1
|
Choi Y, Kim N, Park JH, Song CH, Oh HJ. Expression Rates of Sex Hormone Receptors with Their Clinical Correlates in Gastric Cancer Patients and Normal Controls. World J Mens Health 2025; 43:43.e6. [PMID: 39843179 DOI: 10.5534/wjmh.240272] [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: 11/02/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 01/24/2025] Open
Abstract
PURPOSE Sex hormones affect development and prognosis of gastric cancer (GC). This study aimed to compare the sex hormone receptor expression between control and GC, and to evaluate its correlation with patient characteristics. MATERIALS AND METHODS 110 patients (74 with GC, 36 controls) underwent immunohistochemistry (IHC) and reverse transcription-polymerase chain reaction (RT-PCR) for estrogen receptors (ERs) α and β and androgen receptor (AR). The effect of ERs and AR on the clinicopathological and tumor characteristics were analyzed. RESULTS The positive rate of ERα, ERβ, and AR in GC tissue was 64.9%, 78.4%, and 60.8% by IHC and 41.4%, 27.6%, and 48.3% in RT-PCR respectively. In control, the positive rate of those was 16.7%, 80.6%, and 38.9% by IHC and 22.2%, 58.3%, and 19.4% in RT-PCR respectively. The IHC and RT-PCR results showed concordance with each other, and ERα and AR expressions were positively correlated with cancer, while ERβ showed the opposite pattern. ERα expression was correlated with Helicobacter pylori negativity (p<0.001), diffuse or mixed-type histology (p=0.014), and undifferentiated histology (p<0.001), and AR expression was related to H. pylori negativity (p<0.001), cardiac cancer (p=0.040), and undifferentiated histology (p<0.001). The higher expression rate of ERα in males and that of AR in females seemed to be related with cancer, showing sex differences. CONCLUSIONS The expression rates of ERα, ERβ, and AR were different depending on sex, histologic type and H. pylori infection status, which may explain sex-based differences in GC.
Collapse
Affiliation(s)
- Yonghoon Choi
- Department of Internal Medicine and Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine and Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| | - Ji Hyun Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Chin-Hee Song
- Department of Internal Medicine and Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyeon Jeong Oh
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
2
|
Latacz E, Verheul SML, Sillis Y, van Dam PJ, Doukas M, Grunhagen DJ, Nyström H, Dirix P, Dirix L, Van Laere S, Verhoef C, Vermeulen P. Molecular characterization of the histopathological growth patterns of colorectal cancer liver metastases by RNA sequencing of targeted samples at the tumor-liver interface. Clin Exp Metastasis 2024; 42:1. [PMID: 39666203 DOI: 10.1007/s10585-024-10319-w] [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: 09/15/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024]
Abstract
The behaviour of metastases in patients with liver-metastatic colorectal cancer (CRC) is still not adequately considered during treatment planning. However, studies in large cohorts have shown that the disease course in these patients depends on the histopathological growth pattern (HGP) of the liver metastases, with the desmoplastic (or encapsulated) pattern responsible for a favourable outcome and the replacement pattern for an unfavourable course. To increase our knowledge of cancer biology in general as well as to design clinical trials that take into account the diverse behaviour of liver metastases, it is necessary to know the cellular and molecular determinants of these growth patterns. For that purpose, we compared the transcriptome of tumour tissue (prospective cohort; n = 57) sampled very precisely at the transition of metastasis and adjacent liver, between the desmoplastic and replacement HGP. In addition, the mutational profiles for 46 genes related to CRC were extracted from the RNA sequencing reads. First, we show that the genetic constitution of a liver metastasis from colorectal cancer does not determine its HGP. Second, we show clear differences between HGPs regarding the expression of genes belonging to the Molecular Signatures Database hallmark gene sets. Biological themes of the replacement HGP reflect cancer cell proliferation and glucose metabolism, while the desmoplastic HGP is characterized by inflammation and immune response, and angiogenesis. This study supports the view that HGPs are a reflection of the biology of CRC liver metastases and suggests the HGPs are driven epigenetically rather than by specific gene mutations.
