; 9:2897. [PMID: 30042390 PMCID: PMC6057940 DOI: 10.1038/s41467-018-05220-6] [Citation(s) in RCA: 312] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 06/21/2018] [Indexed: 12/20/2022] Open
Abstract
The cellular and molecular basis of stromal cell recruitment, activation and crosstalk in carcinomas is poorly understood, limiting the development of targeted anti-stromal therapies. In mouse models of triple negative breast cancer (TNBC), Hedgehog ligand produced by neoplastic cells reprograms cancer-associated fibroblasts (CAFs) to provide a supportive niche for the acquisition of a chemo-resistant, cancer stem cell (CSC) phenotype via FGF5 expression and production of fibrillar collagen. Stromal treatment of patient-derived xenografts with smoothened inhibitors (SMOi) downregulates CSC markers expression and sensitizes tumors to docetaxel, leading to markedly improved survival and reduced metastatic burden. In the phase I clinical trial EDALINE, 3 of 12 patients with metastatic TNBC derived clinical benefit from combination therapy with the SMOi Sonidegib and docetaxel chemotherapy, with one patient experiencing a complete response. These studies identify Hedgehog signaling to CAFs as a novel mediator of CSC plasticity and an exciting new therapeutic target in TNBC. Stromal cell recruitment, activation and crosstalk with cancer cells is poorly understood. Here, the authors demonstrate that cancer cell-derived Hedgehog ligand triggers stromal remodeling that in turn induces a cancer-stem-cell like, drug-resistant phenotype of nearby cancer cells while treatment with smoothened inhibitors reverses these phenotypes.
Collapse
Affiliation(s)
- Aurélie S Cazet
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Mun N Hui
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,The Chris O' Brien Lifehouse, Camperdown, NSW, 2050, Australia.,Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Benjamin L Elsworth
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Bristol, BS8 2BN, UK
| | - Sunny Z Wu
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Daniel Roden
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Chia-Ling Chan
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - Joanna N Skhinas
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - Raphaël Collot
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - Jessica Yang
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - Kate Harvey
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - M Zahied Johan
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, 5000, Australia.,Faculty of Health Sciences, School of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Caroline Cooper
- Pathology Queensland and School of Medicine, University of Queensland, St Lucia, QLD, 4006, Australia
| | - Radhika Nair
- Rajiv Gandhi Centre for Biotechnology, Thycaud Post, Poojappura, Thiruvananthapuram, Kerala, 695014, India
| | - David Herrmann
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Andrea McFarland
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia
| | - Niantao Deng
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Manuel Ruiz-Borrego
- Department of Medical Oncology, Hospital Universitario Virgen del Rocío, 41013, Sevilla, Spain
| | - Federico Rojo
- Department of Pathology, Hospital Universitario Fundación Jiménez Díaz, 28040, Madrid, Spain
| | - José M Trigo
- Department of Medical Oncology, Hospital Clínico Universitario Virgen de la Victoria, IBIMA, 29010, Málaga, Spain
| | - Susana Bezares
- GEICAM, Spanish Breast Cancer Group, Madrid, 28703, Spain
| | | | - Elgene Lim
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia.,St Vincent's Hospital, 2010, Darlinghurst, NSW, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Sandra O'Toole
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - D Neil Watkins
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia.,St Vincent's Hospital, 2010, Darlinghurst, NSW, Australia
| | - Thomas R Cox
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia
| | - Michael S Samuel
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, 5000, Australia.,Faculty of Health Sciences, School of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Miguel Martín
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, 28040, Madrid, Spain
| | - Alexander Swarbrick
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia. .,The Kinghorn Cancer Centre, Darlinghurst, NSW, 2010, Australia. .,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, 2010, Australia.
| |
Collapse
|
15
|
Weaver SR, Hernandez LL. Could use of Selective Serotonin Reuptake Inhibitors During Lactation Cause Persistent Effects on Maternal Bone? J Mammary Gland Biol Neoplasia 2018; 23:5-25. [PMID: 29603039 DOI: 10.1007/s10911-018-9390-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/13/2018] [Indexed: 02/07/2023] Open
Abstract
The lactating mammary gland elegantly coordinates maternal homeostasis to provide calcium for milk. During lactation, the monoamine serotonin regulates the synthesis and release of various mammary gland-derived factors, such as parathyroid hormone-related protein (PTHrP), to stimulate bone resorption. Recent evidence suggests that bone mineral lost during prolonged lactation is not fully recovered following weaning, possibly putting women at increased risk of fracture or osteoporosis. Selective Serotonin Reuptake Inhibitor (SSRI) antidepressants have also been associated with reduced bone mineral density and increased fracture risk. Therefore, SSRI exposure while breastfeeding may exacerbate lactational bone loss, compromising long-term bone health. Through an examination of serotonin and calcium homeostasis during lactation, lactational bone turnover and post-weaning recovery of bone mineral, and the effect of peripartum depression and SSRI on the mammary gland and bone, this review will discuss the hypothesis that peripartum SSRI exposure causes persistent reductions in bone mineral density through mammary-derived PTHrP signaling with bone.
Collapse
Affiliation(s)
- Samantha R Weaver
- Endocrine and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Laura L Hernandez
- Department of Dairy Science, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
16
|
EMT programs promote basal mammary stem cell and tumor-initiating cell stemness by inducing primary ciliogenesis and Hedgehog signaling. Proc Natl Acad Sci U S A 2017; 114:E10532-E10539. [PMID: 29158396 DOI: 10.1073/pnas.1711534114] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tissue regeneration relies on adult stem cells (SCs) that possess the ability to self-renew and produce differentiating progeny. In an analogous manner, the development of certain carcinomas depends on a small subset of tumor cells, called "tumor-initiating cells" (TICs), with SC-like properties. Mammary SCs (MaSCs) reside in the basal compartment of the mammary epithelium, and their neoplastic counterparts, mammary TICs (MaTICs), are thought to serve as the TICs for the claudin-low subtype of breast cancer. MaSCs and MaTICs both use epithelial-mesenchymal transition (EMT) programs to acquire SC properties, but the mechanism(s) connecting EMT programs to stemness remain unclear. Here we show that this depends on primary cilia, which are nonmotile, cell-surface structures that serve as platforms for receiving cues and enable activation of various signaling pathways. We show that MaSC and MaTIC EMT programs induce primary cilia formation and Hedgehog (Hh) signaling, which has previously been implicated in both MaSC and MaTIC function. Moreover, ablation of these primary cilia is sufficient to repress Hh signaling, the stemness of MaSCs, and the tumor-forming potential of MaTICs. Together, our findings establish primary ciliogenesis and consequent Hh signaling as a key mechanism by which MaSC and MaTIC EMT programs promote stemness and thereby support mammary tissue outgrowth and tumors of basal origin.
