1
|
Behairi N, Samer A, Sahraoui L, Mataam DH, Trari R, Flissi B, Belguendouz H, Amir ZC, Touil-Boukoffa C. Neuroinflammation, neurodegeneration and alteration of spatial memory in BALB/c mice through ampicillin-induced gut dysbiosis; NOS2 and NFL involvement in a microbiota-gut-brain axis model. J Neuroimmunol 2024; 392:578374. [PMID: 38797060 DOI: 10.1016/j.jneuroim.2024.578374] [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: 02/19/2024] [Revised: 04/05/2024] [Accepted: 05/18/2024] [Indexed: 05/29/2024]
Abstract
We aimed to investigate ampicillin (AMP) mechanisms in microbiota-gut-brain axis. We evaluated its effect on two gut and brain regions and behavioral performances. We administred AMP (1 g/l) to BALB/c mice for 21 days. Then, we analyzed body weigth change, stool consistency scoring, gut length, intestinal microbiota composition, nitric oxide synthase 2 (NOS2) expression and tissue integrity. We subsequently evaluated NOS2, GFAP, CD68 and NFL cerebral expression and spatial memory.Interestingly, our data showed gut microbiota disruption, NOS2 upregulation and tissue damage, associated to cerebral NOS2, GFAP, CD68 and NFL over-expression and behavioral alteration. Antiobiotic therapy should be prescribed with great caution.
Collapse
Affiliation(s)
- Nassima Behairi
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Arezki Samer
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Lynda Sahraoui
- Laboratory of Animal Health and Production, Higher National Veterinary School of Issad-Abbes Oued-Smar, Algiers, Algeria
| | - Djehane Houria Mataam
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Ryad Trari
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Billel Flissi
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Houda Belguendouz
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Zine-Charaf Amir
- Department of Anatomy and Pathological Cytology, University Hospital Center Mustapha Pacha, 1945 Pl. May 1st, Sidi M'Hamed, 16000 Algiers, Algeria
| | - Chafia Touil-Boukoffa
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria.
| |
Collapse
|
2
|
Jiao X, Di Sante G, Casimiro MC, Tantos A, Ashton AW, Li Z, Quach Y, Bhargava D, Di Rocco A, Pupo C, Crosariol M, Lazar T, Tompa P, Wang C, Yu Z, Zhang Z, Aldaaysi K, Vadlamudi R, Mann M, Skordalakes E, Kossenkov A, Du Y, Pestell RG. A cyclin D1 intrinsically disordered domain accesses modified histone motifs to govern gene transcription. Oncogenesis 2024; 13:4. [PMID: 38191593 PMCID: PMC10774418 DOI: 10.1038/s41389-023-00502-1] [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: 02/20/2023] [Revised: 11/09/2023] [Accepted: 12/05/2023] [Indexed: 01/10/2024] Open
Abstract
The essential G1-cyclin, CCND1, is frequently overexpressed in cancer, contributing to tumorigenesis by driving cell-cycle progression. D-type cyclins are rate-limiting regulators of G1-S progression in mammalian cells via their ability to bind and activate CDK4 and CDK6. In addition, cyclin D1 conveys kinase-independent transcriptional functions of cyclin D1. Here we report that cyclin D1 associates with H2BS14 via an intrinsically disordered domain (IDD). The same region of cyclin D1 was necessary for the induction of aneuploidy, induction of the DNA damage response, cyclin D1-mediated recruitment into chromatin, and CIN gene transcription. In response to DNA damage H2BS14 phosphorylation occurs, resulting in co-localization with γH2AX in DNA damage foci. Cyclin D1 ChIP seq and γH2AX ChIP seq revealed ~14% overlap. As the cyclin D1 IDD functioned independently of the CDK activity to drive CIN, the IDD domain may provide a rationale new target to complement CDK-extinction strategies.
Collapse
Affiliation(s)
- Xuanmao Jiao
- Baruch S. Blumberg Institute, Doylestown, PA, 18902, USA
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | | | - Mathew C Casimiro
- Baruch S. Blumberg Institute, Doylestown, PA, 18902, USA
- Department of Science and Mathematics, Abraham Baldwin Agricultural College, Tifton, GA, 31794, USA
| | - Agnes Tantos
- Institute of Enzymology, Hun-Ren Research Centre for Natural Sciences, Budapest, Hungary
| | - Anthony W Ashton
- Baruch S. Blumberg Institute, Doylestown, PA, 18902, USA
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
- Division of Cardiovascular Medicine, Lankenau Institute for Medical Research, Wynnewood, PA, 19003, USA
| | - Zhiping Li
- Baruch S. Blumberg Institute, Doylestown, PA, 18902, USA
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Yen Quach
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | | | | | - Claudia Pupo
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Marco Crosariol
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Tamas Lazar
- VIB-VUB Center for Structural Biology, Vrije Universiteit Brussel, Brussels, 1050, Belgium
| | - Peter Tompa
- Institute of Enzymology, Hun-Ren Research Centre for Natural Sciences, Budapest, Hungary
- VIB-VUB Center for Structural Biology, Vrije Universiteit Brussel, Brussels, 1050, Belgium
| | - Chenguang Wang
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Zuoren Yu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhao Zhang
- Baruch S. Blumberg Institute, Doylestown, PA, 18902, USA
| | - Kawthar Aldaaysi
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba
| | - Ratna Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Sciences Center, San Antonio, TX, 78229, USA
| | - Monica Mann
- Department of Obstetrics and Gynecology, University of Texas Health Sciences Center, San Antonio, TX, 78229, USA
| | | | | | - Yanming Du
- Baruch S. Blumberg Institute, Doylestown, PA, 18902, USA
| | - Richard G Pestell
- Baruch S. Blumberg Institute, Doylestown, PA, 18902, USA.
- Xavier University School of Medicine at Aruba, Oranjestad, Aruba.
- The Wistar Institute, Philadelphia, PA, 19107, USA.
| |
Collapse
|
3
|
Cell-free probiotic supernatant (CFS) treatment alleviates indomethacin-induced enterocolopathy in BALB/c mice by down-modulating inflammatory response and oxidative stress: potential alternative targeted treatment. Inflammopharmacology 2022; 30:1685-1703. [PMID: 35505268 DOI: 10.1007/s10787-022-00996-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/08/2022] [Indexed: 11/05/2022]
Abstract
Probiotics and their metabolites appear to be a promising approach that targets both the intestinal inflammation and dysbiosis in bowel diseases. In this context, the emergence of the probiotic cell-free supernatant (CFS) has attracted more attention as a safe and targeted alternative therapy with reduced side effects. The use of nonsteroidal anti-inflammatory drugs (NSAIDs) can cause significant intestinal alterations and inflammation, leading to experimental enterocolopathy resembling Crohn disease. Therefore, we investigated the effect of CFS supplementation on the inflammation and the mucosal intestinal alterations induced by NSAIDs, indomethacin. In the current study, a murine model of intestinal inflammation was generated by the oral gavage (o.g) of indomethacin (10 mg/kg) to BALB/C mice. A group of mice treated with indomethacin was concomitantly treated orally by CFS for 5 days. The Body Health Condition index was monitored, and histological scores were evaluated. Moreover, oxidative and pro-inflammatory markers were assessed. Interestingly, we observed that CFS treatment attenuated the severity of the intestinal inflammation in our enterocolopathy model and resulted in the improvement of the clinical symptoms and the histopathological features. Notably, nitric oxide, tumor necrosis factor alpha, malondialdehyde, and myeloperoxidase levels were down-modulated by CFS supplementation. Concomitantly, an attenuation of NF-κB p65, iNOS, COX2 expression in the ileum and the colon was reported. Collectively, our data suggest that CFS treatment has a beneficial effect in experimental enterocolopathy model and could constitute a good therapeutic candidate for alleviating inflammatory responses and to maintain mucosal homeostasis during chronic and severe conditions of intestinal inflammation.
Collapse
|
4
|
Cruciferous Vegetables and Their Bioactive Metabolites: from Prevention to Novel Therapies of Colorectal Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1534083. [PMID: 35449807 PMCID: PMC9017484 DOI: 10.1155/2022/1534083] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/16/2022] [Indexed: 02/07/2023]
Abstract
The Brassicaceae family, known as cruciferous vegetables, includes many economically important species, mainly edible oil plants, vegetable species, spice plants, and feed plants. Cruciferous vegetables are foods rich in nutritive composition and are also a good source of dietary fiber. Besides, cruciferous vegetables contain various bioactive chemicals known as glucosinolates and S-methyl cysteine sulfoxide, including sulphur-containing cancer-protective chemicals. Numerous studies have reported that daily intake of sulphurous vegetables helps prevent cancer formation and reduces cancer incidence, especially in colorectal cancer, through various mechanisms. The potential mechanisms of these compounds in preventing cancer in experimental studies are as follows: protecting cells against DNA damage, inactivating carcinogenic substances, showing antiviral and antibacterial effects, triggering apoptosis in cells with disrupted structure, inhibiting tumour cell migration causing metastasis and the development of tumour-feeding vessels (angiogenesis). These beneficial anticancer effects of cruciferous vegetables are generally associated with glucosinolates in their composition and some secondary metabolites, as well as other phenolic compounds, seed oils, and dietary fiber in the literature. This review aims to examine to the roles of cruciferous vegetables and their important bioactive metabolites in the prevention and treatment of colorectal cancer.
Collapse
|
5
|
Takakura H, Horinaka M, Imai A, Aono Y, Nakao T, Miyamoto S, Iizumi Y, Watanabe M, Narita T, Ishikawa H, Mutoh M, Sakai T. Sodium salicylate and 5-aminosalicylic acid synergistically inhibit the growth of human colon cancer cells and mouse intestinal polyp-derived cells. J Clin Biochem Nutr 2022; 70:93-102. [PMID: 35400827 PMCID: PMC8921728 DOI: 10.3164/jcbn.21-74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 10/20/2021] [Indexed: 12/24/2022] Open
Abstract
As colon cancer is one of the most common cancers in the world, practical prevention strategies for colon cancer are needed. Recently, treatment with aspirin and/or 5-aminosalicylic acid-related agents was reported to reduce the number of intestinal polyps in patients with familial adenomatous polyposis. To evaluate the mechanism of aspirin and 5-aminosalicylic acid for suppressing the colon polyp growth, single and combined effects of 5-aminosalicylic acid and sodium salicylate (metabolite of aspirin) were tested in the two human colon cancer cells with different cyclooxygenase-2 expression levels and intestinal polyp-derived cells from familial adenomatous polyposis model mouse. The combination induced cell-cycle arrest at the G1 phase along with inhibition of cell growth and colony-forming ability in these cells. The combination reduced cyclin D1 via proteasomal degradation and activated retinoblastoma protein. The combination inhibited the colony-forming ability of mouse colonic mucosa cells by about 50% and the colony-forming ability of mouse intestinal polyp-derived cells by about 90%. The expression level of cyclin D1 in colon mucosa cells was lower than that in intestinal polyp-derived cells. These results suggest that this combination may be more effective in inhibiting cell growth of intestinal polyps through cyclin D1 down-regulation.
Collapse
Affiliation(s)
- Hideki Takakura
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine
| | - Mano Horinaka
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine
| | - Ayaka Imai
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine
| | - Yuichi Aono
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine
| | - Toshimasa Nakao
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine
| | - Shingo Miyamoto
- Epidemiology and Prevention Division, Center for Public Health Sciences, National Cancer Center
| | - Yosuke Iizumi
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine
| | - Motoki Watanabe
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine
| | - Takumi Narita
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine
| | - Hideki Ishikawa
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine
| | - Michihiro Mutoh
- Epidemiology and Prevention Division, Center for Public Health Sciences, National Cancer Center
| | - Toshiyuki Sakai
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine
| |
Collapse
|
6
|
Modeling Intestinal Stem Cell Function with Organoids. Int J Mol Sci 2021; 22:ijms222010912. [PMID: 34681571 PMCID: PMC8535974 DOI: 10.3390/ijms222010912] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/11/2022] Open
Abstract
Intestinal epithelial cells (IECs) are crucial for the digestive process and nutrient absorption. The intestinal epithelium is composed of the different cell types of the small intestine (mainly, enterocytes, goblet cells, Paneth cells, enteroendocrine cells, and tuft cells). The small intestine is characterized by the presence of crypt-villus units that are in a state of homeostatic cell turnover. Organoid technology enables an efficient expansion of intestinal epithelial tissue in vitro. Thus, organoids hold great promise for use in medical research and in the development of new treatments. At present, the cholinergic system involved in IECs and intestinal stem cells (ISCs) are attracting a great deal of attention. Thus, understanding the biological processes triggered by epithelial cholinergic activation by acetylcholine (ACh), which is produced and released from neuronal and/or non-neuronal tissue, is of key importance. Cholinergic signaling via ACh receptors plays a pivotal role in IEC growth and differentiation. Here, we discuss current views on neuronal innervation and non-neuronal control of the small intestinal crypts and their impact on ISC proliferation, differentiation, and maintenance. Since technology using intestinal organoid culture systems is advancing, we also outline an organoid-based organ replacement approach for intestinal diseases.