Collapse
Affiliation(s)
- Emily Latacz
- Translational Cancer Research Unit, Ziekenhuis aan de Stroom (ZAS), Campus Augustinus, Antwerp, Belgium
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Sanne M L Verheul
- Department of Gastrointestinal Surgery and Surgical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Yasmine Sillis
- Translational Cancer Research Unit, Ziekenhuis aan de Stroom (ZAS), Campus Augustinus, Antwerp, Belgium
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | | | - Michail Doukas
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dirk J Grunhagen
- Department of Gastrointestinal Surgery and Surgical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hanna Nyström
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - Piet Dirix
- Translational Cancer Research Unit, Ziekenhuis aan de Stroom (ZAS), Campus Augustinus, Antwerp, Belgium
| | - Luc Dirix
- Translational Cancer Research Unit, Ziekenhuis aan de Stroom (ZAS), Campus Augustinus, Antwerp, Belgium
| | - Steven Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Cornelis Verhoef
- Department of Gastrointestinal Surgery and Surgical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Peter Vermeulen
- Translational Cancer Research Unit, Ziekenhuis aan de Stroom (ZAS), Campus Augustinus, Antwerp, Belgium.
- Department of Gastrointestinal Surgery and Surgical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Imani S, Aminnezhad S, Alikarami M, Abedi Z, Mosleh IS, Maghsoudloo M, Taheri Z. Exploration of drug repurposing for Mpox outbreaks targeting gene signatures and host-pathogen interactions. Sci Rep 2024; 14:29436. [PMID: 39604570 PMCID: PMC11603026 DOI: 10.1038/s41598-024-79897-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Monkeypox (Mpox) is a growing public health concern, with complex interactions within host systems contributing to its impact. This study employs multi-omics approaches to uncover therapeutic targets and potential drug repurposing opportunities to better understand Mpox's molecular pathogenesis. We developed an in silico host-pathogen interaction (HPI) network and applied weighted gene co-expression network analysis (WGCNA) to explore interactions between Mpox and host proteins. Subtype-specific host-pathogen protein-protein interaction networks were constructed, and key modules from the HPI and WGCNA were integrated to identify significant host proteins. To predict upstream signaling pathways and transcription factors, we used eXpression2Kinases and ChIP-X Enrichment Analysis. The multi-Steiner trees method was applied to compare our findings with those from FDA-approved antiviral drugs. Analysis of 55 differentially expressed genes in Mpox infection revealed 11 kinases and 15 transcription factors as key regulators. We identified 16 potential drug targets, categorized into 8 proviral genes (ESR2, ERK1, ERK2, P38, JNK1, CDK4, GSK3B, STAT3) designated for inhibition, and 8 antiviral genes (IKKA, HDAC1, HIPK2, TF65, CSK21, HIPK2, ESR2, GSK3B) designated for activation. Proviral genes are involved in the AKT, Wnt, and STAT3 pathways, while antiviral genes impact the AP-1, NF-κB, apoptosis, and IFN pathways. Promising FDA-approved candidates were identified, including kinase inhibitors, steroid hormone receptor agonists, STAT3 inhibitors, and notably Niclosamide. This study enhances our understanding of Mpox by identifying key therapeutic targets and potential repurposable drugs, providing a valuable framework for developing new treatments.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China.
| | - Sargol Aminnezhad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Moslem Alikarami
- Research and Development Center, Dina Pharmed Exir Salamat Co, Tehran, Iran
| | - Zahra Abedi
- School of Biotechnology College of Science, University of Tehran, Tehran, Iran
| | - Iman Samei Mosleh
- Plant Functional Genomics Lab, Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Vienna, Austria
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zahra Taheri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
4
|
Monteiro FL, Stepanauskaite L, Archer A, Williams C. Estrogen receptor beta expression and role in cancers. J Steroid Biochem Mol Biol 2024; 242:106526. [PMID: 38657699 DOI: 10.1016/j.jsbmb.2024.106526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/06/2024] [Accepted: 04/21/2024] [Indexed: 04/26/2024]
Abstract
Estrogen drives the growth of some cancers, such as breast cancer, via estrogen receptor alpha (ERα). Estrogen also activates ERβ, but whether ERβ is expressed and has a role in different cancers is debated. The use of nonspecific antibodies has contributed to the confusion, and this review delves into ERβ's controversial role in cancer and focuses on tumor expression that can be supported by non-antibody-dependent assays. We discuss its expression at the transcript level and focus on its potential role in lymphoma, granulosa cell tumors, testicular, and adrenal cancers, emphasizing recent findings and the complexities that necessitate further research.