Collapse
|
17
|
Monkkonen T, Lewis MT. New paradigms for the Hedgehog signaling network in mammary gland development and breast Cancer. Biochim Biophys Acta Rev Cancer 2017; 1868:315-332. [PMID: 28624497 PMCID: PMC5567999 DOI: 10.1016/j.bbcan.2017.06.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 12/12/2022]
Abstract
The Hedgehog signaling network regulates organogenesis, cell fate, proliferation, survival, and stem cell self-renewal in many mammalian tissues. Aberrant activation of the Hedgehog signaling network is present in ~25% of all cancers, including breast. Altered expression of Hedgehog network genes in the mammary gland can elicit phenotypes at many stages of development. However, synthesizing a cohesive mechanistic model of signaling at different stages of development has been difficult. Emerging data suggest that this difficulty is due, in part, to non-canonical and tissue compartment-specific (i.e., epithelial, versus stromal, versus systemic) functions of Hedgehog network components. With respect to systemic functions, Hedgehog network genes regulate development of endocrine organs that impinge on mammary gland development extrinsically. These new observations offer insight into previously conflicting data, and have bearing on the potential for anti-Hedgehog therapeutics in the treatment of breast cancer.
Collapse
Affiliation(s)
- Teresa Monkkonen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; University of California, San Francisco, Dept. of Pathology, 513 Parnassus Ave., San Francisco, CA 94118, USA
| | - Michael T Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Radiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
18
|
Zhao C, Cai S, Shin K, Lim A, Kalisky T, Lu WJ, Clarke MF, Beachy PA. Stromal Gli2 activity coordinates a niche signaling program for mammary epithelial stem cells. Science 2017; 356:science.aal3485. [PMID: 28280246 DOI: 10.1126/science.aal3485] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 03/01/2017] [Indexed: 12/14/2022]
Abstract
The stem cell niche is a complex local signaling microenvironment that sustains stem cell activity during organ maintenance and regeneration. The mammary gland niche must support its associated stem cells while also responding to systemic hormonal regulation that triggers pubertal changes. We find that Gli2, the major Hedgehog pathway transcriptional effector, acts within mouse mammary stromal cells to direct a hormone-responsive niche signaling program by activating expression of factors that regulate epithelial stem cells as well as receptors for the mammatrophic hormones estrogen and growth hormone. Whereas prior studies implicate stem cell defects in human disease, this work shows that niche dysfunction may also cause disease, with possible relevance for human disorders and in particular the breast growth pathogenesis associated with combined pituitary hormone deficiency.
Collapse
Affiliation(s)
- Chen Zhao
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shang Cai
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kunyoo Shin
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyumgbuk 37673, South Korea
| | - Agnes Lim
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tomer Kalisky
- Faculty of Engineering and Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Wan-Jin Lu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael F Clarke
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Philip A Beachy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA. .,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
19
|
Monkkonen T, Landua JD, Visbal AP, Lewis MT. Epithelial and non-epithelial Ptch1 play opposing roles to regulate proliferation and morphogenesis of the mouse mammary gland. Development 2017; 144:1317-1327. [PMID: 28275010 PMCID: PMC5399619 DOI: 10.1242/dev.140434] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 02/07/2017] [Indexed: 12/14/2022]
Abstract
Patched 1 (Ptch1) has epithelial, stromal and systemic roles in murine mammary gland organogenesis, yet specific functions remain undefined. Cre-recombinase-mediated Ptch1 ablation in mammary epithelium increased proliferation and branching, but did not phenocopy transgenic expression of activated smoothened (SmoM2). The epithelium showed no evidence of canonical hedgehog signaling, and hyperproliferation was not blocked by smoothened (SMO) inhibition, suggesting a non-canonical function of PTCH1. Consistent with this possibility, nuclear localization of cyclin B1 was increased. In non-epithelial cells, heterozygous Fsp-Cre-mediated Ptch1 ablation increased proliferation and branching, with dysplastic terminal end buds (TEB) and ducts. By contrast, homozygous Ptch1 ablation decreased proliferation and branching, producing stunted ducts filled with luminal cells showing altered ovarian hormone receptor expression. Whole-gland transplantation into wild-type hosts or estrogen/progesterone treatment rescued outgrowth and hormone receptor expression, but not the histological changes. Bone marrow transplantation failed to rescue outgrowth. Ducts of Fsp-Cre;Ptch1fl/fl mice were similar to Fsp-Cre;SmoM2 ducts, but Fsp-Cre;SmoM2 outgrowths were not stunted, suggesting that the histology might be mediated by Smo in the local stroma, with systemic Ptch1 required for ductal outgrowth and proper hormone receptor expression in the mammary epithelium. Summary: Systemic and tissue-specific depletion of patched 1 in epithelial and stromal compartments of the mammary gland defines functions in ductal patterning, proliferation and gene expression.
Collapse
Affiliation(s)
- Teresa Monkkonen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - John D Landua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Adriana P Visbal
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Michael T Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA .,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
20
|
Veltmaat JM. Prenatal Mammary Gland Development in the Mouse: Research Models and Techniques for Its Study from Past to Present. Methods Mol Biol 2017; 1501:21-76. [PMID: 27796947 DOI: 10.1007/978-1-4939-6475-8_2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mammary gland development starts during prenatal life, when at designated positions along the ventrolateral boundary of the embryonic or fetal trunk, surface ectodermal cells coalesce to form primordia for mammary glands, instead of differentiating into epidermis. With the wealth of genetically engineered mice available as research models, our understanding of the prenatal phase of mammary development has recently greatly advanced. This understanding includes the recognition of molecular and mechanistic parallels between prenatal and postnatal mammary morphogenesis and even tumorigenesis, much of which can moreover be extrapolated to human. This makes the murine embryonic mammary gland a useful model for a myriad of questions pertaining to normal and pathological breast development. Hence, unless indicated otherwise, this review describes embryonic mammary gland development in mouse only, and lists mouse models that have been examined for defects in embryonic mammary development. Techniques that originated in the field of developmental biology, such as explant culture and tissue recombination, were adapted specifically to research on the embryonic mammary gland. Detailed protocols for these techniques have recently been published elsewhere. This review describes how the development and adaptation of these techniques moved the field forward from insights on (comparative) morphogenesis of the embryonic mammary gland to the understanding of tissue and molecular interactions and their regulation of morphogenesis and functional development of the embryonic mammary gland. It is here furthermore illustrated how generic molecular biology and biochemistry techniques can be combined with these older, developmental biology techniques, to address relevant research questions. As such, this review should provide a solid starting point for those wishing to familiarize themselves with this fascinating and important subdomain of mammary gland biology, and guide them in designing a relevant research strategy.
Collapse
Affiliation(s)
- Jacqueline M Veltmaat
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.
| |
Collapse
|
21
|
Bussche L, Rauner G, Antonyak M, Syracuse B, McDowell M, Brown AMC, Cerione RA, Van de Walle GR. Microvesicle-mediated Wnt/β-Catenin Signaling Promotes Interspecies Mammary Stem/Progenitor Cell Growth. J Biol Chem 2016; 291:24390-24405. [PMID: 27733685 DOI: 10.1074/jbc.m116.726117] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 10/02/2016] [Indexed: 01/08/2023] Open
Abstract
Signaling mechanisms that regulate mammary stem/progenitor cell (MaSC) self-renewal are essential for developmental changes that occur in the mammary gland during pregnancy, lactation, and involution. We observed that equine MaSCs (eMaSCs) maintain their growth potential in culture for an indefinite period, whereas canine MaSCs (cMaSCs) lose their growth potential in long term cultures. We then used this system to investigate the role of microvesicles (MVs) in promoting self-renewal properties. We found that Wnt3a and Wnt1 were expressed at higher levels in MVs isolated from eMaSCs compared with those from cMaSCs. Furthermore, eMaSC-MVs were able to induce Wnt/β-catenin signaling in different target cells, including cMaSCs. Interestingly, the induction of Wnt/β-catenin signaling in cMaSCs was prolonged when using eMaSC-MVs compared with recombinant Wnt proteins, indicating that MVs are not only important for transport of Wnt proteins, but they also enhance their signaling activity. Finally, we demonstrate that the eMaSC-MVs-mediated activation of the Wnt/β-catenin signaling pathway in cMaSCs significantly improves the ability of cMaSCs to grow as mammospheres and, importantly, that this effect is abolished when eMaSC-MVs are treated with Wnt ligand inhibitors. This suggests that this novel form of intercellular communication plays an important role in self-renewal.