Collapse
|
7
|
Neurotensin Regulates Proliferation and Stem Cell Function in the Small Intestine in a Nutrient-Dependent Manner. Cell Mol Gastroenterol Hepatol 2021; 13:501-516. [PMID: 34560309 PMCID: PMC8688554 DOI: 10.1016/j.jcmgh.2021.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Intestinal stem cells (ISCs) are sensitive to dietary alterations and nutrient availability. Neurotensin (NT), a gut peptide localized predominantly to the small bowel and released by fat ingestion, stimulates the growth of intestinal mucosa under basal conditions and during periods of nutrient deprivation, suggesting a possible role for NT on ISC function. METHODS Leucine-rich repeat-containing G-protein coupled receptor 5-Enhanced Green Fluorescent Protein (Lgr5-EGFP) NT wild type (Nt+/+) and Lgr5-EGFP NT knockout (Nt-/-) mice were fed ad libitum or fasted for 48 hours. Small intestine tissue and crypts were examined by gene expression analyses, fluorescence-activated cell sorting, Western blot, immunohistochemistry, and crypt-derived organoid culture. Drosophila expressing NT in midgut enteroendocrine cells were fed a standard diet or low-energy diet and esg-green fluorescent protein+ ISCs were quantified via immunofluorescence. RESULTS Loss of NT impaired crypt cell proliferation and ISC function in a manner dependent on nutrient status. Under nutrient-rich conditions, NT stimulated extracellular signal-regulated kinases 1 and 2 signaling and the expression of genes that promote cell-cycle progression, leading to crypt cell proliferation. Under conditions of nutrient depletion, NT stimulated WNT/β-catenin signaling and promoted an ISC gene signature, leading to enhanced ISC function. NT was required for the induction of WNT/β-catenin signaling and ISC-specific gene expression during nutrient depletion, and loss of NT reduced crypt cell proliferation and impaired ISC function and Lgr5 expression in the intestine during fasting. Conversely, the expression of NT in midgut enteroendocrine cells of Drosophila prevented loss of ISCs during nutrient depletion. CONCLUSIONS Collectively, our findings establish an evolutionarily conserved role for NT in ISC maintenance during nutritional stress. GSE182828.
Collapse
|
8
|
Findeiss E, Schwarz SC, Evsyukov V, Rösler TW, Höllerhage M, Chakroun T, Nykänen NP, Shen Y, Wurst W, Kohl M, Tost J, Höglinger GU. Comprehensive miRNome-Wide Profiling in a Neuronal Cell Model of Synucleinopathy Implies Involvement of Cell Cycle Genes. Front Cell Dev Biol 2021; 9:561086. [PMID: 33748099 PMCID: PMC7969723 DOI: 10.3389/fcell.2021.561086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 02/11/2021] [Indexed: 12/13/2022] Open
Abstract
Growing evidence suggests that epigenetic mechanisms like microRNA-mediated transcriptional regulation contribute to the pathogenesis of parkinsonism. In order to study the influence of microRNAs (miRNAs), we analyzed the miRNome 2 days prior to major cell death in α-synuclein-overexpressing Lund human mesencephalic neurons, a well-established cell model of Parkinson’s disease (PD), by next-generation sequencing. The expression levels of 23 miRNAs were significantly altered in α-synuclein-overexpressing cells, 11 were down- and 12 upregulated (P < 0.01; non-adjusted). The in silico analysis of known target genes of these miRNAs was complemented by the inclusion of a transcriptome dataset (BeadChip) of the same cellular system, revealing the G0/G1 cell cycle transition to be markedly enriched. Out of 124 KEGG-annotated cell cycle genes, 15 were present in the miRNA target gene dataset and six G0/G1 cell cycle genes were found to be significantly altered upon α-synuclein overexpression, with five genes up- (CCND1, CCND2, and CDK4 at P < 0.01; E2F3, MYC at P < 0.05) and one gene downregulated (CDKN1C at P < 0.001). Additionally, several of these altered genes are targeted by miRNAs hsa-miR-34a-5p and hsa-miR-34c-5p, which also modulate α-synuclein expression levels. Functional intervention by siRNA-mediated knockdown of the cell cycle gene cyclin D1 (CCND1) confirmed that silencing of cell cycle initiation is able to substantially reduce α-synuclein-mediated cytotoxicity. The present findings suggest that α-synuclein accumulation induces microRNA-mediated aberrant cell cycle activation in post-mitotic dopaminergic neurons. Thus, the mitotic cell cycle pathway at the level of miRNAs might offer interesting novel therapeutic targets for PD.
Collapse
Affiliation(s)
- Elisabeth Findeiss
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases, Munich, Germany.,Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Sigrid C Schwarz
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases, Munich, Germany
| | - Valentin Evsyukov
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases, Munich, Germany.,Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany.,Department of Neurology, Hannover Medical School, Hanover, Germany
| | - Thomas W Rösler
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases, Munich, Germany.,Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthias Höllerhage
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases, Munich, Germany.,Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany.,Department of Neurology, Hannover Medical School, Hanover, Germany
| | - Tasnim Chakroun
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases, Munich, Germany.,Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Niko-Petteri Nykänen
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases, Munich, Germany.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Yimin Shen
- Laboratory for Epigenetics and Environment, Center National de Recherche en Génomique Humaine, CEA-Institut de Biologie François Jacob, Evry, France
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Center Munich, Munich, Germany.,Genome Engineering, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Michael Kohl
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Jörg Tost
- Laboratory for Epigenetics and Environment, Center National de Recherche en Génomique Humaine, CEA-Institut de Biologie François Jacob, Evry, France
| | - Günter U Höglinger
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases, Munich, Germany.,Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany.,Department of Neurology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
9
|
Yang L, Yang H, Chu Y, Song Y, Ding L, Zhu B, Zhai W, Wang X, Kuang Y, Ren F, Jia B, Wu W, Ye X, Wang Y, Chang Z. CREPT is required for murine stem cell maintenance during intestinal regeneration. Nat Commun 2021; 12:270. [PMID: 33431892 PMCID: PMC7801528 DOI: 10.1038/s41467-020-20636-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Intestinal stem cells (ISCs) residing in the crypts are critical for the continual self-renewal and rapid recovery of the intestinal epithelium. The regulatory mechanism of ISCs is not fully understood. Here we report that CREPT, a recently identified tumor-promoting protein, is required for the maintenance of murine ISCs. CREPT is preferably expressed in the crypts but not in the villi. Deletion of CREPT in the intestinal epithelium of mice (Vil-CREPTKO) results in lower body weight and slow migration of epithelial cells in the intestine. Vil-CREPTKO intestine fails to regenerate after X-ray irradiation and dextran sulfate sodium (DSS) treatment. Accordingly, the deletion of CREPT decreases the expression of genes related to the proliferation and differentiation of ISCs and reduces Lgr5+ cell numbers at homeostasis. We identify that CREPT deficiency downregulates Wnt signaling by impairing β-catenin accumulation in the nucleus of the crypt cells during regeneration. Our study provides a previously undefined regulator of ISCs.
Collapse
Affiliation(s)
- Liu Yang
- State Key Laboratory of Membrane Biology, School of Medicine, Center for Synthetic and Systems Biology, Tsinghua University, 100084, Beijing, China
| | - Haiyan Yang
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Yunxiang Chu
- Department of Gastroenterology, Emergency General Hospital, 100028, Beijing, China
| | - Yunhao Song
- State Key Laboratory of Membrane Biology, School of Medicine, Center for Synthetic and Systems Biology, Tsinghua University, 100084, Beijing, China
| | - Lidan Ding
- State Key Laboratory of Membrane Biology, School of Medicine, Center for Synthetic and Systems Biology, Tsinghua University, 100084, Beijing, China
| | - Bingtao Zhu
- State Key Laboratory of Membrane Biology, School of Medicine, Center for Synthetic and Systems Biology, Tsinghua University, 100084, Beijing, China
| | - Wanli Zhai
- State Key Laboratory of Membrane Biology, School of Medicine, Center for Synthetic and Systems Biology, Tsinghua University, 100084, Beijing, China
| | - Xuning Wang
- Department of Gastroenterology, Chinese PLA General Hospital, 100700, Beijing, China
| | - Yanshen Kuang
- Department of Gastroenterology, Chinese PLA General Hospital, 100700, Beijing, China
| | - Fangli Ren
- State Key Laboratory of Membrane Biology, School of Medicine, Center for Synthetic and Systems Biology, Tsinghua University, 100084, Beijing, China
| | - Baoqing Jia
- Department of Gastroenterology, Chinese PLA General Hospital, 100700, Beijing, China
| | - Wei Wu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Xiongjun Ye
- Urology and Lithotripsy Center, Peking University People's Hospital, 100034, Beijing, China.
| | - Yinyin Wang
- State Key Laboratory of Membrane Biology, School of Medicine, Center for Synthetic and Systems Biology, Tsinghua University, 100084, Beijing, China.
| | - Zhijie Chang
- State Key Laboratory of Membrane Biology, School of Medicine, Center for Synthetic and Systems Biology, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
10
|
Yang L, Yang H, Chu Y, Song Y, Ding L, Zhu B, Zhai W, Wang X, Kuang Y, Ren F, Jia B, Wu W, Ye X, Wang Y, Chang Z. CREPT is required for murine stem cell maintenance during intestinal regeneration. Nat Commun 2021. [DOI: 10.1038/s41467-020-20636-9 order by 38439--] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
AbstractIntestinal stem cells (ISCs) residing in the crypts are critical for the continual self-renewal and rapid recovery of the intestinal epithelium. The regulatory mechanism of ISCs is not fully understood. Here we report that CREPT, a recently identified tumor-promoting protein, is required for the maintenance of murine ISCs. CREPT is preferably expressed in the crypts but not in the villi. Deletion of CREPT in the intestinal epithelium of mice (Vil-CREPTKO) results in lower body weight and slow migration of epithelial cells in the intestine. Vil-CREPTKO intestine fails to regenerate after X-ray irradiation and dextran sulfate sodium (DSS) treatment. Accordingly, the deletion of CREPT decreases the expression of genes related to the proliferation and differentiation of ISCs and reduces Lgr5+ cell numbers at homeostasis. We identify that CREPT deficiency downregulates Wnt signaling by impairing β-catenin accumulation in the nucleus of the crypt cells during regeneration. Our study provides a previously undefined regulator of ISCs.
Collapse
|
11
|
Moloudizargari M, Moradkhani F, Hekmatirad S, Fallah M, Asghari MH, Reiter RJ. Therapeutic targets of cancer drugs: Modulation by melatonin. Life Sci 2020; 267:118934. [PMID: 33385405 DOI: 10.1016/j.lfs.2020.118934] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/27/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022]
Abstract
The biological functions of melatonin range beyond the regulation of the circadian rhythm. With regard to cancer, melatonin's potential to suppress cancer initiation, progression, angiogenesis and metastasis as well as sensitizing malignant cells to conventional chemo- and radiotherapy are among its most interesting effects. The targets at which melatonin initiates its anti-cancer effects are in common with those of a majority of existing anti-cancer agents, giving rise to the notion that this molecule is a pleiotropic agent sharing many features with other antineoplastic drugs in terms of their mechanisms of action. Among these common mechanisms of action are the regulation of several major intracellular pathways including mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase (ERK) and protein kinase B (AKT/PKB) signaling. The important mediators affected by melatonin include cyclins, nuclear factor-κB (NF-κB), heat shock proteins (HSPs) and c-Myc, all of which can serve as potential targets for cancer drugs. Melatonin also exerts some of its anti-cancer effects via inducing epigenetic modifications, DNA damage and mitochondrial disruption in malignant cells. The regulation of these mediators by melatonin mitigates tumor growth and invasiveness via modulating their downstream responsive genes, housekeeping enzymes, telomerase reverse transcriptase, apoptotic gene expression, angiogenic factors and structural proteins involved in metastasis. Increasing our knowledge on how melatonin affects its target sites will help find ways of exploiting the beneficial effects of this ubiquitously-acting molecule in cancer therapy. Acknowledging this, here we reviewed the most studied target pathways attributed to the anti-cancer effects of melatonin, highlighting their therapeutic potential.