Collapse
Affiliation(s)
- Fátima L Monteiro
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden
| | - Lina Stepanauskaite
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden
| | - Amena Archer
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden
| | - Cecilia Williams
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden.
| |
Collapse
|
5
|
Wu J, Bai Y, Lu Y, Yu Z, Zhang S, Yu B, Chen L, Li J. Role of sex steroids in colorectal cancer: pathomechanisms and medical applications. Am J Cancer Res 2024; 14:3200-3221. [PMID: 39113870 PMCID: PMC11301278 DOI: 10.62347/oebs6893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/26/2024] [Indexed: 08/10/2024] Open
Abstract
Given that the colon represents the most extensive hormone-responsive tissue in the human body, it prompts a compelling inquiry into whether the progression of its cancer is intimately linked to hormonal dynamics. Consequently, the interplay between sex steroids - a pivotal constituent of hormones - and colorectal cancer has increasingly captivated scientific interest. Upon a comprehensive review of pertinent literature both domestically and internationally, this study delineates the present landscape of three pivotal steroids - estrogen, progestin, and androgen - in the context of colorectal cancer. More specifically, this investigation probes into the potential utility of these steroids in providing therapeutic interventions, diagnostic insights, and prognostic indicators. Furthermore, this study also delves into the mechanistic pathways through which sex steroid interventions exert influence on colorectal cancer. It was discovered that the trio of sex steroid hormones partakes in an array of biological processes, thereby influencing the onset and progression of colorectal cancer. In conclusion, this study posits that a profound interconnection exists between colorectal cancer and sex steroids, suggesting that elucidating the targets of their action mechanisms could unveil novel avenues for the diagnosis and prevention of colorectal cancer.
Collapse
Affiliation(s)
- Jianglan Wu
- Hunan University of Traditional Chinese MedicineChangsha 410208, Hunan, China
| | - Yanan Bai
- Hunan University of Traditional Chinese MedicineChangsha 410208, Hunan, China
| | - Yuwen Lu
- Hunan University of Traditional Chinese MedicineChangsha 410208, Hunan, China
| | - Zixuan Yu
- Hunan University of Traditional Chinese MedicineChangsha 410208, Hunan, China
| | - Shumeng Zhang
- Hunan University of Traditional Chinese MedicineChangsha 410208, Hunan, China
| | - Bin Yu
- Department of Gastroenterology, The First Affiliated Hospital of Hunan University of Traditional Chinese MedicineChangsha 410007, Hunan, China
| | - Lingli Chen
- Hunan Provincial Key Laboratory of Pathogenic Biology Based on Integrated Chinese and Western Medicine, Hunan University of Traditional Chinese MedicineChangsha 410208, Hunan, China
| | - Jie Li
- Hunan University of Traditional Chinese MedicineChangsha 410208, Hunan, China
| |
Collapse
|
6
|
Rawłuszko-Wieczorek AA, Lipowicz J, Nowacka M, Ostrowska K, Pietras P, Blatkiewicz M, Ruciński M, Jagodziński PP, Nowicki M. Estrogen receptor β affects hypoxia response in colorectal cancer cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166894. [PMID: 37748565 DOI: 10.1016/j.bbadis.2023.166894] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023]
Abstract
The occurrence of colorectal cancer (CRC) is inversely correlated with estrogen receptor beta (ERβ) presence. Additionally, multiple studies associate low ERβ expression with poorer overall survival of CRC patients. Molecular pathways involved in ERβ - related reduced tumorigenesis include enhanced apoptosis, decreased proliferation, or repression of oncogenes. Moreover, the development of solid tumors, such as CRC, is often associated with an increased tumor mass that results in decreased oxygen partial tension, known as hypoxia, clinically associated with decreased prognosis and therapeutic resistance. Our high-throughput study suggests that ERβ also represses a hypoxic response in CRC cells. We observed a significantly altered transcriptional profile in HCT116 ERβ overexpressing cells that was further stimulated by E2 treatment under hypoxic conditions. The achieved data for downregulation of VEGFA, PDGFA and ANGPTL4 were validated in a time course experiment in DLD-1 cells. In addition, using an ERβ construct with a mutated DNA binding domain we observed that the downregulation of selected genes is dependent on the direct binding of this receptor to regulatory region genes. In addition, we observed that ERβ may affect the expression of the main hypoxia regulator, HIF1A, at the transcriptional and translational levels. In summary, ERβ alters the hypoxic outcome in CRC cells.