Collapse
Affiliation(s)
| | - Gat Rauner
- From the Baker Institute for Animal Health and
| | - Marc Antonyak
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853 and
| | | | | | - Anthony M C Brown
- the Department of Cell & Developmental Biology, Weill Cornell Medical College, New York, New York 10065
| | - Richard A Cerione
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853 and
| | | |
Collapse
|
22
|
Song L, Wang W, Liu D, Zhao Y, He J, Wang X, Dai Z, Zhang H, Li X. Targeting of sonic hedgehog-Gli signaling: A potential therapeutic target for patients with breast cancer. Oncol Lett 2016; 12:1027-1033. [PMID: 27446389 PMCID: PMC4950736 DOI: 10.3892/ol.2016.4722] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 02/05/2016] [Indexed: 11/06/2022] Open
Abstract
Breast cancer is the most common malignant cancer among women. The Hedgehog (Hh) signaling pathway serves a key role in malignant cancer cell growth and migration. However, little is known with regard to the specific function of the Hh signaling pathway in human breast cancer. The current study investigated the specific role of Hh signaling in the human breast cancer cell line MDA-MB-231. Expression of components of Shh-Gli signaling, as well as the Gli-responsive genes B-cell lymphoma 2 (Bcl-2) and cyclin D1, were investigated in MDA-MB-231 cells using western blotting. The effects of Shh-Gli signaling on MDA-MB-231 proliferation were analyzed by MTT assay. The role of E-cadherin in the epithelial-mesenchymal transition process was determined by western blot while matrix metalloproteinase (MMP)-9/MMP-2 secretion was studied by enzyme-linked immunosorbent assay. The results indicated that Shh-Gli signaling was activated in MDA-MB-231 cells, significantly enhancing cell viability. Overexpression of Gli positively regulated the transcription of Bcl-2 and cyclin D1 thereby regulating MDA-MB-231 cell proliferation and survival. Treatment of MDA-MB-231 cells with human sonic hedgehog, n-terminus for 72 h significantly reduced E-cadherin protein levels and enhanced secretion of MMP-9 and MMP-2. These findings suggest that Shh-Gli signaling is significantly activated in human breast cancer cells, and is accompanied by enhanced cell viability, proliferation and migration capacities.
Collapse
Affiliation(s)
- Lingqin Song
- Department of Tumor Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Weifeng Wang
- Department of Tumor Surgery, The First People's Hospital of Xianyang, Xianyang, Shaanxi 712000, P.R. China
| | - Di Liu
- Department of Tumor Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yang Zhao
- Department of Tumor Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jianjun He
- Department of Tumor Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xijing Wang
- Department of Tumor Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Zhijun Dai
- Department of Tumor Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Huimin Zhang
- Department of Tumor Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiao Li
- Department of Tumor Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
23
|
Lin CY, Barry-Holson KQ, Allison KH. Breast cancer stem cells: are we ready to go from bench to bedside? Histopathology 2015; 68:119-37. [DOI: 10.1111/his.12868] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Chieh-Yu Lin
- Department of Pathology; Stanford University; Stanford CA USA
| | | | | |
Collapse
|
24
|
Laporta J, Keil KP, Weaver SR, Cronick CM, Prichard AP, Crenshaw TD, Heyne GW, Vezina CM, Lipinski RJ, Hernandez LL. Serotonin regulates calcium homeostasis in lactation by epigenetic activation of hedgehog signaling. Mol Endocrinol 2014; 28:1866-74. [PMID: 25192038 DOI: 10.1210/me.2014-1204] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Calcium homeostasis during lactation is critical for maternal and neonatal health. We previously showed that nonneuronal/peripheral serotonin [5-hydroxytryptamine (5-HT)] causes the lactating mammary gland to synthesize and secrete PTHrP in an acute fashion. Here, using a mouse model, we found that genetic inactivation of tryptophan hydroxylase 1 (Tph1), which catalyzes the rate-limiting step in peripheral 5-HT synthesis, reduced circulating and mammary PTHrP expression, osteoclast activity, and maternal circulating calcium concentrations during the transition from pregnancy to lactation. Tph1 inactivation also reduced sonic hedgehog signaling in the mammary gland during lactation. Each of these deficiencies was rescued by daily injections of 5-hydroxy-L-tryptophan (an immediate precursor of 5-HT) to Tph1-deficient dams. We used immortalized mouse embryonic fibroblasts to demonstrate that 5-HT induces PTHrP through a sonic hedgehog-dependent signal transduction mechanism. We also found that 5-HT altered DNA methylation of the Shh gene locus, leading to transcriptional initiation at an alternate start site and formation of a variant transcript in mouse embryonic fibroblasts in vitro and in mammary tissue in vivo. These results support a new paradigm of 5-HT-mediated Shh regulation involving DNA methylation remodeling and promoter switching. In addition to having immediate implications for lactation biology, identification and characterization of a novel functional regulatory relationship between nonneuronal 5-HT, hedgehog signaling, and PTHrP offers new avenues for the study of these important factors in development and disease.
Collapse
Affiliation(s)
- Jimena Laporta
- Departments of Dairy Science (J.L., S.R.W., C.M.C., A.P.P., L.L.H.), Comparative Biosciences (K.P.K., G.W.H., C.M.V., R.J.L.), and Animal Science (T.D.C.), University of Wisconsin-Madison, Madison, Wisconsin 53706
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mitchell EH, Serra R. Normal mammary development and function in mice with Ift88 deleted in MMTV- and K14-Cre expressing cells. Cilia 2014; 3:4. [PMID: 24594320 PMCID: PMC3942223 DOI: 10.1186/2046-2530-3-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 02/14/2014] [Indexed: 12/21/2022] Open
Abstract
Background Primary cilia (PC) are non-motile microtubule based organelles present on almost every cell type and are known to serve as critical organizing centers for several signaling pathways crucial to embryonic and postnatal development. Alterations in the Hh pathway, the most studied signaling pathway regulated by PC, affect mammary gland development as well as maintenance of the stem and progenitor cell populations. Results We developed mouse models with deletion of PC in mammary luminal epithelial, basal epithelial, and stromal cells for evaluation of the function of PC in mammary development via MMTV-Cre, K14-Cre, and Prx1-Cre mediated deletion, respectively. The activity of Cre was confirmed using ROSA26 reporters. Mammary stem and progenitor cells were enriched through growth as mammospheres. Adenovirus-Cre mediated deletion of Ift88 was used to determine a role for PC in this population of cells. Disruption of Ift88 and PC were confirmed in using PCR and immunofluorescent methods. Prx1-Cre; Ift88Del mice demonstrated defects in terminal end buds during puberty. However, these Ift88Del glands exhibited typical terminal end bud formation as well as normal ductal histology when transplanted into wild type hosts, indicating that the phenotype observed was not intrinsic to the mammary gland. Furthermore, no discernable alterations to mammary development were observed in MMTV-Cre- or K14-Cre; Ift88Del lines. These mice were able to feed and support several litters of pups even though wide spread depletion of PC was confirmed. Cells grown in mammosphere culture were enriched for PC containing cells suggesting PC are preferentially expressed on mammary stem and progenitor cells. Deletion of Ift88 in mammary epithelial cells resulted in a significant reduction in the number of primary mammospheres established; however, there was no effect on outgrowth of secondary mammospheres in PC-depleted cells. Conclusions PC regulate systemic factors that can affect mammary development in early puberty. PC on MMTV- or K14-expressing epithelial cells are not required for normal mammary development or function. PC are expressed at high levels on cells in mammosphere cultures. PC may be required for cells to establish mammospheres in culture; however, PC are not required for renewal of the cultures.