Collapse
Affiliation(s)
- Milad Moloudizargari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moradkhani
- Department of Medical Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Hekmatirad
- Department of Pharmacology and Toxicology, School of Medicine, Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Marjan Fallah
- Medicinal Plant Research Centre, Faculty of Pharmacy, Ayatollah Amoli Branch, Islamic Azad University, Amol, Iran
| | - Mohammad Hossein Asghari
- Department of Pharmacology and Toxicology, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health, San Antonio, TX, USA.
| |
Collapse
|
12
|
Manivannan A, Lee ES, Han K, Lee HE, Kim DS. Versatile Nutraceutical Potentials of Watermelon-A Modest Fruit Loaded with Pharmaceutically Valuable Phytochemicals. Molecules 2020; 25:E5258. [PMID: 33187365 PMCID: PMC7698065 DOI: 10.3390/molecules25225258] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 11/16/2022] Open
Abstract
Watermelon (Citrulus lantus) is an important horticultural crop which belongs to the Curcubitaceae family. The nutraceutical potential of watermelon has been illustrated by several researchers, which makes it a better choice of functional food. Watermelon has been used to treat various ailments, such as cardio-vascular diseases, aging related ailments, obesity, diabetes, ulcers, and various types of cancers. The medicinal properties of watermelon are attributed by the presence of important phytochemicals with pharmaceutical values such as lycopene, citrulline, and other polyphenolic compounds. Watermelon acts as vital source of l-citrulline, a neutral-alpha amino acid which is the precursor of l-arginine, an essential amino acid necessary for protein synthesis. Supplementation of l-citrulline and lycopene displayed numerous health benefits in in vitro and in vivo studies. Similarly, the dietary intake of watermelon has proven benefits as functional food in humans for weight management. Apart from the fruits, the extracts prepared from the seeds, sprouts, and leaves also evidenced medicinal properties. The present review provides a comprehensive overview of benefits of watermelon for the treatment of various ailments.
Collapse
Affiliation(s)
| | | | | | | | - Do-Sun Kim
- Vegetable Research Division, National Institute of Horticultural and Herbal Science, Rural Development Administration, Jeonju 55365, Korea; (A.M.); (E.-S.L.); (K.H.); (H.-E.L.)
| |
Collapse
|
13
|
Chen K, Jiao X, Ashton A, Di Rocco A, Pestell TG, Sun Y, Zhao J, Casimiro MC, Li Z, Lisanti MP, McCue PA, Shen D, Achilefu S, Rui H, Pestell RG. The membrane-associated form of cyclin D1 enhances cellular invasion. Oncogenesis 2020; 9:83. [PMID: 32948740 PMCID: PMC7501870 DOI: 10.1038/s41389-020-00266-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 08/22/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
The essential G1-cyclin, CCND1, is a collaborative nuclear oncogene that is frequently overexpressed in cancer. D-type cyclins bind and activate CDK4 and CDK6 thereby contributing to G1–S cell-cycle progression. In addition to the nucleus, herein cyclin D1 was also located in the cytoplasmic membrane. In contrast with the nuclear-localized form of cyclin D1 (cyclin D1NL), the cytoplasmic membrane-localized form of cyclin D1 (cyclin D1MEM) induced transwell migration and the velocity of cellular migration. The cyclin D1MEM was sufficient to induce G1–S cell-cycle progression, cellular proliferation, and colony formation. The cyclin D1MEM was sufficient to induce phosphorylation of the serine threonine kinase Akt (Ser473) and augmented extranuclear localized 17β-estradiol dendrimer conjugate (EDC)-mediated phosphorylation of Akt (Ser473). These studies suggest distinct subcellular compartments of cell cycle proteins may convey distinct functions.
Collapse
Affiliation(s)
- Ke Chen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Anthony Ashton
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Agnese Di Rocco
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Timothy G Pestell
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Jun Zhao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Mathew C Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA.,Dept of Science and Math, Abraham Baldwin Agricultural college, Tifton, GA, 31794, Georgia
| | - Zhiping Li
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
| | - Michael P Lisanti
- Biomedical Research Centre (BRC), Translational Medicine, School of Environment and Life Sciences, University of Salford, Manchester, United Kingdom
| | - Peter A McCue
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Duanwen Shen
- Departments of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA
| | - Samuel Achilefu
- Departments of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA.,Departments of Radiology, Washington University, St. Louis, MO, 63110, USA.,Departments of Biochemistry & Molecular Biophysics, Washington University, St. Louis, MO, 63110, USA
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA. .,The Wistar Cancer Center, Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
14
|
Palmer N, Kaldis P. Less-well known functions of cyclin/CDK complexes. Semin Cell Dev Biol 2020; 107:54-62. [PMID: 32386818 DOI: 10.1016/j.semcdb.2020.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/31/2022]
Abstract
Cyclin-dependent kinases (CDKs) are activated by cyclins, which play important roles in dictating the actions of CDK/cyclin complexes. Cyclin binding influences the substrate specificity of these complexes in addition to their susceptibility to inhibition or degradation. CDK/cyclin complexes are best known to promote cell cycle progression in the mitotic cell cycle but are also crucial for important cellular processes not strictly associated with cellular division. This chapter primarily explores the understudied topic of CDK/cyclin complex functionality during the DNA damage response. We detail how CDK/cyclin complexes perform dual roles both as targets of DNA damage checkpoint signaling as well as effectors of DNA repair. Additionally, we discuss the potential CDK-independent roles of cyclins in these processes and the impact of such roles in human diseases such as cancer. Our goal is to place the spotlight on these important functions of cyclins either acting as independent entities or within CDK/cyclin complexes which have attracted less attention in the past. We consider that this will be important for a more complete understanding of the intricate functions of cell cycle proteins in the DNA damage response.
Collapse
Affiliation(s)
- Nathan Palmer
- Institute of Molecular and Cell Biology (IMCB), A⁎STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore; National University of Singapore (NUS), Department of Biochemistry, Singapore, 117597, Republic of Singapore
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), A⁎STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore; National University of Singapore (NUS), Department of Biochemistry, Singapore, 117597, Republic of Singapore; Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Box 50332, SE-202 13, Malmö, Sweden.
| |
Collapse
|
15
|
Di Sante G, Pagé J, Jiao X, Nawab O, Cristofanilli M, Skordalakes E, Pestell RG. Recent advances with cyclin-dependent kinase inhibitors: therapeutic agents for breast cancer and their role in immuno-oncology. Expert Rev Anticancer Ther 2019; 19:569-587. [PMID: 31219365 PMCID: PMC6834352 DOI: 10.1080/14737140.2019.1615889] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 05/03/2019] [Indexed: 12/18/2022]
Abstract
Introduction: Collaborative interactions between several diverse biological processes govern the onset and progression of breast cancer. These processes include alterations in cellular metabolism, anti-tumor immune responses, DNA damage repair, proliferation, anti-apoptotic signals, autophagy, epithelial-mesenchymal transition, components of the non-coding genome or onco-mIRs, cancer stem cells and cellular invasiveness. The last two decades have revealed that each of these processes are also directly regulated by a component of the cell cycle apparatus, cyclin D1. Area covered: The current review is provided to update recent developments in the clinical application of cyclin/CDK inhibitors to breast cancer with a focus on the anti-tumor immune response. Expert opinion: The cyclin D1 gene encodes the regulatory subunit of a proline-directed serine-threonine kinase that phosphorylates several substrates. CDKs possess phosphorylation site selectivity, with the phosphate-acceptor residue preceding a proline. Several important proteins are substrates including all three retinoblastoma proteins, NRF1, GCN5, and FOXM1. Over 280 cyclin D3/CDK6 substrates have b\een identified. Given the diversity of substrates for cyclin/CDKs, and the altered thresholds for substrate phosphorylation that occurs during the cell cycle, it is exciting that small molecular inhibitors targeting cyclin D/CDK activity have encouraging results in specific tumors.
Collapse
Affiliation(s)
- Gabriele Di Sante
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, USA
| | - Jessica Pagé
- Xavier University School of Medicine, Woodbury, NY, USA
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, USA
| | - Omar Nawab
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, USA
- Xavier University School of Medicine, Woodbury, NY, USA
| | - Massimo Cristofanilli
- Department of Medicine-Hematology and Oncology, Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, USA
- Xavier University School of Medicine, Woodbury, NY, USA
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
16
|
Effects of watermelon powder supplementation on colitis in high-fat diet-fed and dextran sodium sulfate-treated rats. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
17
|
Harmsen S, Rogalla S, Huang R, Spaliviero M, Neuschmelting V, Hayakawa Y, Lee Y, Tailor Y, Toledo-Crow R, Kang JW, Samii JM, Karabeber H, Davis RM, White JR, van de Rijn M, Gambhir SS, Contag CH, Wang TC, Kircher MF. Detection of Premalignant Gastrointestinal Lesions Using Surface-Enhanced Resonance Raman Scattering-Nanoparticle Endoscopy. ACS NANO 2019; 13:1354-1364. [PMID: 30624916 PMCID: PMC6428194 DOI: 10.1021/acsnano.8b06808] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cancers of the gastrointestinal (GI) tract are among the most frequent and most lethal cancers worldwide. An important reason for this high mortality is that early disease is typically asymptomatic, and patients often present with advanced, incurable disease. Even in high-risk patients who routinely undergo endoscopic screening, lesions can be missed due to their small size or subtle appearance. Thus, current imaging approaches lack the sensitivity and specificity to accurately detect incipient GI tract cancers. Here we report our finding that a single dose of a high-sensitivity surface-enhanced resonance Raman scattering nanoparticle (SERRS-NP) enables reliable detection of precancerous GI lesions in animal models that closely mimic disease development in humans. Some of these animal models have not been used previously to evaluate imaging probes for early cancer detection. The studies were performed using a commercial Raman imaging system, a newly developed mouse Raman endoscope, and finally a clinically applicable Raman endoscope for larger animal studies. We show that this SERRS-NP-based approach enables robust detection of small, premalignant lesions in animal models that faithfully recapitulate human esophageal, gastric, and colorectal tumorigenesis. This method holds promise for much earlier detection of GI cancers than currently possible and could lead therefore to marked reduction of morbidity and mortality of these tumor types.