Collapse
Affiliation(s)
| | - Julia Lipowicz
- Department of Histology, Poznań University of Medical Sciences, Poland
| | - Marta Nowacka
- Department of Histology, Poznań University of Medical Sciences, Poland
| | - Kamila Ostrowska
- Department of Histology, Poznań University of Medical Sciences, Poland; Department of Head and Neck Oncology, Poznań University of Medical Sciences, The Greater Poland Cancer Centre, Poland
| | - Paulina Pietras
- Department of Histology, Poznań University of Medical Sciences, Poland
| | | | - Marcin Ruciński
- Department of Histology, Poznań University of Medical Sciences, Poland
| | - Paweł Piotr Jagodziński
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Poland
| | - Michał Nowicki
- Department of Histology, Poznań University of Medical Sciences, Poland
| |
Collapse
|
7
|
Birgersson M, Indukuri R, Lindquist L, Stepanauskaite L, Luo Q, Deng Q, Archer A, Williams C. Ovarian ERβ cistrome and transcriptome reveal chromatin interaction with LRH-1. BMC Biol 2023; 21:277. [PMID: 38031019 PMCID: PMC10688478 DOI: 10.1186/s12915-023-01773-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Estrogen receptor beta (ERβ, Esr2) plays a pivotal role in folliculogenesis and ovulation, yet its exact mechanism of action is mainly uncharacterized. RESULTS We here performed ERβ ChIP-sequencing of mouse ovaries followed by complementary RNA-sequencing of wild-type and ERβ knockout ovaries. By integrating the ERβ cistrome and transcriptome, we identified its direct target genes and enriched biological functions in the ovary. This demonstrated its strong impact on genes regulating organism development, cell migration, lipid metabolism, response to hypoxia, and response to estrogen. Cell-type deconvolution analysis of the bulk RNA-seq data revealed a decrease in luteal cells and an increased proportion of theca cells and a specific type of cumulus cells upon ERβ loss. Moreover, we identified a significant overlap with the gene regulatory network of liver receptor homolog 1 (LRH-1, Nr5a2) and showed that ERβ and LRH-1 extensively bound to the same chromatin locations in granulosa cells. Using ChIP-reChIP, we corroborated simultaneous ERβ and LRH-1 co-binding at the ERβ-repressed gene Greb1 but not at the ERβ-upregulated genes Cyp11a1 and Fkbp5. Transactivation assay experimentation further showed that ERβ and LRH-1 can inhibit their respective transcriptional activity at classical response elements. CONCLUSIONS By characterizing the genome-wide endogenous ERβ chromatin binding, gene regulations, and extensive crosstalk between ERβ and LRH-1, along with experimental corroborations, our data offer genome-wide mechanistic underpinnings of ovarian physiology and fertility.
Collapse
Affiliation(s)
- Madeleine Birgersson
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Rajitha Indukuri
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
| | - Linnéa Lindquist
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Lina Stepanauskaite
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Qing Luo
- Department of Physiology and Pharmacology, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Amena Archer
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Cecilia Williams
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden.
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden.