Collapse
Affiliation(s)
| | - Rosa Serra
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, 1918 University Blvd,, 660 MCLM, Birmingham, AL, 35294-0005, USA.
| |
Collapse
|
26
|
Lee MY, Sun L, Veltmaat JM. Hedgehog and Gli signaling in embryonic mammary gland development. J Mammary Gland Biol Neoplasia 2013; 18:133-8. [PMID: 23677624 PMCID: PMC3691482 DOI: 10.1007/s10911-013-9291-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 04/30/2013] [Indexed: 12/21/2022] Open
Abstract
The first mouse mutation associated with a heritable defect in embryonic mammary gland development was Extratoes. It represents a functional null-mutation of the gene encoding Gli3, which is best known as a transcription factor mediating canonical Hedgehog (Hh) signaling. Here we review the roles of Hh and Gli proteins in murine embryonic mammary development. We propose that an off-state for Hh signaling, mediated by Gli3-repressor, is determinant for induction of a mammary instead of hair follicle fate in the trunk surface ectoderm.
Collapse
Affiliation(s)
- May Yin Lee
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Singapore, 138673 Singapore
- Present Address: Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, 808 Route de Lennik, Brussels, 1070 Belgium
| | - Li Sun
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Singapore, 138673 Singapore
| | - Jacqueline M. Veltmaat
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Singapore, 138673 Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Health System, MD10, 4 Medical Drive, Singapore, 117597 Singapore
| |
Collapse
|
27
|
Nigam A. Breast cancer stem cells, pathways and therapeutic perspectives 2011. Indian J Surg 2013; 75:170-80. [PMID: 24426422 PMCID: PMC3689383 DOI: 10.1007/s12262-012-0616-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 06/06/2012] [Indexed: 01/16/2023] Open
Abstract
The evidence for the existence of a heterogeneous population of cancer stem cells (CSCs) responsible for the initiation and maintenance of cancer has been characterized for several tumors recently. Purification and molecular characterization of normal human mammary stem cells from cultured mammospheres has been achieved, providing evidence supporting a model in which breast tumor heterogeneity is a reflection of a number of CSC-like cells in the tumor. A number of experimental methodologies have been developed to characterize epithelial stem cells, including the expression of cell surface or intracellular markers, mammosphere formation, exclusion of fluorescent dye by a side population, retention of the radionucleotide label, etc. Methodologies have also been successfully employed to identify tumorigenic cells within breast cancers. The most important characteristics of stem cells are the capacity for self-renewal and the regulation of the balance between self-renewal and differentiation. In the mammary gland, signaling pathways, such as Hedgehog (Hh), Wnt/β-catenin, and Notch, play a role in embryogenesis and organogenesis and maintenance of tissues in the adult through regulation of the balance between self-renewal and differentiation of stem cells. Breast TAAs include epitopes from proteins, such as carcinoembryonic antigen and NYBR-1, which are involved in tissue differentiation. Targeting BCSCs may be achieved by a number of approaches such as chemotherapy sensitization of BCSCs, differentiating therapy, targeting stem cell elimination, targeting signaling pathways and drug transporters, and inhibition of regulatory pathways involved in self-renewal. Targeting cells which have the potential to metastasize will be an important aspect of the BCSC field as these are the cells that cause the majority of morbidity and mortality from breast cancer.
Collapse
Affiliation(s)
- Anjana Nigam
- Department of Surgery, Pt.J.N.M.Medical College, Raipur, 492001 CG India
| |
Collapse
|
28
|
Hui M, Cazet A, Nair R, Watkins DN, O'Toole SA, Swarbrick A. The Hedgehog signalling pathway in breast development, carcinogenesis and cancer therapy. Breast Cancer Res 2013; 15:203. [PMID: 23547970 PMCID: PMC3672663 DOI: 10.1186/bcr3401] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Despite the progress achieved in breast cancer screening and therapeutic innovations, the basal-like subtype of breast cancer (BLBC) still represents a particular clinical challenge. In order to make an impact on survival in this type of aggressive breast cancer, new targeted therapeutic agents are urgently needed. Aberrant activation of the Hedgehog (Hh) signalling pathway has been unambiguously tied to cancer development and progression in a variety of solid malignancies, and the recent approval of vismodegib, an orally bioavailable small-molecule inhibitor of Smoothened, validates Hh signalling as a valuable therapeutic target. A number of recent publications have highlighted a role for Hh signalling in breast cancer models and clinical specimens. Interestingly, Hh ligand overexpression is associated with the BLBC phenotype and a poor outcome in terms of metastasis and breast cancer-related death. In this review, we provide a comprehensive overview of the canonical Hh signalling pathway in mammals, highlight its roles in mammary gland development and breast carcinogenesis and discuss its potential therapeutic value in BLBC.