Collapse
Affiliation(s)
- Stefan Harmsen
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Pediatrics, Stanford University, Stanford, California 94305, United States
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Stephan Rogalla
- Department of Pediatrics, Stanford University, Stanford, California 94305, United States
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Ruimin Huang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Massimiliano Spaliviero
- Urology Service, Department of Surgery, Sidney Kimmel Center for Prostate and Urologic Cancers, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Volker Neuschmelting
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Neurosurgery, University Hospital Cologne, Cologne 50937, Germany
| | - Yoku Hayakawa
- Department of Medicine, Columbia University, New York, New York 10032, United States
| | - Yoomi Lee
- Department of Medicine, Columbia University, New York, New York 10032, United States
| | - Yagnesh Tailor
- Department of Medicine, Columbia University, New York, New York 10032, United States
| | - Ricardo Toledo-Crow
- Research Engineering Lab, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Jeon Woong Kang
- Laser Biomedical Research Center, G. R. Harrison Spectroscopy Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jason M. Samii
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Hazem Karabeber
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Ryan M. Davis
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Julie R. White
- Tri-Institutional Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University, and Weill Cornell Medical College, New York, New York 10065, United States
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Matt van de Rijn
- Department of Pathology, Stanford University, Stanford, California 94305, United States
| | - Sanjiv S. Gambhir
- Department of Radiology, Stanford University, Stanford, California 94305, United States
- Department of Bioengineering, Department of Materials Science & Engineering, Molecular Imaging Program at Stanford, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California 94305, United States
| | - Christopher H. Contag
- Department of Pediatrics, Stanford University, Stanford, California 94305, United States
- Department of Microbiology and Immunology, Stanford University, Stanford, California 94305, United States
- Institute of Quantitative Health Science and Engineering, Department of Biomedical Engineering, and Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824, United States
- Corresponding Authors., .,
| | - Timothy C. Wang
- Department of Medicine, Columbia University, New York, New York 10032, United States
- Corresponding Authors., .,
| | - Moritz F. Kircher
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
- Department of Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
- Department of Imaging, Dana-Farber Cancer Institute & Harvard Medical School, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
- Corresponding Authors., .,
| |
Collapse
|
18
|
Li Z, Jiao X, Di Sante G, Ertel A, Casimiro MC, Wang M, Katiyar S, Ju X, Klopfenstein DV, Tozeren A, Dampier W, Chepelev I, Jeltsch A, Pestell RG. Cyclin D1 integrates G9a-mediated histone methylation. Oncogene 2019; 38:4232-4249. [PMID: 30718920 PMCID: PMC6542714 DOI: 10.1038/s41388-019-0723-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 12/03/2018] [Accepted: 01/08/2019] [Indexed: 12/26/2022]
Abstract
Lysine methylation of histones and non-histone substrates by the SET domain containing protein lysine methyltransferase (KMT) G9a/EHMT2 governs transcription contributing to apoptosis, aberrant cell growth, and pluripotency. The positioning of chromosomes within the nuclear three-dimensional space involves interactions between nuclear lamina (NL) and the lamina-associated domains (LAD). Contact of individual LADs with the NL are dependent upon H3K9me2 introduced by G9a. The mechanisms governing the recruitment of G9a to distinct subcellular sites, into chromatin or to LAD, is not known. The cyclin D1 gene product encodes the regulatory subunit of the holoenzyme that phosphorylates pRB and NRF1 thereby governing cell-cycle progression and mitochondrial metabolism. Herein, we show that cyclin D1 enhanced H3K9 dimethylation though direct association with G9a. Endogenous cyclin D1 was required for the recruitment of G9a to target genes in chromatin, for G9a-induced H3K9me2 of histones, and for NL-LAD interaction. The finding that cyclin D1 is required for recruitment of G9a to target genes in chromatin and for H3K9 dimethylation, identifies a novel mechanism coordinating protein methylation.
Collapse
Affiliation(s)
- Zhiping Li
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, 3805 Old Easton Rd., Doylestown, PA, 18902, USA
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, 3805 Old Easton Rd., Doylestown, PA, 18902, USA
| | - Gabriele Di Sante
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, 3805 Old Easton Rd., Doylestown, PA, 18902, USA
| | - Adam Ertel
- Department of Cancer Biology, Thomas Jefferson University, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Mathew C Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, 3805 Old Easton Rd., Doylestown, PA, 18902, USA
| | - Min Wang
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, 3805 Old Easton Rd., Doylestown, PA, 18902, USA
| | - Sanjay Katiyar
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, 3805 Old Easton Rd., Doylestown, PA, 18902, USA
| | - Xiaoming Ju
- Department of Cancer Biology, Thomas Jefferson University, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - D V Klopfenstein
- Center for Integrated Bioinformatics, School of Biomedical Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Aydin Tozeren
- Center for Integrated Bioinformatics, School of Biomedical Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - William Dampier
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA, 19104, USA
| | - Iouri Chepelev
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Albert Jeltsch
- Department of Biochemistry, Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, Allmandring 31, D-70569, Stuttgart, Germany
| | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, 3805 Old Easton Rd., Doylestown, PA, 18902, USA. .,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 637551, Singapore.
| |
Collapse
|
19
|
Peng YG, Zhang L. Wedelolactone suppresses cell proliferation and migration through AKT and AMPK signaling in melanoma. J DERMATOL TREAT 2018; 30:389-395. [PMID: 30252545 DOI: 10.1080/09546634.2018.1527996] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Ya-Guang Peng
- Huaiyin District, Shandong Provincial Hospital affiliated to Shandong University, Jinan City, Shandong Province, China
| | - Li Zhang
- Huaiyin District, Shandong Provincial Hospital affiliated to Shandong University, Jinan City, Shandong Province, China
| |
Collapse
|
20
|
Yu S, Yin Y, Wang Q, Wang L. Dual gene deficient models of Apc Min/+ mouse in assessing molecular mechanisms of intestinal carcinogenesis. Biomed Pharmacother 2018; 108:600-609. [PMID: 30243094 DOI: 10.1016/j.biopha.2018.09.056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/08/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023] Open
Abstract
The ApcMin/+ mouse, carrying an inactivated allele of the adenomatous polyposis coli (Apc) gene, is a widely used animal model of human colorectal tumorigenesis. While crossed with other gene knockout or knock-in mice, these mice possess advantages in investigation of human intestinal tumorigenesis. Intestinal tumor pathogenesis involves multiple gene alterations; thus, various double gene deficiency models could provide novel insights into molecular mechanisms of tumor biology, as well as gene-gene interactions involved in intestinal tumor development and assessment of novel strategies for preventing and treating intestinal cancer. This review discusses approximately 100 double gene deficient mice and their associated intestinal tumor development and progression phenotypes. The dual gene knockouts based on the Apc mutation background consist of inflammation and immune-related, cell cycle-related, Wnt/β-catenin signaling-related, tumor growth factor (TGF)-signaling-related, drug metabolism-related, and transcription factor genes, as well as some oncogenes and tumor suppressors. Future studies should focus on conditional or inducible dual or multiple mouse gene knockout models to investigate the molecular mechanisms underlying intestinal tumor development, as well as potential drug targets.
Collapse
Affiliation(s)
- Shuwen Yu
- Department of Pharmacy, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| | - Yanhui Yin
- Department of Pharmacy, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Qian Wang
- Department of Pharmacy, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Lu Wang
- Department of Pharmacy, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| |
Collapse
|
21
|
Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation. Proc Natl Acad Sci U S A 2018; 115:E6760-E6769. [PMID: 29967145 DOI: 10.1073/pnas.1800621115] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Zyxin is a member of the focal adhesion complex and plays a critical role in actin filament polymerization and cell motility. Several recent studies showed that Zyxin is a positive regulator of Yki/YAP (Yes-associated protein) signaling. However, little is known about the mechanisms by which Zyxin itself is regulated and how Zyxin affects Hippo-YAP activity. We first showed that Zyxin is phosphorylated by CDK1 during mitosis. Depletion of Zyxin resulted in significantly impaired colon cancer cell proliferation, migration, anchorage-independent growth, and tumor formation in xenograft animal models. Mitotic phosphorylation is required for Zyxin activity in promoting growth. Zyxin regulates YAP activity through the colon cancer oncogene CDK8. CDK8 knockout phenocopied Zyxin knockdown in colon cancer cells, while ectopic expression of CDK8 substantially restored the tumorigenic defects of Zyxin-depletion cells. Mechanistically, we showed that CDK8 directly phosphorylated YAP and promoted its activation. Fully activated YAP is required to support the growth in CDK8-knockout colon cancer cells in vitro and in vivo. Together, these observations suggest that Zyxin promotes colon cancer tumorigenesis in a mitotic-phosphorylation-dependent manner and through CDK8-mediated YAP activation.
Collapse
|
22
|
Russo I, Carrizzo A, Bochicchio S, Piazza O, Lamberti G, Barba AA, Vecchione C, Zeppa P, Iovino P, Bucci C, Santonicola A, Ciacci C. siRNA Delivery for Control of Cyclin D1 and E2F1 Expression in Crohn's Disease. Transl Med UniSa 2018; 17:25-33. [PMID: 30083520 PMCID: PMC6067069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Evidence in inflammatory bowel diseases (IBD) supports a connection between inflammation and cancer due to the alteration of the cell cycle with loss of control at the G1/S checkpoint. In this study, we analyze the expression and modulation of CyD1 and E2F1 in colon explants from Crohn's disease (CD) patients. We used ex vivo culture of colon explants from 4 CD patients and 2 healthy controls, stimulated with lipopolysaccharide from Escherichia Coli (EC-LPS). Commercial siRNAs for CyD1 and E2F1 inhibition were encapsulated in Invivofectamine® and in purposely produced nanoliposomal vectors to silencing CyD1 and E2F1 expression. Western blot analysis was used to investigate the effect of siRNA on CyD1, E2F1 and cyclooxygenase 2 (COX-2) expression. In CD patients colon explants, CyD1 and E2F1 increased after the inflammatory stimulus but siRNA silencing attenuated their expression and controlled the COX-2 expression too. These data represent a prelimiary exploration of in vitro siRNA use.
Collapse
Affiliation(s)
- Ilaria Russo
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Albino Carrizzo
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Sabrina Bochicchio
- Department of Industrial Engineering, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) – ITALY,Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) - ITALY
| | - Ornella Piazza
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Gaetano Lamberti
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) - ITALY
| | - Anna Angela Barba
- Department of Industrial Engineering, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) – ITALY
| | - Carmine Vecchione
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Pio Zeppa
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Paola Iovino
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Cristina Bucci
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Antonella Santonicola
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Carolina Ciacci
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| |
Collapse
|
23
|
Russo I, Carrizzo A, Bochicchio S, Piazza O, Lamberti G, Barba AA, Vecchione C, Zeppa P, Iovino P, Bucci C, Santonicola A, Ciacci C. siRNA Delivery for Control of Cyclin D1 and E2F1 Expression in Crohn's Disease. Transl Med UniSa 2018; 17:22-30. [PMID: 30050877 PMCID: PMC6056255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Evidence in inflammatory bowel diseases (IBD) supports a connection between inflammation and cancer due to the alteration of the cell cycle with loss of control at the G1/S checkpoint. In this study, we analyze the expression and modulation of CyD1 and E2F1 in colon explants from Crohn's disease (CD) patients. We used ex vivo culture of colon explants from 4 CD patients and 2 healthy controls, stimulated with lipopolysaccharide from Escherichia Coli (EC-LPS). Commercial siRNAs for CyD1 and E2F1 inhibition were encapsulated in Invivofectamine® and in purposely produced nanoliposomal vectors to silencing CyD1 and E2F1 expression. Western blot analysis was used to investigate the effect of siRNA on CyD1, E2F1 and cyclooxygenase 2 (COX-2) expression. In CD patients colon explants, CyD1 and E2F1 increased after the inflammatory stimulus but siRNA silencing attenuated their expression and controlled the COX-2 expression too. These data represent a prelimiary exploration of in vitro siRNA use.