| |
Collapse
|
8
|
Iloki Assanga SB, Lewis Luján LM, McCarty MF. Targeting beta-catenin signaling for prevention of colorectal cancer - Nutraceutical, drug, and dietary options. Eur J Pharmacol 2023; 956:175898. [PMID: 37481200 DOI: 10.1016/j.ejphar.2023.175898] [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: 03/11/2023] [Revised: 06/09/2023] [Accepted: 06/29/2023] [Indexed: 07/24/2023]
Abstract
Progressive up-regulation of β-catenin signaling is very common in the transformation of colorectal epithelium to colorectal cancer (CRC). Practical measures for opposing such signaling hence have potential for preventing or slowing such transformation. cAMP/PKA activity in colon epithelium, as stimulated by COX-2-generated prostaglandins and β2-adrenergic signaling, boosts β-catenin activity, whereas cGMP/PKG signaling has the opposite effect. Bacterial generation of short-chain fatty acids (as supported by unrefined high-carbohydrate diets, berberine, and probiotics), dietary calcium, daily aspirin, antioxidants opposing cox-2 induction, and nicotine avoidance, can suppress cAMP production in colonic epithelium, whereas cGMP can be boosted via linaclotides, PDE5 inhibitors such as sildenafil or icariin, and likely high-dose biotin. Selective activation of estrogen receptor-β by soy isoflavones, support of adequate vitamin D receptor activity with UV exposure or supplemental vitamin D, and inhibition of CK2 activity with flavanols such as quercetin, can also oppose β-catenin signaling in colorectal epithelium. Secondary bile acids, the colonic production of which can be diminished by low-fat diets and berberine, can up-regulate β-catenin activity by down-regulating farnesoid X receptor expression. Stimulation of PI3K/Akt via insulin, IGF-I, TLR4, and EGFR receptors boosts β-catenin levels via inhibition of glycogen synthase-3β; plant-based diets can down-regulate insulin and IGF-I levels, exercise training and leanness can keep insulin low, anthocyanins and their key metabolite ferulic acid have potential for opposing TLR4 signaling, and silibinin is a direct antagonist for EGFR. Partially hydrolyzed phytate can oppose growth factor-mediated down-regulation of β-catenin by inhibiting Akt activation. Multifactorial strategies for safely opposing β-catenin signaling can be complemented with measures that diminish colonic mutagenesis and DNA hypomethylation - such as avoidance of heme-rich meat and charred or processed meats, consumption of phase II-inductive foods and nutraceuticals (e.g., Crucifera), and assurance of adequate folate status.
Collapse
Affiliation(s)
- Simon Bernard Iloki Assanga
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora, Blvd Luis Encinas y Rosales S/N Col. Centro, Hermosillo, Sonora, C.P. 83000, Mexico.
| | - Lidianys María Lewis Luján
- Technological Institute of Hermosillo (ITH), Ave. Tecnológico y Periférico Poniente S/N, Col. Sahuaro, Hermosillo, Sonora, C.P. 83170, México.
| | | |
Collapse
|
9
|
Gut Epithelial Inositol Polyphosphate Multikinase Alleviates Experimental Colitis via Governing Tuft Cell Homeostasis. Cell Mol Gastroenterol Hepatol 2022; 14:1235-1256. [PMID: 35988719 PMCID: PMC9579329 DOI: 10.1016/j.jcmgh.2022.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Inositol polyphosphate multikinase (IPMK), an essential enzyme for inositol phosphate metabolism, has been known to mediate major biological events such as growth. Recent studies have identified single-nucleotide polymorphisms in the IPMK gene associated with inflammatory bowel disease predisposition. Therefore, we aimed to investigate the functional significance of IPMK in gut epithelium. METHODS We generated intestinal epithelial cell (IEC)-specific Ipmk knockout (IPMKΔIEC) mice, and assessed their vulnerability against dextran sulfate sodium-induced experimental colitis. Both bulk and single-cell RNA sequencing were performed to analyze IPMK-deficient colonic epithelial cells and colonic tuft cells. RESULTS Although IPMKΔIEC mice developed normally and showed no intestinal abnormalities during homeostasis, Ipmk deletion aggravated dextran sulfate sodium-induced colitis, with higher clinical colitis scores, and increased epithelial barrier permeability. Surprisingly, Ipmk deletion led to a significant decrease in the number of tuft cells without influencing other IECs. Single-cell RNA sequencing of mouse colonic tuft cells showed 3 distinct populations of tuft cells, and further showed that a transcriptionally inactive population was expanded markedly in IPMKΔIEC mice, while neuronal-related cells were relatively decreased. CONCLUSIONS Cholinergic output from tuft cells is known to be critical for the restoration of intestinal architecture upon damage, supporting that tuft cell-defective IPMKΔIEC mice are more prone to colitis. Thus, intestinal epithelial IPMK is a critical regulator of colonic integrity and tissue regeneration by determining tuft cell homeostasis and affecting cholinergic output.