Collapse
|
29
|
García-Zaragoza E, Pérez-Tavarez R, Ballester A, Lafarga V, Jiménez-Reinoso A, Ramírez Á, Murillas R, Gallego MI. Intraepithelial paracrine Hedgehog signaling induces the expansion of ciliated cells that express diverse progenitor cell markers in the basal epithelium of the mouse mammary gland. Dev Biol 2012; 372:28-44. [DOI: 10.1016/j.ydbio.2012.09.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 09/10/2012] [Accepted: 09/11/2012] [Indexed: 10/27/2022]
|
30
|
Karamboulas C, Ailles L. Developmental signaling pathways in cancer stem cells of solid tumors. Biochim Biophys Acta Gen Subj 2012. [PMID: 23196196 DOI: 10.1016/j.bbagen.2012.11.008] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The intricate regulation of several signaling pathways is essential for embryonic development and adult tissue homeostasis. Cancers commonly display aberrant activity within these pathways. A population of cells identified in several cancers, termed cancer stem cells (CSCs) show similar properties to normal stem cells and evidence suggests that altered developmental signaling pathways play an important role in maintaining CSCs and thereby the tumor itself. SCOPE OF REVIEW This review will focus on the roles of the Notch, Wnt and Hedgehog pathways in the brain, breast and colon cancers. We describe the roles these pathways play in normal tissue homeostasis through the regulation of stem cell fate in these three tissues, and the experimental evidence indicating that the role of these pathways in cancers of these is directly linked to CSCs. MAJOR CONCLUSIONS A large body of evidence is accumulating to indicate that the deregulation of Notch, Wnt and Hedgehog pathways play important roles in both normal and cancer stem cells. We are only beginning to understand how these pathways interact, how they are coordinated during normal development and adult tissue homeostasis, and how they are deregulated during cancer. However, it is becoming increasingly clear that if we are to target CSCs therapeutically, it will likely be necessary to develop combination therapies. GENERAL SIGNIFICANCE If CSCs are the driving force behind tumor maintenance and growth then understanding the molecular mechanisms regulating CSCs is essential. Such knowledge will contribute to better targeted therapies that could significantly enhance cancer treatments and patient survival. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Christina Karamboulas
- Ontario Cancer Institute, Campbell Family Cancer Research Institute, University Health Network, Toronto, Ontario, Canada M5G 1L7
| | | |
Collapse
|
31
|
Malhotra GK, Zhao X, Band H, Band V. Shared signaling pathways in normal and breast cancer stem cells. J Carcinog 2011; 10:38. [PMID: 22279423 PMCID: PMC3263309 DOI: 10.4103/1477-3163.91413] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 10/20/2011] [Indexed: 12/31/2022] Open
Abstract
Recent advances in our understanding of breast cancer biology have led to the identification of a subpopulation of cells within tumors that appear to be responsible for initiating and propagating the cancer. These tumor initiating cells are not only unique in their ability to generate tumors, but also share many similarities with elements of normal adult tissue stem cells, and have therefore been termed cancer stem cells (CSCs). These CSCs often inappropriately use many of the same signaling pathways utilized by their normal stem cell counterparts which may present a challenge to the development of CSC specific therapies. Here, we discuss three major stem cell signaling pathways (Notch, Wnt, and Hedgehog); with a focus on their function in normal mammary gland development and their misuse in breast cancer stem cell fate determination.
Collapse
Affiliation(s)
- Gautam K Malhotra
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, USA
| | | | | | | |
Collapse
|
32
|
Garner OB, Bush KT, Nigam KB, Yamaguchi Y, Xu D, Esko JD, Nigam SK. Stage-dependent regulation of mammary ductal branching by heparan sulfate and HGF-cMet signaling. Dev Biol 2011; 355:394-403. [PMID: 21586278 PMCID: PMC3118867 DOI: 10.1016/j.ydbio.2011.04.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 04/18/2011] [Accepted: 04/30/2011] [Indexed: 02/08/2023]
Abstract
Specific interactions of growth factors with heparan sulfate may function as "switches" to regulate stages of branching morphogenesis in developing mammalian organs, such as breast, lung, salivary gland and kidney, but the evidence derives mostly from studies of explanted tissues or cell culture (Shah et al., 2004). We recently provided in vivo evidence that inactivation of Ndst1, the predominant N-deacetylase/N-sulfotransferase gene essential for the formation of mature heparan sulfate, results in a highly specific defect in murine lobuloalveolar development (Crawford et al., 2010). Here, we demonstrate a highly penetrant dramatic defect in primary branching by mammary epithelial-specific inactivation of Ext1, a subunit of the copolymerase complex that catalyzes the formation of the heparan sulfate chain. In contrast to Ext1 deletion, inactivation of Hs2st (which encodes an enzyme required for 2-O-sulfation of uronic acids in heparan sulfate) did not inhibit ductal formation but displayed markedly decreased secondary and ductal side-branches as well as fewer bifurcated terminal end buds. Targeted conditional deletion of c-Met, the receptor for HGF, in mammary epithelial cells showed similar defects in secondary and ductal side-branching, but did not result in any apparent defect in bifurcation of terminal end buds. Although there is published evidence indicating a role for 2-O sulfation in HGF binding, primary epithelial cells isolated from Hs2st conditional deletions were able to activate Erk in the presence of HGF and there appeared to be only a slight reduction in HGF-mediated c-Met phosphorylation in these cells compared to control. Thus, both c-Met and Hs2st play important, but partly independent, roles in secondary and ductal side-branching. When considered together with previous studies of Ndst1-deficient glands, the data presented here raise the possibility of partially-independent regulation by heparan sulfate-dependent pathways of primary ductal branching, terminal end bud bifurcation, secondary branching, ductal side-branching and lobuloalveolar formation.
Collapse
Affiliation(s)
- Omai B. Garner
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, 92093
| | - Kevin T. Bush
- Department of Pediatrics, University of California, San Diego, La Jolla, California, 92093
| | - Kabir B. Nigam
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
| | - Yu Yamaguchi
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California, 92093
| | - Ding Xu
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, 92093
| | - Jeffrey D. Esko
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, 92093
| | - Sanjay K. Nigam
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Pediatrics, University of California, San Diego, La Jolla, California, 92093
- Department of Bioengineering, University of California, San Diego, La Jolla, California, 92093
| |
Collapse
|
33
|
O'Toole SA, Machalek DA, Shearer RF, Millar EK, Nair R, Schofield P, McLeod D, Cooper CL, McNeil CM, McFarland A, Nguyen A, Ormandy CJ, Qiu MR, Rabinovich B, Martelotto LG, Vu D, Hannigan GE, Musgrove EA, Christ D, Sutherland RL, Watkins DN, Swarbrick A. Hedgehog Overexpression Is Associated with Stromal Interactions and Predicts for Poor Outcome in Breast Cancer. Cancer Res 2011; 71:4002-14. [DOI: 10.1158/0008-5472.can-10-3738] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Bussard KM, Smith GH. The mammary gland microenvironment directs progenitor cell fate in vivo. Int J Cell Biol 2011; 2011:451676. [PMID: 21647291 PMCID: PMC3103901 DOI: 10.1155/2011/451676] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 03/11/2011] [Indexed: 12/21/2022] Open
Abstract
The mammary gland is a unique organ that continually undergoes postnatal developmental changes. In mice, the mammary gland is formed via signals from terminal end buds, which direct ductal growth and elongation. Intriguingly, it is likely that the entire cellular repertoire of the mammary gland is formed from a single antecedent cell. Furthermore, in order to produce progeny of varied lineages (e.g., luminal and myoepithelial cells), signals from the local tissue microenvironment influence mammary stem/progenitor cell fate. Data have shown that cells from the mammary gland microenvironment reprogram adult somatic cells from other organs (testes, nerve) into cells that produce milk and express mammary epithelial cell proteins. Similar results were found for human tumorigenic epithelial carcinoma cells. Presently, it is unclear how the deterministic power of the mammary gland microenvironment controls epithelial cell fate. Regardless, signals generated by the microenvironment have a profound influence on progenitor cell differentiation in vivo.