Collapse
Affiliation(s)
- Ilaria Russo
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Albino Carrizzo
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Sabrina Bochicchio
- Department of Industrial Engineering, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) – ITALY,Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) - ITALY
| | - Ornella Piazza
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Gaetano Lamberti
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) - ITALY
| | - Anna Angela Barba
- Department of Industrial Engineering, University of Salerno, via Giovanni Paolo II, 132, Fisciano (SA) – ITALY
| | - Carmine Vecchione
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Pio Zeppa
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Paola Iovino
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Cristina Bucci
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Antonella Santonicola
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Carolina Ciacci
- Department of Medicine and Surgery, Scuola Medica Salernitana, University of Salerno, 84081, Baronissi (SA), Italy, IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| |
Collapse
|
24
|
Wang G, Gormley M, Qiao J, Zhao Q, Wang M, Di Sante G, Deng S, Dong L, Pestell T, Ju X, Casimiro MC, Addya S, Ertel A, Tozeren A, Li Q, Yu Z, Pestell RG. Cyclin D1-mediated microRNA expression signature predicts breast cancer outcome. Theranostics 2018; 8:2251-2263. [PMID: 29721077 PMCID: PMC5928887 DOI: 10.7150/thno.23877] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/25/2017] [Indexed: 01/03/2023] Open
Abstract
Background: Genetic classification of breast cancer based on the coding mRNA suggests the evolution of distinct subtypes. Whether the non-coding genome is altered concordantly with the coding genome and the mechanism by which the cell cycle directly controls the non-coding genome is poorly understood. Methods: Herein, the miRNA signature maintained by endogenous cyclin D1 in human breast cancer cells was defined. In order to determine the clinical significance of the cyclin D1-mediated miRNA signature, we defined a miRNA expression superset from 459 breast cancer samples. We compared the coding and non-coding genome of breast cancer subtypes. Results: Hierarchical clustering of human breast cancers defined four distinct miRNA clusters (G1-G4) associated with distinguishable relapse-free survival by Kaplan-Meier analysis. The cyclin D1-regulated miRNA signature included several oncomirs, was conserved in multiple breast cancer cell lines, was associated with the G2 tumor miRNA cluster, ERα+ status, better outcome and activation of the Wnt pathway. The coding and non-coding genome were discordant within breast cancer subtypes. Seed elements for cyclin D1-regulated miRNA were identified in 63 genes of the Wnt signaling pathway including DKK. Cyclin D1 restrained DKK1 via the 3'UTR. In vivo studies using inducible transgenics confirmed cyclin D1 induces Wnt-dependent gene expression. Conclusion: The non-coding genome defines breast cancer subtypes that are discordant with their coding genome subtype suggesting distinct evolutionary drivers within the tumors. Cyclin D1 orchestrates expression of a miRNA signature that induces Wnt/β-catenin signaling, therefore cyclin D1 serves both upstream and downstream of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Guangxue Wang
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Michael Gormley
- Department of Cancer Biology, Thomas Jefferson University, 233 South 10 th St. Philadelphia PA 19107
| | - Jing Qiao
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Qian Zhao
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Min Wang
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center and Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Suite, 222, Wynnewood, PA. 19096
| | - Gabriele Di Sante
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center and Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Suite, 222, Wynnewood, PA. 19096
| | - Shengqiong Deng
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Shanghai Gongli Hospital, the Second Military Medical University, Shanghai 200120, China
| | - Lin Dong
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Tim Pestell
- Department of Cancer Biology, Thomas Jefferson University, 233 South 10 th St. Philadelphia PA 19107
| | - Xiaoming Ju
- Department of Cancer Biology, Thomas Jefferson University, 233 South 10 th St. Philadelphia PA 19107
| | - Mathew C. Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center and Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Suite, 222, Wynnewood, PA. 19096
| | - Sankar Addya
- Department of Cancer Biology, Thomas Jefferson University, 233 South 10 th St. Philadelphia PA 19107
| | - Adam Ertel
- Department of Cancer Biology, Thomas Jefferson University, 233 South 10 th St. Philadelphia PA 19107
| | - Ayden Tozeren
- Center for Integrated Bioinformatics, Drexel University, Philadelphia, PA 19104
- School of Biomedical Engineering, Systems and Health Sciences, Drexel University, Philadelphia, PA 19104
| | - Qinchuan Li
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zuoren Yu
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center and Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Suite, 222, Wynnewood, PA. 19096
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Richard G. Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center and Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Suite, 222, Wynnewood, PA. 19096
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637551, Singapore
| |
Collapse
|
25
|
Bong YS, Assefnia S, Tuohy T, Neklason DW, Burt RW, Ahn J, Bueno De Mesquita PJ, Byers SW. A role for the vitamin D pathway in non-intestinal lesions in genetic and carcinogen models of colorectal cancer and in familial adenomatous polyposis. Oncotarget 2018; 7:80508-80520. [PMID: 27768599 PMCID: PMC5348337 DOI: 10.18632/oncotarget.12768] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 10/11/2016] [Indexed: 12/14/2022] Open
Abstract
Vitamin D is implicated in the etiology of cancers of the gastrointestinal tract, usually characterized by alteration in the APC/β-catenin/TCF tumor suppressor pathway. The vitamin D receptor (VDR) is also implicated in cardiovascular and skin diseases as well as in immunity. Activated VDR can indirectly alter β-catenin nuclear localization and directly suppress β-catenin/TCF mediated transcriptional activity. We treated VDR null mice with the carcinogen azoxymethane (AOM) and generated mice bearing a mutated APC (hypomorph) on a VDR null background (Apc1638N/+Vdr−/−). VDR null mice do not develop GI or extra-colonic tumors but loss of VDR decreased intestinal tumor latency and increased progression to adenocarcinoma in both models. AOM treatment of VDR null mice also caused squamous cell carcinoma of the anus. Although levels and distribution of total or activated β-catenin in the epithelial component of tumors were unaffected by loss of VDR, β-catenin dependent cyclin D1 expression was affected suggesting a direct VDR effect on β-catenin co-activator activity. Extra-colonic mucosa manifestations in Apc1638N/+Vdr−/− animals included increased nuclear β-catenin in submucosal stromal cells, spleno- and cardiomegaly and large epidermoid cysts characteristic of the FAP variant, Gardner's syndrome. Consistent with this, SNPs in the VDR, vitamin D binding protein and CYP24 as well as mutations in APC distal to codon 850 were strongly associated with Gardners syndrome in a cohort of 457 FAP patients, This work suggests that alterations in the vitamin D/VDR axis are important in Gardner's syndrome, as well as in the etiology of anal cancer.
Collapse
Affiliation(s)
- Yong-Sik Bong
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, DC, United States of America
| | - Shahin Assefnia
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, DC, United States of America
| | - Therese Tuohy
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Deborah W Neklason
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Randall W Burt
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Jaeil Ahn
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University School of Medicine, Washington, DC, United States of America
| | - Paul J Bueno De Mesquita
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, DC, United States of America
| | - Stephen W Byers
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University School of Medicine, Washington, DC, United States of America
| |
Collapse
|
26
|
Pestell TG, Jiao X, Kumar M, Peck AR, Prisco M, Deng S, Li Z, Ertel A, Casimiro MC, Ju X, Di Rocco A, Di Sante G, Katiyar S, Shupp A, Lisanti MP, Jain P, Wu K, Rui H, Hooper DC, Yu Z, Goldman AR, Speicher DW, Laury-Kleintop L, Pestell RG. Stromal cyclin D1 promotes heterotypic immune signaling and breast cancer growth. Oncotarget 2017; 8:81754-81775. [PMID: 29137220 PMCID: PMC5669846 DOI: 10.18632/oncotarget.19953] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/09/2017] [Indexed: 12/28/2022] Open
Abstract
The cyclin D1 gene encodes the regulatory subunit of a holoenzyme that drives cell autonomous cell cycle progression and proliferation. Herein we show cyclin D1 abundance is increased >30-fold in the stromal fibroblasts of patients with invasive breast cancer, associated with poor outcome. Cyclin D1 transformed hTERT human fibroblast to a cancer-associated fibroblast phenotype. Stromal fibroblast expression of cyclin D1 (cyclin D1Stroma) in vivo, enhanced breast epithelial cancer tumor growth, restrained apoptosis, and increased autophagy. Cyclin D1Stroma had profound effects on the breast tumor microenvironment increasing the recruitment of F4/80+ and CD11b+ macrophages and increasing angiogenesis. Cyclin D1Stroma induced secretion of factors that promoted expansion of stem cells (breast stem-like cells, embryonic stem cells and bone marrow derived stem cells). Cyclin D1Stroma resulted in increased secretion of proinflammatory cytokines (CCL2, CCL7, CCL11, CXCL1, CXCL5, CXCL9, CXCL12), CSF (CSF1, GM-CSF1) and osteopontin (OPN) (30-fold). OPN was induced by cyclin D1 in fibroblasts, breast epithelial cells and in the murine transgenic mammary gland and OPN was sufficient to induce stem cell expansion. These results demonstrate that cyclin D1Stroma drives tumor microenvironment heterocellular signaling, promoting several key hallmarks of cancer.
Collapse
Affiliation(s)
- Timothy G Pestell
- Departments of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA, USA
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, USA
| | - Mukesh Kumar
- Departments of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA, USA
| | - Amy R Peck
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Marco Prisco
- Departments of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA, USA
| | - Shengqiong Deng
- Departments of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA, USA.,Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiping Li
- Departments of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA, USA
| | - Adam Ertel
- Departments of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA, USA
| | - Mathew C Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, USA
| | - Xiaoming Ju
- Departments of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA, USA
| | - Agnese Di Rocco
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, USA
| | - Gabriele Di Sante
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, USA
| | - Sanjay Katiyar
- Departments of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA, USA
| | - Alison Shupp
- Departments of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA, USA
| | - Michael P Lisanti
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre, University of Salford, Salford, Greater Manchester, England, UK
| | - Pooja Jain
- Department of Microbiology and Immunology, Institute for Molecular Medicine & Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Douglas C Hooper
- Department of Microbiology, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA, USA
| | - Zuoren Yu
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, USA.,Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Aaron R Goldman
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | - David W Speicher
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | | | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, USA.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
27
|
Reelin protects from colon pathology by maintaining the intestinal barrier integrity and repressing tumorigenic genes. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2126-2134. [PMID: 28572005 DOI: 10.1016/j.bbadis.2017.05.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/15/2017] [Accepted: 05/29/2017] [Indexed: 12/17/2022]
Abstract
We previously reported that reelin, an extracellular matrix protein first known for its key role in neuronal migration, reduces the susceptibility to dextran sulphate sodium (DSS)-colitis. The aim of the current study was to determine whether reelin protects from colorectal cancer and how reelin defends from colon pathology. In the colon of wild-type and of mice lacking reelin (reeler mice) we have analysed the: i) epithelium cell renewal processes, ii) morphology, iii) Sox9, Cdx2, Smad5, Cyclin D1, IL-6 and IFNγ mRNA abundance in DSS-treated and untreated mice, and iv) development of azoxymethane/DSS-induced colorectal cancer, using histological and real time-PCR methodologies. The reeler mutation increases colitis-associated tumorigenesis, with increased tumours number and size. It also impairs the intestinal barrier because it reduces cell proliferation, migration, differentiation and apoptosis; decreases the number and maturation of goblet cells, and expands the intercellular space of the desmosomes. The intestinal barrier impairment might explain the increased susceptibility to colon pathology exhibited by the reeler mice and is at least mediated by the down-regulation of Sox9 and Cdx2. In response to DSS-colitis, the reeler colon increases the mRNA abundance of IL-6, Smad5 and Cyclin D1 and decreases that of IFNγ, conditions that might result in the increased colitis-associated tumorigenesis found in the reeler mice. In conclusion, the results highlight a role for reelin in maintaining intestinal epithelial cell homeostasis and providing resistance against colon pathology.
Collapse
|
28
|
Abstract
Cancer is characterized by uncontrolled tumour cell proliferation resulting from aberrant activity of various cell cycle proteins. Therefore, cell cycle regulators are considered attractive targets in cancer therapy. Intriguingly, animal models demonstrate that some of these proteins are not essential for proliferation of non-transformed cells and development of most tissues. By contrast, many cancers are uniquely dependent on these proteins and hence are selectively sensitive to their inhibition. After decades of research on the physiological functions of cell cycle proteins and their relevance for cancer, this knowledge recently translated into the first approved cancer therapeutic targeting of a direct regulator of the cell cycle. In this Review, we focus on proteins that directly regulate cell cycle progression (such as cyclin-dependent kinases (CDKs)), as well as checkpoint kinases, Aurora kinases and Polo-like kinases (PLKs). We discuss the role of cell cycle proteins in cancer, the rationale for targeting them in cancer treatment and results of clinical trials, as well as the future therapeutic potential of various cell cycle inhibitors.