Collapse
|
10
|
Nishi K, Fu W, Kiyama R. Novel estrogen-responsive genes (ERGs) for the evaluation of estrogenic activity. PLoS One 2022; 17:e0273164. [PMID: 35976950 PMCID: PMC9385026 DOI: 10.1371/journal.pone.0273164] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/03/2022] [Indexed: 11/19/2022] Open
Abstract
Estrogen action is mediated by various genes, including estrogen-responsive genes (ERGs). ERGs have been used as reporter-genes and markers for gene expression. Gene expression profiling using a set of ERGs has been used to examine statistically reliable transcriptomic assays such as DNA microarray assays and RNA sequencing (RNA-seq). However, the quality of ERGs has not been extensively examined. Here, we obtained a set of 300 ERGs that were newly identified by six sets of RNA-seq data from estrogen-treated and control human breast cancer MCF-7 cells. The ERGs exhibited statistical stability, which was based on the coefficient of variation (CV) analysis, correlation analysis, and examination of the functional association with estrogen action using database searches. A set of the top 30 genes based on CV ranking were further evaluated quantitatively by RT-PCR and qualitatively by a functional analysis using the GO and KEGG databases and by a mechanistic analysis to classify ERα/β-dependent or ER-independent types of transcriptional regulation. The 30 ERGs were characterized according to (1) the enzymes, such as metabolic enzymes, proteases, and protein kinases, (2) the genes with specific cell functions, such as cell-signaling mediators, tumor-suppressors, and the roles in breast cancer, (3) the association with transcriptional regulation, and (4) estrogen-responsiveness. Therefore, the ERGs identified here represent various cell functions and cell signaling pathways, including estrogen signaling, and thus, may be useful to evaluate estrogenic activity.
Collapse
Affiliation(s)
- Kentaro Nishi
- Department of Life Science, Faculty of Life Science, Kyushu Sangyo University Matsukadai, Higashi-ku, Fukuoka, Japan
| | - Wenqiang Fu
- Department of Life Science, Faculty of Life Science, Kyushu Sangyo University Matsukadai, Higashi-ku, Fukuoka, Japan
| | - Ryoiti Kiyama
- Department of Life Science, Faculty of Life Science, Kyushu Sangyo University Matsukadai, Higashi-ku, Fukuoka, Japan
| |
Collapse
|
11
|
Estrogen Receptor β (ESR2) Transcriptome and Chromatin Binding in a Mantle Cell Lymphoma Tumor Model Reveal the Tumor-Suppressing Mechanisms of Estrogens. Cancers (Basel) 2022; 14:cancers14133098. [PMID: 35804870 PMCID: PMC9264873 DOI: 10.3390/cancers14133098] [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: 05/27/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Mantle cell lymphoma (MCL) is much more common in males than in females. The reason for this is not clear, but research has indicated that the female sex hormones, estrogens, have a protective effect on MCL development. To study this further, mice were transplanted with MCL cells and treated with an estrogen that selectively activates ESR2, the main nuclear estrogen receptor in lymphoma cells. The activation of ESR2 resulted in reduced MCL tumor growth of MCL tumors that were both sensitive and resistant to a newly developed drug (ibrutinib). The mechanism for this effect was investigated by analyzing gene expression and ESR2 binding to target genes. The results show that the affected genes were enriched in several malignancy-related biological processes, including MCL. Furthermore, the results suggested an interplay between the lymphoma cells and the tumor microenvironment in response to ESR2 activation. Altogether, the results clarify the mechanisms of ESR2-mediated MCL growth impairment by estrogens and provide a possible explanation for the sex difference in incidence. Furthermore, targeting ESR2 may be an option when considering the treatment of MCL. Abstract Mantle cell lymphoma (MCL) is a non-Hodgkin lymphoma with one of the highest male-to-female incidence ratios. The reason for this is not clear, but epidemiological as well as experimental data have suggested a role for estrogens, particularly acting through estrogen receptor β (ESR2). To study the ESR2 effects on MCL progression, MCL cells sensitive and resistant to the Bruton tyrosine kinase inhibitor ibrutinib were grafted to mice and treated with the ESR2-selective agonist diarylpropionitrile (DPN). The results showed that the DPN treatment of mice grafted with both ibrutinib-sensitive and -resistant MCL tumors resulted in impaired tumor progression. To identify the signaling pathways involved in the impaired tumor progression following ESR2 agonist treatment, the transcriptome and ESR2 binding to target genes were investigated by genome-wide chromatin immunoprecipitation in Granta-519 MCL tumors. DPN-regulated genes were enriched in several biological processes that included cell–cell adhesion, endothelial–mesenchymal transition, nuclear factor-kappaB signaling, vasculogenesis, lymphocyte proliferation, and apoptosis. In addition, downregulation of individual genes, such as SOX11 and MALAT1, that play a role in MCL progression was also observed. Furthermore, the data suggested an interplay between the lymphoma cells and the tumor microenvironment in response to the ESR2 agonist. In conclusion, the results clarify the mechanisms by which estrogens, via ESR2, impair MCL tumor progression and provide a possible explanation for the sex-dependent difference in incidence. Furthermore, targeting ESR2 with a selective agonist may be an additional option when considering the treatment of both ibrutinib-sensitive and -resistant MCL tumors.