Collapse
Affiliation(s)
| | - Gilbert H. Smith
- Mammary Biology and Tumorigenesis Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
35
|
Visbal AP, LaMarca HL, Villanueva H, Toneff MJ, Li Y, Rosen JM, Lewis MT. Altered differentiation and paracrine stimulation of mammary epithelial cell proliferation by conditionally activated Smoothened. Dev Biol 2011; 352:116-27. [PMID: 21276786 PMCID: PMC3057274 DOI: 10.1016/j.ydbio.2011.01.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Revised: 01/14/2011] [Accepted: 01/19/2011] [Indexed: 11/18/2022]
Abstract
The Hedgehog (Hh) signaling network is critical for patterning and organogenesis in mammals, and has been implicated in a variety of cancers. Smoothened (Smo), the gene encoding the principal signal transducer, is overexpressed frequently in breast cancer, and constitutive activation in MMTV-SmoM2 transgenic mice caused alterations in mammary gland morphology, increased proliferation, and changes in stem/progenitor cell number. Both in transgenic mice and in clinical specimens, proliferative cells did not usually express detectable Smo, suggesting the hypothesis that Smo functioned in a non-cell autonomous manner to stimulate proliferation. Here, we employed a genetically tagged mouse model carrying a Cre-recombinase-dependent conditional allele of constitutively active Smo (SmoM2) to test this hypothesis. MMTV-Cre- or adenoviral-Cre-mediated SmoM2 expression in the luminal epithelium, but not in the myoepithelium, was required for the hyper-proliferative phenotypes. High levels of proliferation were observed in cells adjacent or in close-proximity to Smo expressing cells demonstrating that SmoM2 expressing cells were stimulating proliferation via a paracrine or juxtacrine mechanism. In contrast, Smo expression altered luminal cell differentiation in a cell-autonomous manner. SmoM2 expressing cells, purified by fluorescence activated cell sorting (FACS) via the genetic fluorescent tag, expressed high levels of Ptch2, Gli1, Gli2, Jag2 and Dll-1, and lower levels of Notch4 and Hes6, in comparison to wildtype cells. These studies provide insight into the mechanism of Smo activation in the mammary gland and its possible roles in breast tumorigenesis. In addition, these results also have potential implications for the interpretation of proliferative phenotypes commonly observed in other organs as a consequence of hedgehog signaling activation.
Collapse
Affiliation(s)
- Adriana P. Visbal
- Developmental Biology Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Heather L. LaMarca
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hugo Villanueva
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Michael J. Toneff
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yi Li
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jeffrey M. Rosen
- Developmental Biology Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Michael T. Lewis
- Developmental Biology Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
36
|
Souzaki M, Kubo M, Kai M, Kameda C, Tanaka H, Taguchi T, Tanaka M, Onishi H, Katano M. Hedgehog signaling pathway mediates the progression of non-invasive breast cancer to invasive breast cancer. Cancer Sci 2011; 102:373-81. [PMID: 21091847 PMCID: PMC11159393 DOI: 10.1111/j.1349-7006.2010.01779.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The purpose of this study is to clarify the contribution of the Hedgehog signaling pathway (Hh pathway) to the progression from ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC). A total of 149 surgically resected mammary disease specimens and 12 sentinel lymph nodes with micro-metastasis (Ly-met) were studied. The degree of Hh pathway activation was estimated from the Gli1 nuclear staining ratio (%Gli1 nuclear translocation) in cancer cells. The invasiveness of breast cancer cells was determined using Matrigel assays. A serial increase of %Gli1 nuclear translocation to IDC from non-neoplastic diseases was confirmed. In tumor specimens, %Gli1 nuclear translocation correlated with the invasiveness of each type of mammary disease and also correlated with invasion-related histopathological parameters. The %Gli1 nuclear translocation in lymph nodes with micro-metastasis was similar to that in primary sites and higher than that in DCIS with microinvasion and DCIS. Blockade of the Hh pathway decreased the invasiveness of breast cancer cells. In IDC, %Gli1 nuclear translocation correlated with the expression of estrogen receptor-α. Estrogen increased %Gli1 nuclear translocation and the invasiveness of estrogen receptor-α-positive cells. The Hh pathway mediates progression from a non-invasive phenotype to an invasive phenotype and %Gli1 nuclear translocation may be useful as a predictive marker for evaluating the ability of invasiveness.
Collapse
Affiliation(s)
- Masae Souzaki
- Department of Cancer Therapy and Research, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chia YH, Ma CX. Hedgehog Pathway Inhibitors: Potential Applications in Breast Cancer. CURRENT BREAST CANCER REPORTS 2010. [DOI: 10.1007/s12609-010-0031-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
38
|
Visbal AP, Lewis MT. Hedgehog signaling in the normal and neoplastic mammary gland. Curr Drug Targets 2010; 11:1103-11. [PMID: 20545610 PMCID: PMC5499530 DOI: 10.2174/138945010792006753] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 05/04/2010] [Indexed: 02/07/2023]
Abstract
The hedgehog signal transduction network is a critical regulator of metazoan development. Inappropriate activation of this network is implicated in several different cancers, including breast. Genetic evidence in mice as well as molecular biological studies in human cells clearly indicate that activated signaling can lead to mammary hyperplasia and, in some cases, tumor formation. However, the exact role(s) activated hedgehog signaling plays in the development or progression of breast cancer also remain unclear. In this review, we have discussed recent data regarding the mechanism(s) by which the hedgehog network may signal in the mammary gland, as well as the data implicating activated signaling as a contributing factor to breast cancer development. Finally, we provide a brief update on the available hedgehog signaling inhibitors with respect to ongoing clinical trials, some of which will include locally advanced or metastatic breast cancers. Given the growing intensity with which the hedgehog signaling network is being studied in the normal and neoplastic mammary gland, a more complete understanding of this network should allow more effective targeting of its activities in breast cancer treatment or prevention.
Collapse
Affiliation(s)
- Adriana P. Visbal
- The Lester and Sue Smith Breast Center, Baylor College of Medicine,
One Baylor Plaza, BCM600; Room N1210, Houston TX 77030, TEL: 713-798-3296, FAX:
713-798-1659
- Program in Developmental Biology
| | - Michael T. Lewis
- The Lester and Sue Smith Breast Center, Baylor College of Medicine,
One Baylor Plaza, BCM600; Room N1210, Houston TX 77030, TEL: 713-798-3296, FAX:
713-798-1659
- Departments of Molecular and Cellular Biology and Radiology
| |
Collapse
|
39
|
Barakat MT, Humke EW, Scott MP. Learning from Jekyll to control Hyde: Hedgehog signaling in development and cancer. Trends Mol Med 2010; 16:337-48. [PMID: 20696410 PMCID: PMC3651907 DOI: 10.1016/j.molmed.2010.05.003] [Citation(s) in RCA: 172] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Revised: 05/13/2010] [Accepted: 05/17/2010] [Indexed: 02/07/2023]
Abstract
The Hedgehog (Hh) cascade controls cell proliferation, differentiation and patterning of tissues during embryogenesis but is largely suppressed in the adult. The Hh pathway can become reactivated in cancer. Here, we assimilate data from recent studies to understand how and when the Hh pathway is turned on to aid the neoplastic process. Hh signaling is now known to have a role in established tumors, enabling categorization of tumors based on the role Hh signaling plays in their growth. This categorization has relevance for prognosis and targeted therapeutics. In the first category, abnormal Hh signaling initiates the tumor. In the second category, Hh signaling helps maintain the tumor. In the third category, Hh signaling is implicated but its role is not yet defined.