Collapse
Affiliation(s)
- Tobias Otto
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
29
|
Badenes M, Trindade A, Pissarra H, Lopes-da-Costa L, Duarte A. Delta-like 4/Notch signaling promotes Apc Min/+ tumor initiation through angiogenic and non-angiogenic related mechanisms. BMC Cancer 2017; 17:50. [PMID: 28086833 PMCID: PMC5237288 DOI: 10.1186/s12885-016-3036-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 12/27/2016] [Indexed: 01/27/2023] Open
Abstract
Background Delta like 4 (Dll4)/Notch signaling is a key regulator of tumor angiogenesis. Additionally, the role of Dll4 has been studied on tumor stem cells. However, as these cells are implicated in tumor angiogenesis, it is conceivable that the effect of Dll4 on these cells may be a consequence of its angiogenic function. Our aim was to evaluate the expression and dissect the functions of Dll4 in the ApcMin/+ model of colorectal cancer. Methods We evaluated the protein expression pattern of Dll4 and other Notch members in the ApcMin/+ tumors relatively to the normal gut and compared endothelial-specific with ubiquitous Dll4 knockout mice on an ApcMin/+ background. Results All Notch pathway members were present in the normal small and large intestine and in the adenomas of the same regions. Dll4, all Notch receptors and Hes1 expression seemed upregulated in the tumors, with some regional differences. The same members and Hes5, instead of Hes1, presented ectopic expression in the tumor parenchyma. Dll4 expression was most pronounced in the tumor cells but it was also present in the tumor blood vessels and in other stromal cells. Ubiquitous and endothelial-specific Dll4 deletion led to an equivalent reduction of tumor growth because of a similarly marked tumoral angiogenic phenotype promoting non-productive vasculature and consequently hypoxia and apoptosis. The ubiquitous Dll4 inhibition led to a stronger decrease of tumor multiplicity than the endothelial-specific deletion by further reducing tumor proliferation and tumor stem cell density through upregulation of the cyclin-dependent kinase inhibitors 1C and 1B and downregulation of Myc, Cyclin D1 and D2 independently of β-catenin activation. This phenotype was associated to the observed increased epithelial differentiation deviated towards the secretory lineages by Atoh1 and Klf4 upregulation only in the ubiquitous Dll4 mutants. Conclusions Dll4 seems to promote ApcMin/+ tumorigenesis through both angiogenic and non-angiogenic related mechanisms. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-3036-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marina Badenes
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - Alexandre Trindade
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - Hugo Pissarra
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - Luís Lopes-da-Costa
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - António Duarte
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal.
| |
Collapse
|
30
|
Ju X, Jiao X, Ertel A, Casimiro MC, Di Sante G, Deng S, Li Z, Di Rocco A, Zhan T, Hawkins A, Stoyanova T, Andò S, Fatatis A, Lisanti MP, Gomella LG, Languino LR, Pestell RG. v-Src Oncogene Induces Trop2 Proteolytic Activation via Cyclin D1. Cancer Res 2016; 76:6723-6734. [PMID: 27634768 DOI: 10.1158/0008-5472.can-15-3327] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 07/18/2016] [Accepted: 08/18/2016] [Indexed: 01/06/2023]
Abstract
Proteomic analysis of castration-resistant prostate cancer demonstrated the enrichment of Src tyrosine kinase activity in approximately 90% of patients. Src is known to induce cyclin D1, and a cyclin D1-regulated gene expression module predicts poor outcome in human prostate cancer. The tumor-associated calcium signal transducer 2 (TACSTD2/Trop2/M1S1) is enriched in the prostate, promoting prostate stem cell self-renewal upon proteolytic activation via a γ-secretase cleavage complex (PS1, PS2) and TACE (ADAM17), which releases the Trop2 intracellular domain (Trop2 ICD). Herein, v-Src transformation of primary murine prostate epithelial cells increased the proportion of prostate cancer stem cells as characterized by gene expression, epitope characteristics, and prostatosphere formation. Cyclin D1 was induced by v-Src, and Src kinase induction of Trop2 ICD nuclear accumulation required cyclin D1. Cyclin D1 induced abundance of the Trop2 proteolytic cleavage activation components (PS2, TACE) and restrained expression of the inhibitory component of the Trop2 proteolytic complex (Numb). Patients with prostate cancer with increased nuclear Trop2 ICD and cyclin D1, and reduced Numb, had reduced recurrence-free survival probability (HR = 4.35). Cyclin D1, therefore, serves as a transducer of v-Src-mediated induction of Trop2 ICD by enhancing abundance of the Trop2 proteolytic activation complex. Cancer Res; 76(22); 6723-34. ©2016 AACR.
Collapse
Affiliation(s)
- Xiaoming Ju
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Xuanmao Jiao
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Adam Ertel
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Mathew C Casimiro
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Gabriele Di Sante
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Shengqiong Deng
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Zhiping Li
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Agnese Di Rocco
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Tingting Zhan
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Adam Hawkins
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Tanya Stoyanova
- Department of Microbiology, Immunology, and Molecular Genetics University of California, Los Angeles, California
| | - Sebastiano Andò
- Faculty of Pharmacy, Nutrition, and Health Science, University of Calabria, Arcavacata, Rende CS, Italy
| | - Alessandro Fatatis
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Pharmacology and Physiology and Laboratory Medicine, Drexel University, Philadelphia, Pennsylvania
| | - Michael P Lisanti
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Stem Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Leonard G Gomella
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Lucia R Languino
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Richard G Pestell
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. .,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
31
|
Li Y, Lauriola M, Kim D, Francesconi M, D'Uva G, Shibata D, Malafa MP, Yeatman TJ, Coppola D, Solmi R, Cheng JQ. Adenomatous polyposis coli (APC) regulates miR17-92 cluster through β-catenin pathway in colorectal cancer. Oncogene 2016; 35:4558-4568. [PMID: 26804172 PMCID: PMC4960006 DOI: 10.1038/onc.2015.522] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 11/09/2015] [Accepted: 11/30/2015] [Indexed: 02/06/2023]
Abstract
Adenomatous polyposis coli (APC) mutation is the most common genetic change in sporadic colorectal cancer (CRC). Although deregulations of miRNAs have been frequently reported in this malignancy, APC-regulated miRNAs have not been extensively documented. Here, by using an APC-inducible cell line and array analysis, we identified a total of 26 deregulated miRNAs. Among them, members of miR-17-92 cluster were dramatically inhibited by APC and induced by enforced expression of β-catenin. Furthermore, we demonstrate that activated β-catenin resulted from APC loss binds to and activates the miR-17-92 promoter. Notably, enforced expression of miR-19a overrides APC tumor suppressor activity, and knockdown of miR-19a in cancer cells with compromised APC function reduced their aggressive features in vitro. Finally, we observed that expression of miR-19a significantly correlates with β-catenin levels in colorectal cancer specimens, and it is associated to the aggressive stage of tumor progression. Thus, our study reveals that miR-17-92 cluster is directly regulated by APC/β-catenin pathway and could be a potential therapeutic target in colon cancers with aberrant APC/β-catenin signaling.
Collapse
Affiliation(s)
- Yajuan Li
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Mattia Lauriola
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, via Massarenti, 9, 40138 Bologna, Italy
| | - Donghwa Kim
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Mirko Francesconi
- EMBL-CRG Systems Biology Unit, Centre for Genomic Regulation (CRG), Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Gabriele D'Uva
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, via Massarenti, 9, 40138 Bologna, Italy
| | - Dave Shibata
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Mokenge P Malafa
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Timothy J Yeatman
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Domenico Coppola
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Rossella Solmi
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, via Massarenti, 9, 40138 Bologna, Italy
| | - Jin Q Cheng
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
32
|
Kinase-independent role of cyclin D1 in chromosomal instability and mammary tumorigenesis. Oncotarget 2016; 6:8525-38. [PMID: 25940700 PMCID: PMC4496164 DOI: 10.18632/oncotarget.3267] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 01/31/2015] [Indexed: 01/26/2023] Open
Abstract
Cyclin D1 is an important molecular driver of human breast cancer but better understanding of its oncogenic mechanisms is needed, especially to enhance efforts in targeted therapeutics. Currently, pharmaceutical initiatives to inhibit cyclin D1 are focused on the catalytic component since the transforming capacity is thought to reside in the cyclin D1/CDK activity. We initiated the following study to directly test the oncogenic potential of catalytically inactive cyclin D1 in an in vivo mouse model that is relevant to breast cancer. Herein, transduction of cyclin D1−/− mouse embryonic fibroblasts (MEFs) with the kinase dead KE mutant of cyclin D1 led to aneuploidy, abnormalities in mitotic spindle formation, autosome amplification, and chromosomal instability (CIN) by gene expression profiling. Acute transgenic expression of either cyclin D1WT or cyclin D1KE in the mammary gland was sufficient to induce a high CIN score within 7 days. Sustained expression of cyclin D1KE induced mammary adenocarcinoma with similar kinetics to that of the wild-type cyclin D1. ChIP-Seq studies demonstrated recruitment of cyclin D1WT and cyclin D1KE to the genes governing CIN. We conclude that the CDK-activating function of cyclin D1 is not necessary to induce either chromosomal instability or mammary tumorigenesis.
Collapse
|
33
|
Casimiro MC, Di Sante G, Ju X, Li Z, Chen K, Crosariol M, Yaman I, Gormley M, Meng H, Lisanti MP, Pestell RG. Cyclin D1 Promotes Androgen-Dependent DNA Damage Repair in Prostate Cancer Cells. Cancer Res 2015; 76:329-38. [PMID: 26582866 DOI: 10.1158/0008-5472.can-15-0999] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 10/08/2015] [Indexed: 11/16/2022]
Abstract
Therapy resistance and poor outcome in prostate cancer is associated with increased expression of cyclin D1. Androgens promote DNA double-strand break repair to reduce DNA damage, and cyclin D1 was also shown to enhance DNA damage repair (DDR). In this study, we investigated the significance of cyclin D1 in androgen-induced DDR using established prostate cancer cells and prostate tissues from cyclin D1 knockout mice. We demonstrate that endogenous cyclin D1 further diminished the dihydrotestosterone (DHT)-dependent reduction of γH2AX foci in vitro. We also show that cyclin D1 was required for the androgen-dependent DNA damage response both in vitro and in vivo. Furthermore, cyclin D1 was required for androgen-enhanced DDR and radioresistance of prostate cancer cells. Moreover, microarray analysis of primary prostate epithelial cells from cyclin D1-deficient and wild-type mice demonstrated that most of the DHT-dependent gene expression changes are also cyclin D1 dependent. Collectively, our findings suggest that the hormone-mediated recruitment of cyclin D1 to sites of DDR may facilitate the resistance of prostate cancer cells to DNA damage therapies and highlight the need to explore other therapeutic approaches in prostate cancer to prevent or overcome drug resistance.
Collapse
Affiliation(s)
- Mathew C Casimiro
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Gabriele Di Sante
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Xiaoming Ju
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Zhiping Li
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Ke Chen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Science and Technology, Tongji Hospital, Tongji Medical College, Huazhong University of Technology, Shanghai, P.R. China
| | - Marco Crosariol
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Ismail Yaman
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Michael Gormley
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Hui Meng
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Michael P Lisanti
- Department of Stem Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania. University of Manchester, Manchester Breast Center, Manchester, United Kingdom
| | - Richard G Pestell
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania. Kazan Federal University, Kazan, Republic of Tatarstan, Russia.
| |
Collapse
|
34
|
Dasgupta N, Kumar Thakur B, Ta A, Das S. Caveolin-1 is transcribed from a hypermethylated promoter to mediate colonocyte differentiation and apoptosis. Exp Cell Res 2015; 334:323-36. [DOI: 10.1016/j.yexcr.2015.03.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 11/28/2022]
|
35
|
Thomas SS, Makar KW, Li L, Zheng Y, Yang P, Levy L, Rudolph RY, Lampe PD, Yan M, Markowitz SD, Bigler J, Lampe JW, Potter JD. Tissue-specific patterns of gene expression in the epithelium and stroma of normal colon in healthy individuals in an aspirin intervention trial. BMC MEDICAL GENETICS 2015; 16:18. [PMID: 25927723 PMCID: PMC4422425 DOI: 10.1186/s12881-015-0161-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/26/2015] [Indexed: 12/21/2022]
Abstract
Background Regular aspirin use reduces colon adenoma and carcinoma incidence. UDP-glucuronosyltransferases (UGT) are involved in aspirin metabolism and clearance, and variant alleles in UGT1A6 have been shown to alter salicylic acid metabolism and risk of colon neoplasia. Methods In a randomized, cross-over, placebo-controlled trial of 44 healthy men and women, homozygous for UGT1A6*1 or UGT1A6*2, we explored differences between global epithelial and stromal expression, using Affymetrix U133 + 2.0 microarrays and tested effects of 60-day aspirin supplementation (325 mg/d) on epithelial and stromal gene expression and colon prostaglandin E2 (PGE2) levels. Results No statistically significant differences in gene expression were observed in response to aspirin or UGT1A6 genotype, but tissue PGE2 levels were lower with aspirin compared to placebo (p <0.001). Transcripts differentially expressed between epithelium and stroma (N = 4916, P <0.01, false discovery rate <0.001), included a high proportion of genes involved in cell signaling, cellular movement, and cancer. Genes preferentially expressed in epithelium were involved in drug and xenobiotic metabolism, fatty acid and lipid metabolism, apoptosis signaling, and ion transport. Genes preferentially expressed in stroma included those involved in inflammation, cellular adhesion, and extracellular matrix production. Wnt-Tcf4 pathway genes were expressed in both epithelium and stroma but differed by subcellular location. Conclusions These results suggest that, in healthy individuals, subtle effects of aspirin on gene expression in normal colon tissue are likely overwhelmed by inter-individual variability in microarray analyses. Differential expression of critical genes between colonic epithelium and stroma suggest that these tissue types need to be considered separately. Electronic supplementary material The online version of this article (doi:10.1186/s12881-015-0161-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sushma S Thomas
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Karen W Makar
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Lin Li
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Yingye Zheng
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Peiying Yang
- M.D. Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Lisa Levy
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | | | - Paul D Lampe
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Min Yan
- Case Western Reserve University School of Medicine, Cincinnati, OH, 44106, USA.
| | - Sanford D Markowitz
- Case Western Reserve University School of Medicine, Cincinnati, OH, 44106, USA.
| | | | - Johanna W Lampe
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - John D Potter
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| |
Collapse
|
36
|
Immunoexpression of cyclin D1 in colorectal carcinomas is not correlated with survival outcome. J Microsc Ultrastruct 2015; 3:62-67. [PMID: 30023183 PMCID: PMC6014192 DOI: 10.1016/j.jmau.2015.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 01/02/2015] [Indexed: 11/21/2022] Open
Abstract
Background Colon and colorectal cancer (CRC) research has entered a new era with recent updates of molecular events and prognostic markers. Among other prognostic markers, exaggerated expression of nuclear CCND1 has key role in tumour pathogenesis and metastases of CRC and has also been claimed to predict response to treatment. Objectives This study was designed to evaluate the prognostic and predictive value of CCND1 in CRC and the correlation of CCND1 expression with the different clinicopathological parameters. Methods Paraffin blocks from 117 primary CRC were retrieved from the archives of the Department of Pathology at King Abdulaziz University. Tissue microarrays were designed and constructed. The immunostaining of CCND1 was performed and analysed. Results There were more cases with low nuclear immunoexpression of CCND1 in both primary tumours and nodal metastasis (p <0.001). Cyclin D1 did not show association with clinicopathological features except with lymphovascular invasion. Low nuclear immunoexpression of CCND1 was associated with negative lymphovascular invasion (p = 0.046). There was no statistically significant correlation between CCND1 immunoexpression and survival probability (Log Rank = 2.474, p = 0.116). Conclusion Our study indicates that CCND1 immunoexpression cannot be used as a predictor of survival in CRC. It also shows no significant correlation with clinicopathological features except with lymphovascular invasion.
Collapse
|
37
|
Nuclear factor-kappaB sensitizes to benzyl isothiocyanate-induced antiproliferation in p53-deficient colorectal cancer cells. Cell Death Dis 2014; 5:e1534. [PMID: 25412312 PMCID: PMC4260753 DOI: 10.1038/cddis.2014.495] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 10/20/2014] [Accepted: 10/21/2014] [Indexed: 11/29/2022]
Abstract
Benzyl isothiocyanate (BITC), a dietary isothiocyanate derived from cruciferous vegetables, inhibits the proliferation of colorectal cancer cells, most of which overexpress β-catenin as a result of mutations in the genes for adenomatous polyposis coli or mutations in β-catenin itself. Because nuclear factor-κB (NF-κB) is a plausible target of BITC signaling in inflammatory cell models, we hypothesized that it is also involved in BITC-inhibited proliferation of colorectal cancer cells. siRNA-mediated knockdown of the NF-κB p65 subunit significantly decreased the BITC sensitivity of human colorectal cancer HT-29 cells with mutated p53 tumor suppressor protein. Treating HT-29 cells with BITC induced the phosphorylation of IκB kinase, IκB-α and p65, the degradation of IκB-α, the translocation of p65 to the nucleus and the upregulation of NF-κB transcriptional activity. BITC also decreased β-catenin binding to a positive cis element of the cyclin D1 promoter and thus inhibited β-catenin-dependent cyclin D1 transcription, possibly through a direct interaction between p65 and β-catenin. siRNA-mediated knockdown of p65 confirmed that p65 negatively affects cyclin D1 expression. On the other hand, when human colorectal cancer HCT-116 cells with wild-type p53 were treated with BITC, translocation of p65 to the nucleus was inhibited rather than enhanced. p53 knockout increased the BITC sensitivity of HCT-116 cells in a p65-dependent manner, suggesting that p53 negatively regulates p65-dependent effects. Together, these results identify BITC as a novel type of antiproliferative agent that regulates the NF-κB pathway in p53-deficient colorectal cancer cells.
Collapse
|
38
|
Pysz MA, Hao F, Hizli AA, Lum MA, Swetzig WM, Black AR, Black JD. Differential regulation of cyclin D1 expression by protein kinase C α and ϵ signaling in intestinal epithelial cells. J Biol Chem 2014; 289:22268-83. [PMID: 24914206 DOI: 10.1074/jbc.m114.571554] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Cellular accumulation of cyclin D1, a key regulator of cell proliferation and tumorigenesis, is subject to tight control. Our previous studies have identified PKCα as a negative regulator of cyclin D1 in the intestinal epithelium. However, treatment of non-transformed IEC-18 ileal crypt cells with PKC agonists has a biphasic effect on cyclin D1 expression. Initial PKCα-mediated down-regulation is followed by recovery and subsequent accumulation of the cyclin to levels markedly higher than those seen in untreated cells. Using protein overexpression strategies, siRNA, and pharmacological inhibitors, we now demonstrate that the recovery and hyperinduction of cyclin D1 reflect the combined effects of (a) loss of negative signals from PKCα due to agonist-induced PKCα down-regulation and (b) positive effects of PKCϵ. PKCϵ-mediated up-regulation of cyclin D1 requires sustained ERK stimulation and transcriptional activation of the proximal cyclin D1 (CCDN1) promoter, without apparent involvement of changes in protein stability or translation. PKCϵ also up-regulates cyclin D1 expression in colon cancer cells, through mechanisms that parallel those in IEC-18 cells. Although induction of cyclin D1 by PKCϵ is dependent on non-canonical NF-κB activation, the NF-κB site in the proximal promoter is not required. Instead, cyclin D1 promoter activity is regulated by a novel interaction between NF-κB and factors that associate with the cyclic AMP-response element adjacent to the NF-κB site. The differential effects of PKCα and PKCϵ on cyclin D1 accumulation are likely to contribute to the opposing tumor-suppressive and tumor-promoting activities of these PKC family members in the intestinal epithelium.
Collapse
Affiliation(s)
- Marybeth A Pysz
- the Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263
| | - Fang Hao
- the Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263
| | - A Asli Hizli
- the Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263
| | - Michelle A Lum
- From the Eppley Institute for Research in Cancer and Allied Diseases and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198 and
| | - Wendy M Swetzig
- the Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263
| | - Adrian R Black
- the Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263From the Eppley Institute for Research in Cancer and Allied Diseases and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198 and
| | - Jennifer D Black
- the Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263From the Eppley Institute for Research in Cancer and Allied Diseases and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198 and
| |
Collapse
|
39
|
Berry WL, Kim TD, Janknecht R. Stimulation of β-catenin and colon cancer cell growth by the KDM4B histone demethylase. Int J Oncol 2014; 44:1341-8. [PMID: 24481461 DOI: 10.3892/ijo.2014.2279] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 12/21/2013] [Indexed: 11/06/2022] Open
Abstract
The linchpin of colorectal cancer is the oncoprotein and transcriptional cofactor β-catenin, whose overexpression is causative for the neoplastic transformation of colon cells. However, the molecular details of β-catenin dependent gene transcription in cancer cells are still not comprehensively explored. Here, we show that the histone demethylase KDM4B was upregulated in colon and rectal adenocarcinomas and required for efficient growth and clonogenic activity of human HT-29 colon cancer cells. Moreover, KDM4B formed complexes with β-catenin in vitro and in vivo, which involved its central amino acids 353-740. In addition, KDM4B also interacted with the DNA-binding protein TCF4, which is the main factor recruiting β-catenin to chromatin in the intestine. KDM4B downregulation resulted in reduced expression of the β-catenin/TCF4 target genes JUN, MYC and Cyclin D1, all of which encode for oncoproteins. Collectively, our data indicate that KDM4B overexpression supports β-catenin mediated gene transcription and thereby contributes to the genesis of colorectal tumors. Accordingly, inhibition of the KDM4B histone demethylase may represent a novel avenue of fighting colorectal cancer, one of the major causes of cancer death throughout the world.
Collapse
Affiliation(s)
- William L Berry
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Tae-Dong Kim
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ralf Janknecht
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
40
|
Emerging roles of peroxisome proliferator-activated receptor gamma in cancer. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
41
|
Ju X, Casimiro MC, Gormley M, Meng H, Jiao X, Katiyar S, Crosariol M, Chen K, Wang M, Quong AA, Lisanti MP, Ertel A, Pestell RG. Identification of a cyclin D1 network in prostate cancer that antagonizes epithelial-mesenchymal restraint. Cancer Res 2013; 74:508-19. [PMID: 24282282 DOI: 10.1158/0008-5472.can-13-1313] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Improved clinical management of prostate cancer has been impeded by an inadequate understanding of molecular genetic elements governing tumor progression. Gene signatures have provided improved prognostic indicators of human prostate cancer. The TGF-β/BMP-SMAD4 signaling pathway, which induces epithelial-mesenchymal transition (EMT), is known to constrain prostate cancer progression induced by Pten deletion. Herein, cyclin D1 inactivation reduced cellular proliferation in the murine prostate in vivo and in isogenic oncogene-transformed prostate cancer cell lines. The in vivo cyclin D1-mediated molecular signature predicted poor outcome of recurrence-free survival for patients with prostate cancer (K-means HR, 3.75, P = 0.02) and demonstrated that endogenous cyclin D1 restrains TGF-β, Snail, Twist, and Goosecoid signaling. Endogenous cyclin D1 enhanced Wnt and ES cell gene expression and expanded a prostate stem cell population. In chromatin immunoprecipitation sequencing, cyclin D1 occupied genes governing stem cell expansion and induced their transcription. The coordination of EMT restraining and stem cell expanding gene expression by cyclin D1 in the prostate may contribute to its strong prognostic value for poor outcome in biochemical-free recurrence in human prostate cancer.
Collapse
Affiliation(s)
- Xiaoming Ju
- Authors' Affiliations: Departments of Cancer Biology, Medical Oncology, and Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Pestell RG. New roles of cyclin D1. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:3-9. [PMID: 23790801 DOI: 10.1016/j.ajpath.2013.03.001] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 02/26/2013] [Accepted: 03/07/2013] [Indexed: 12/11/2022]
Abstract
Cyclins encode regulatory subunits of holoenzymes that phosphorylate a variety of cellular substrates. Although the classic role of cyclins in cell cycle progression and tumorigenesis has been well characterized, new functions have been identified, including the induction of cellular migration and invasion, enhancement of angiogenesis, inhibition of mitochondrial metabolism, regulation of transcription factor signaling via a DNA-bound form, the induction of chromosomal instability, enhancement of DNA damage sensing and DNA damage repair, and feedback governing expression of the noncoding genome. This review describes the mechanisms of these new functions of cyclin D1.
Collapse
Affiliation(s)
- Richard G Pestell
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA.
| |
Collapse
|
43
|
Shimura T, Fukumoto M, Kunugita N. The role of cyclin D1 in response to long-term exposure to ionizing radiation. Cell Cycle 2013; 12:2738-43. [PMID: 23974042 DOI: 10.4161/cc.25746] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The health-related hazards resulting from long-term exposure to radiation remain unknown. Thus, an appropriate molecular marker is needed to clarify these effects. Cyclin D1 regulates the cell cycle transition from the G 1 phase to the S phase. Cyclin D1 is degraded as a G1/S checkpoint after 10 Gy of single acute radiation exposure, whereas conversely, cyclin D1 is stabilized when human tumor cells are exposed to fractionated radiation (FR) with 0.5 Gy of x-rays for 31 d. In this article, we review new findings regarding cyclin D1 overexpression in response to long-term exposure to FR. Cyclin D1 overexpression is associated with induction of genomic instability in irradiated cells. Therefore, repression of cyclin D1 expression is likely to cancel the harmful effects of long-term exposure to FR. Thus cyclin D1 may be a marker of long-term exposure to radiation and is a putative molecular radioprotection target for radiation safety.