Collapse
Affiliation(s)
- Monique T Barakat
- Department of Developmental Biology, Howard Hughes Medical Institute, Clark Center West W252, 318 Campus Drive, Stanford University School of Medicine, Stanford, CA 94305-5439, USA
| | | | | |
Collapse
|
40
|
Crawford BE, Garner OB, Bishop JR, Zhang DY, Bush KT, Nigam SK, Esko JD. Loss of the heparan sulfate sulfotransferase, Ndst1, in mammary epithelial cells selectively blocks lobuloalveolar development in mice. PLoS One 2010; 5:e10691. [PMID: 20502530 PMCID: PMC2872662 DOI: 10.1371/journal.pone.0010691] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2010] [Accepted: 04/26/2010] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Considerable evidence indicates that heparan sulfate is essential for the development of tissues consisting of branching ducts and tubules. However, there are few examples where specific sulfate residues regulate a specific stage in the formation of such tissues. METHODOLOGY/PRINCIPAL FINDINGS We examined the role of heparan sulfation in mammary gland branching morphogenesis, lactation and lobuloalveolar development by inactivation of heparan sulfate GlcNAc N-deacetylase/N-sulfotransferase genes (Ndst) in mammary epithelial cells using the Cre-loxP system. Ndst1 deficiency resulted in an overall reduction in glucosamine N-sulfation and decreased binding of FGF to mammary epithelial cells in vitro and in vivo. Mammary epithelia lacking Ndst1 underwent branching morphogenesis, filling the gland with ductal tissue by sexual maturity to the same extent as wildtype epithelia. However, lobuloalveolar expansion did not occur in Ndst1-deficient animals, resulting in insufficient milk production to nurture newly born pups. Lactational differentiation of isolated mammary epithelial cells occurred appropriately via stat5 activation, further supporting the notion that the lack of milk production was due to lack of expansion of the lobuloalveoli. CONCLUSIONS/SIGNIFICANCE These findings demonstrate a selective, highly penetrant, cell autonomous effect of Ndst1-mediated sulfation on lobuloalveolar development.
Collapse
Affiliation(s)
- Brett E. Crawford
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California, United States of America
| | - Omai B. Garner
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California, United States of America
| | - Joseph R. Bishop
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - David Y. Zhang
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Kevin T. Bush
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Sanjay K. Nigam
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jeffrey D. Esko
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
41
|
O'Toole SA, Swarbrick A, Sutherland RL. The Hedgehog signalling pathway as a therapeutic target in early breast cancer development. Expert Opin Ther Targets 2009; 13:1095-103. [PMID: 19659449 DOI: 10.1517/14728220903130612] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The Hedgehog (Hh) signalling pathway is a highly conserved developmental pathway, which plays critical roles in patterning of the embryo through epithelial to mesenchymal signalling and the maintenance of stem cells in the adult organism. There is increasing evidence that this pathway is dysregulated in many malignancies, including breast cancer. While there has been a significant decrease in mortality from breast cancer, a number of treatment challenges remain, particularly in those tumours which develop resistance to endocrine-based therapy, or which lack expression of hormone or c-erbB2/HER2 receptors. Therapeutic manipulation of the Hh pathway as a potential cancer therapy is attracting great interest, with preclinical studies and clinical trials underway in a range of malignancies. This review highlights important recent developments that affect the potential of the Hh pathway as a novel therapeutic target in early breast cancer.
Collapse
Affiliation(s)
- Sandra A O'Toole
- Garvan Institute of Medical Research, Cancer Research Program, Darlinghurst 2010, NSW, Australia
| | | | | |
Collapse
|
42
|
Zhang X, Harrington N, Moraes RC, Wu MF, Hilsenbeck SG, Lewis MT. Cyclopamine inhibition of human breast cancer cell growth independent of Smoothened (Smo). Breast Cancer Res Treat 2009; 115:505-21. [PMID: 18563554 PMCID: PMC5300001 DOI: 10.1007/s10549-008-0093-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 06/04/2008] [Indexed: 11/25/2022]
Abstract
Altered hedgehog signaling is implicated in the development of approximately 20-25% of all cancers, especially those of soft tissues. Genetic evidence in mice as well as immunolocalization studies in human breast cancer specimens suggest that deregulated hedgehog signaling may contribute to breast cancer development. Indeed, two recent studies demonstrated that anchorage-dependent growth of some human breast cancer cell lines is impaired by cyclopamine, a potent hedgehog signaling antagonist targeting the Smoothened (SMO) protein. However, specificity of cyclopamine at the dosage required for growth inhibition (> or =10 microM) remained an open question. In this paper we demonstrate that hedgehog signaling antagonists, including cyclopamine, and a second compound, CUR0199691, can inhibit growth of estrogen receptor (ER)-positive and ER-negative tumorigenic breast cancer cells at elevated doses. However, our results indicate that, for most breast cancer cell lines, growth inhibition by these compounds can be independent of detectable Smo gene expression. Rather, our results suggest that cyclopamine and CUR0199691 have unique secondary molecular targets at the dosages required for growth inhibition that are unrelated to hedgehog signaling.
Collapse
MESH Headings
- Apoptosis/drug effects
- Binding, Competitive
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Adhesion/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cholesterol/metabolism
- Drug Resistance, Neoplasm/drug effects
- Female
- Flow Cytometry
- Fluorescent Antibody Technique
- Gene Expression Regulation, Neoplastic/drug effects
- Hedgehog Proteins/genetics
- Hedgehog Proteins/metabolism
- Humans
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Estrogen/metabolism
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Smoothened Receptor
- Veratrum Alkaloids/pharmacology
Collapse
Affiliation(s)
- Xiaomei Zhang
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| | - Nikesha Harrington
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| | - Ricardo C. Moraes
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| | - Meng-Fen Wu
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| | - Susan G. Hilsenbeck
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| |
Collapse
|
43
|
Moraes RC, Chang H, Harrington N, Landua JD, Prigge JT, Lane TF, Wainwright BJ, Hamel PA, Lewis MT. Ptch1 is required locally for mammary gland morphogenesis and systemically for ductal elongation. Development 2009; 136:1423-32. [PMID: 19297414 DOI: 10.1242/dev.023994] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Systemic hormones and local growth factor-mediated tissue interactions are essential for mammary gland development. Using phenotypic and transplantation analyses of mice carrying the mesenchymal dysplasia (mes) allele of patched 1 (Ptch1(mes)), we found that Ptch1(mes) homozygosity led to either complete failure of gland development, failure of post-pubertal ductal elongation, or delayed growth with ductal dysplasia. All ductal phenotypes could be present in the same animal. Whole gland and epithelial fragment transplantation each yielded unique morphological defects indicating both epithelial and stromal functions for Ptch1. However, ductal elongation was rescued in all cases, suggesting an additional systemic function. Epithelial function was confirmed using a conditional null Ptch1 allele via MMTV-Cre-mediated disruption. In Ptch1(mes) homozygotes, failure of ductal elongation correlated with diminished estrogen and progesterone receptor expression, but could not be rescued by exogenous ovarian hormone treatment. By contrast, pituitary isografts were able to rescue the ductal elongation phenotype. Thus, Ptch1 functions in the mammary epithelium and stroma to regulate ductal morphogenesis, and in the pituitary to regulate ductal elongation and ovarian hormone responsiveness.