Collapse
Affiliation(s)
- Tsutomu Shimura
- Department of Environmental Health; National Institute of Public Health 2-3-6 Minami; Wako, Saitama, Japan
| | | | | |
Collapse
|
44
|
Casimiro MC, Wang C, Li Z, Di Sante G, Willmart NE, Addya S, Chen L, Liu Y, Lisanti MP, Pestell RG. Cyclin D1 determines estrogen signaling in the mammary gland in vivo. Mol Endocrinol 2013; 27:1415-28. [PMID: 23864650 DOI: 10.1210/me.2013-1065] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The CCND1 gene, which is frequently overexpressed in cancers, encodes the regulatory subunit of a holoenzyme that phosphorylates the retinoblastoma protein. Although it is known that cyclin D1 regulates estrogen receptor (ER)α transactivation using heterologous reporter systems, the in vivo biological significance of cyclin D1 to estrogen-dependent signaling, and the molecular mechanisms by which cyclin D1 is involved, are yet to be elucidated. Herein, genome-wide expression profiling conducted of 17β-estradiol-treated castrated virgin mice deleted of the Ccnd1 gene demonstrated that cyclin D1 determines estrogen-dependent gene expression for 88% of estrogen-responsive genes in vivo. In addition, expression profiling of 17β-estradiol-stimulated cyclin D1 small interfering RNA treated MCF7 cells shows cyclin D1 is required for estrogen-mediated gene expression in vitro. Genome-wide chromatin immunoprecipitation-Seq analysis revealed a cyclin D1-DNA bound form associated with genes that were regulated by estrogen in a cyclin D1-dependent manner. The cyclin D1-dependent estrogen signaling pathways identified in vivo were highly enriched for extracellular membrane-associated growth factor receptors (epidermal growth factor receptor, ErbB3, and EphB3) and their ligands (amphiregulin, encoded by AREG gene), and matrix metalloproteinase. The AREG protein, a pivotal ligand for epidermal growth factor receptors to promote cellular proliferation, was induced by cyclin D1 via the AREG promoter. Chromatin immunoprecipitation analysis demonstrated the recruitment of cyclin D1 to the breast cancer 1 (Brca1)/ERα binding site of the Areg gene. Cyclin D1 genetic deletion demonstrated the in vivo requirement for cyclin D1 in assembling the estrogen-dependent amplified in breast cancer 1-associated multiprotein complex. The current studies define a requirement for cyclin D1 in estrogen-dependent signaling modules governing growth factor receptor and ligand expression in vivo and reveal a noncanonical function of cyclin D1 at ERα target gene promoters. Cyclin D1 mediates the convergence of ERα and growth factor signaling at a common cis-element of growth factor genes.
Collapse
Affiliation(s)
- Mathew C Casimiro
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Signaling through cyclin D-dependent kinases. Oncogene 2013; 33:1890-903. [PMID: 23644662 DOI: 10.1038/onc.2013.137] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 02/22/2013] [Accepted: 02/27/2013] [Indexed: 12/13/2022]
Abstract
Research over the past quarter century has identified cyclin D-dependent kinases, CDK4 and CDK6, as the major oncogenic drivers among members of the CDK superfamily. CDK4/6 are rendered hyperactive in the majority of human cancers through a multitude of genomic alterations. Sustained activation of these protein kinases provides cancer cells with the power to enter the cell cycle continuously by triggering G1-S-phase transitions and dramatically shortening the duration of the G1 phase. It has also become clear, however, that CDK4/6 effectively counter cancer cell-intrinsic tumor suppression mechanisms, senescence and apoptosis, which must be overcome during cell transformation and kept at bay throughout all stages of tumorigenesis. As a central 'node' in cellular signaling networks, cyclin D-dependent kinases sense a plethora of mitogenic signals to orchestrate specific transcriptional programs. As the complexity of the cellular signaling network regulated by these oncogenic kinases unfolds, much remains to be learned about its architecture, its dynamics and the consequences of its perturbation.
Collapse
|
46
|
Casimiro MC, Crosariol M, Loro E, Li Z, Pestell RG. Cyclins and cell cycle control in cancer and disease. Genes Cancer 2013; 3:649-57. [PMID: 23634253 DOI: 10.1177/1947601913479022] [Citation(s) in RCA: 164] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cyclin D1 overexpression is found in more than 50% of human breast cancers and causes mammary cancer in transgenic mice. Dysregulation of cyclin D1 gene expression or function contributes to the loss of normal cell cycle control during tumorigenesis. Recent studies have demonstrated that cyclin D1 conducts additional specific functions to regulate gene expression in the context of local chromatin, promote cellular migration, and promote chromosomal instability. It is anticipated that these additional functions contribute to the pathology associated with dysregulated cyclin D1 abundance. This article discusses evidence that examines the functional roles that cyclin D1 may play in cancer with an emphasis on other cyclin family members that also may contribute to cancer and disease in a similar fashion.
Collapse
|
47
|
Bai Z, Tai Y, Li W, Zhen C, Gu W, Jian Z, Wang Q, Lin JE, Zhao Q, Gong W, Liang B, Wang C, Zhou T. Gankyrin Activates IL-8 to Promote Hepatic Metastasis of Colorectal Cancer. Cancer Res 2013; 73:4548-58. [DOI: 10.1158/0008-5472.can-12-4586] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
48
|
Datta K, Suman S, Kallakury BVS, Fornace AJ. Heavy ion radiation exposure triggered higher intestinal tumor frequency and greater β-catenin activation than γ radiation in APC(Min/+) mice. PLoS One 2013; 8:e59295. [PMID: 23555653 PMCID: PMC3605451 DOI: 10.1371/journal.pone.0059295] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 02/13/2013] [Indexed: 01/11/2023] Open
Abstract
Risk of colorectal cancer (CRC) after exposure to low linear energy transfer (low-LET) radiation such as γ-ray is highlighted by the studies in atom bomb survivors. On the contrary, CRC risk prediction after exposure to high-LET cosmic heavy ion radiation exposure is hindered due to scarcity of in vivo data. Therefore, intestinal tumor frequency, size, cluster, and grade were studied in APCMin/+ mice (n = 20 per group; 6 to 8 wks old; female) 100 to 110 days after exposure to 1.6 or 4 Gy of heavy ion 56Fe radiation (energy: 1000 MeV/nucleon) and results were compared to γ radiation doses of 2 or 5 Gy, which are equitoxic to 1.6 and 4 Gy 56Fe respectively. Due to relevance of lower doses to radiotherapy treatment fractions and space exploration, we followed 2 Gy γ and equitoxic 1.6 Gy 56Fe for comparative analysis of intestinal epithelial cell (IEC) proliferation, differentiation, and β-catenin signaling pathway alterations between the two radiation types using immunoblot, and immunohistochemistry. Relative to controls and γ-ray, intestinal tumor frequency and grade was significantly higher after 56Fe radiation. Additionally, tumor incidence per unit of radiation (per cGy) was also higher after 56Fe radiation relative to γ radiation. Staining for phospho-histone H3, indicative of IEC proliferation, was more and alcian blue staining, indicative of IEC differentiation, was less in 56Fe than γ irradiated samples. Activation of β-catenin was more in 56Fe-irradiated tumor-free and tumor-bearing areas of the intestinal tissues. When considered along with higher levels of cyclin D1, we infer that relative to γ radiation exposure to 56Fe radiation induced markedly reduced differentiation, and increased proliferative index in IEC resulting in increased intestinal tumors of larger size and grade due to preferentially greater activation of β-catenin and its downstream effectors.
Collapse
Affiliation(s)
- Kamal Datta
- Department of Biochemistry and Molecular & Cell Biology and Lombardi Comprehensives Cancer Center, Georgetown University, Washington, DC, United States of America
- * E-mail: (KD); (AJF)
| | - Shubhankar Suman
- Department of Biochemistry and Molecular & Cell Biology and Lombardi Comprehensives Cancer Center, Georgetown University, Washington, DC, United States of America
| | - Bhaskar V. S. Kallakury
- Department of Pathology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Albert J. Fornace
- Department of Biochemistry and Molecular & Cell Biology and Lombardi Comprehensives Cancer Center, Georgetown University, Washington, DC, United States of America
- Center of Excellence In Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- * E-mail: (KD); (AJF)
| |
Collapse
|
49
|
Expression of cyclin D1a and D1b as predictive factors for treatment response in colorectal cancer. Br J Cancer 2012; 107:1684-91. [PMID: 23099809 PMCID: PMC3493874 DOI: 10.1038/bjc.2012.463] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background: The aim of this study was to investigate the value of the cyclin D1 isoforms D1a and D1b as prognostic factors and their relevance as predictors of response to adjuvant chemotherapy with 5-fluorouracil and levamisole (5-FU/LEV) in colorectal cancer (CRC). Methods: Protein expression of nuclear cyclin D1a and D1b was assessed by immunohistochemistry in 335 CRC patients treated with surgery alone or with adjuvant therapy using 5-FU/LEV. The prognostic and predictive value of these two molecular markers and clinicopathological factors were evaluated statistically in univariate and multivariate survival analyses. Results: Neither cyclin D1a nor D1b showed any prognostic value in CRC or colon cancer patients. However, high cyclin D1a predicted benefit from adjuvant therapy measured in 5-year relapse-free survival (RFS) and CRC-specific survival (CSS) compared to surgery alone in colon cancer (P=0.012 and P=0.038, respectively) and especially in colon cancer stage III patients (P=0.005 and P=0.019, respectively) in univariate analyses. An interaction between treatment group and cyclin D1a could be shown for RFS (P=0.004) and CSS (P=0.025) in multivariate analysis. Conclusion: Our study identifies high cyclin D1a protein expression as a positive predictive factor for the benefit of adjuvant 5-FU/LEV treatment in colon cancer, particularly in stage III colon cancer.
Collapse
|
50
|
Choi YJ, Li X, Hydbring P, Sanda T, Stefano J, Christie AL, Signoretti S, Look AT, Kung AL, von Boehmer H, Sicinski P. The requirement for cyclin D function in tumor maintenance. Cancer Cell 2012; 22:438-51. [PMID: 23079655 PMCID: PMC3487466 DOI: 10.1016/j.ccr.2012.09.015] [Citation(s) in RCA: 271] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Revised: 06/24/2012] [Accepted: 09/18/2012] [Indexed: 11/24/2022]
Abstract
D-cyclins represent components of cell cycle machinery. To test the efficacy of targeting D-cyclins in cancer treatment, we engineered mouse strains that allow acute and global ablation of individual D-cyclins in a living animal. Ubiquitous shutdown of cyclin D1 or inhibition of cyclin D-associated kinase activity in mice bearing ErbB2-driven mammary carcinomas triggered tumor cell senescence, without compromising the animals' health. Ablation of cyclin D3 in mice bearing Notch1-driven T cell acute lymphoblastic leukemias (T-ALL) triggered tumor cell apoptosis. Such selective killing of leukemic cells can also be achieved by inhibiting cyclin D associated kinase activity in mouse and human T-ALL models. Inhibition of cyclin D-kinase activity represents a highly-selective anticancer strategy that specifically targets cancer cells without significantly affecting normal tissues.
Collapse
Affiliation(s)
- Yoon Jong Choi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Xiaoyu Li
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Microbiology and Immunology, Harvard Medical School, Boston, MA 02115
| | - Per Hydbring
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Takaomi Sanda
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Division of Hematology/Oncology, Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Joanna Stefano
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Amanda L. Christie
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - A. Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Division of Hematology/Oncology, Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Andrew L. Kung
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Harald von Boehmer
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Microbiology and Immunology, Harvard Medical School, Boston, MA 02115
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Correspondence:
| |
Collapse
|