Collapse
Affiliation(s)
- Ricardo C Moraes
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kasper M, Jaks V, Fiaschi M, Toftgård R. Hedgehog signalling in breast cancer. Carcinogenesis 2009; 30:903-11. [PMID: 19237605 DOI: 10.1093/carcin/bgp048] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most common cause of cancer death among women worldwide. In order to improve the treatment of this disease, a more complete understanding of its biological basis is necessary. Since the Hedgehog (Hh) pathway was recently found to be required for growth and propagation of a number of different cancers, we discuss here the possible involvement of this pathway in the normal biology and development of cancer in the mammary gland. The use of mouse mammary cancer models has assisted the process of dissecting the mechanisms behind Hh-driven mammary tumour formation and growth. Based on recent studies, we conclude that the inhibition of Hh signalling in breast tumours may interfere with the maintenance of a putative cancer stem cell compartment and the abnormal stimulation of tumour stroma. Therefore, the components of the Hh signalling cascade may provide a set of drug targets, which could be implemented into novel combinatorial strategies for the treatment of breast cancer.
Collapse
Affiliation(s)
- Maria Kasper
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden
| | | | | | | |
Collapse
|
45
|
Expression of Gli1 correlates with the transition of breast cancer cells to estrogen-independent growth. Breast Cancer Res Treat 2009; 119:39-51. [PMID: 19191023 DOI: 10.1007/s10549-009-0323-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2008] [Accepted: 01/16/2009] [Indexed: 12/11/2022]
Abstract
The failure of breast cancer treatment is largely due to the development of estrogen independence. Current data illustrate that Hedgehog (Hh) signaling may play an important role in breast cancer development. Here, we show that the expression of the Hh effector protein, Gli1 was significantly higher in estrogen-independent breast cancer cells than in estrogen-dependent cells. Our data showed for the first time that stable expression of Gli1 in ER positive breast cancer cell lines MCF-7 and T47D can induce estrogen-independent proliferation and promote G1/S phase transition, which associated with cyclin-Rb axi. Gli1 can also attenuate the response of proliferation to estrogenic stimulation, which was correlated with down-regulation of expression of ERalpha and PR, as well as down-regulation of transactivation of ERalpha. Our results suggest that up-regulation of Gli1 in breast cancer cells may be one of the mechanisms responsible for developing estrogen independence and this process may be regulated through down-regulation of expression and transactivation of ERalpha.
Collapse
|
46
|
Abstract
Mammary stem cells have recently been identified and purified on the basis of surface antigens and transplantation assays. In addition, recent reports have identified a small sub-population of highly tumorigenic cells within primary and metastatic breast tumors and in a number of breast cancer cell lines. This suggests that, similarly to its normal physiological counterpart, a cancer stem cell may be at the origin of breast cancer. These observations have dramatic biological and clinical implications, as they dictate a revision of our understanding of breast cancer and of our therapeutic strategies. The aim of this article is to review recent data regarding normal mammary epithelial stem cells and evidence in support of the cancer stem cell hypothesis in the breast, and to provide further insight into how taking this subpopulation of cells into account may affect the way we treat epithelial cancers in the future.
Collapse
Affiliation(s)
- Massimiliano Cariati
- Department of Academic Oncology, King's College London, 3rd Floor Bermondsey Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
| |
Collapse
|
47
|
The glandular stem/progenitor cell niche in airway development and repair. Ann Am Thorac Soc 2008; 5:682-8. [PMID: 18684717 DOI: 10.1513/pats.200801-003aw] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Airway submucosal glands (SMGs) are major secretory structures that lie beneath the epithelium of the cartilaginous airway. These glands are believed to play important roles in normal lung function and airway innate immunity by secreting antibacterial factors, mucus, and fluid into the airway lumen. Recent studies have suggested that SMGs may additionally serve as a protective niche for adult epithelial stem/progenitor cells of the proximal airways. As in the case of other adult stem cell niches, SMGs are believed to provide the localized environmental signals required to both maintain and mobilize stem/progenitor cells, in the setting of normal cellular turnover or injury. Aberrant proliferation and differentiation of glandular stem/progenitor cells may be associated with several hypersecretory lung diseases, including chronic bronchitis, asthma, and cystic fibrosis. To better understand the molecular mechanisms that regulate the specification and proliferation of glandular stem/progenitor cells in lung diseases associated with SMG hypertrophy and hyperplasia, researchers have begun to search for the molecular signals and cell types responsible for establishing the glandular stem/progenitor cell niche, and to dissect how these determinants of the niche change in the setting of proximal airway injury and repair. Such studies have revealed certain similarities between stem/progenitor cell niches of the distal conducting airways and the SMGs of the proximal airways.
Collapse
|
48
|
Johnson ET, Nicola T, Roarty K, Yoder BK, Haycraft CJ, Serra R. Role for primary cilia in the regulation of mouse ovarian function. Dev Dyn 2008; 237:2053-60. [PMID: 18629867 DOI: 10.1002/dvdy.21612] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Ift88 is a component of the intraflagellar transport complex required for formation and maintenance of cilia. Disruption of Ift88 results in depletion of cilia. The goal of the current study was to determine the role of primary cilia in ovarian function. Deletion of Ift88 in ovary using Cre-Lox recombination in mice resulted in a severe delay in mammary gland development including lack of terminal end bud structures, alterations in the estrous cycle, and impaired ovulation. Because estrogen drives the formation of end buds and Cre was expressed in the granulosa cells of the ovary, we tested the hypothesis that addition of estradiol to the mutant mice would compensate for defects in ovarian function and rescue the mammary gland phenotype. Mammary gland development including the formation of end bud structures resumed in mutant mice that were injected with estradiol. Together the results suggest that cilia are required for ovarian function.
Collapse
Affiliation(s)
- Ellen T Johnson
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0005, USA
| | | | | | | | | | | |
Collapse
|
49
|
Expression of Indian Hedgehog signaling molecules in breast cancer. J Cancer Res Clin Oncol 2008; 135:235-40. [PMID: 18636275 DOI: 10.1007/s00432-008-0451-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Accepted: 06/21/2008] [Indexed: 01/20/2023]
Abstract
PURPOSE To investigate the clinicopathological significance and expression pattern of Hedgehog (Hh) signaling molecules in breast normal glands and invasive ductal carcinoma. MATERIALS AND METHODS A total of 142 cases, including 21 of normal breast and 121 of invasive ductal carcinoma of the breast, were immunohistochemically analyzed for Ihh, Ptch, Smo, Gli-1, Gli-2, and Gli-3 protein expression. RESULTS All of Hh signaling molecules were greatly enhanced in invasive ductal carcinoma compared with the normal breast epithelia. The expressions of Ihh, Smo, and Gli-2 were increased in PR negative cases, and the expressions of Ihh, Ptch, and Gli-1/2/3 were statistically correlated with increased proliferating index of Ki-67 in invasive ductal carcinoma. Ihh and Gli-1/2/3 expressions were correlated with node metastasis. Additionally, the protein expressions of Ihh, Ptch, and Gli-2 were correlated with the clinical stage of breast cancer. CONCLUSIONS Hedgehog signaling molecules play an important role in the progression of invasive ductal carcinoma of breast.
Collapse
|
50
|
|