Collapse
|
12
|
Indukuri R, Damdimopoulos A, Williams C. An Optimized ChIP-Seq Protocol to Determine Chromatin Binding of Estrogen Receptor Beta. Methods Mol Biol 2022; 2418:203-221. [PMID: 35119668 DOI: 10.1007/978-1-0716-1920-9_13] [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] [Indexed: 06/14/2023]
Abstract
Estrogen regulates transcription through two nuclear receptors, ERα and ERβ, in a tissue and cellular-dependent manner. Both the receptors bind estrogen and activate transcription through direct or indirect interactions with DNA. Revealing their interactions with the chromatin is key to understanding their transcriptional activities and their biological functions. Chromatin-immunoprecipitation followed by sequencing (ChIP-Seq) is a powerful technique to map protein-DNA interactions at precise genomic locations. The genome-wide binding of ERα has been extensively studied. Similar studies of ERβ, however, have been more difficult, in part due to a lack of endogenous expression in cell lines and lack of specific antibodies. In this chapter, we provide an optimized stepwise ChIP protocol for a well-validated ERβ antibody, which is applicable for ChIP-Seq analysis of cell lines with exogenous expression of ERβ.
Collapse
Affiliation(s)
- Rajitha Indukuri
- SciLifeLab, Department of Protein Science, KTH-Royal Institute of Technology, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Anastasios Damdimopoulos
- Bioinformatics and Expression Core, Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Cecilia Williams
- SciLifeLab, Department of Protein Science, KTH-Royal Institute of Technology, Solna, Sweden.
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
13
|
Song D, He H, Indukuri R, Huang Z, Stepanauskaite L, Sinha I, Haldosén LA, Zhao C, Williams C. ERα and ERβ Homodimers in the Same Cellular Context Regulate Distinct Transcriptomes and Functions. Front Endocrinol (Lausanne) 2022; 13:930227. [PMID: 35872983 PMCID: PMC9299245 DOI: 10.3389/fendo.2022.930227] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
The two estrogen receptors ERα and ERβ are nuclear receptors that bind estrogen (E2) and function as ligand-inducible transcription factors. They are homologues and can form dimers with each other and bind to the same estrogen-response element motifs in the DNA. ERα drives breast cancer growth whereas ERβ has been reported to be anti-proliferative. However, they are rarely expressed in the same cells, and it is not fully investigated to which extent their functions are different because of inherent differences or because of different cellular context. To dissect their similarities and differences, we here generated a novel estrogen-dependent cell model where ERα homodimers can be directly compared to ERβ homodimers within the identical cellular context. By using CRISPR-cas9 to delete ERα in breast cancer MCF7 cells with Tet-Off-inducible ERβ expression, we generated MCF7 cells that express ERβ but not ERα. MCF7 (ERβ only) cells exhibited regulation of estrogen-responsive targets in a ligand-dependent manner. We demonstrated that either ER was required for MCF7 proliferation, but while E2 increased proliferation via ERα, it reduced proliferation through a G2/M arrest via ERβ. The two ERs also impacted migration differently. In absence of ligand, ERβ increased migration, but upon E2 treatment, ERβ reduced migration. E2 via ERα, on the other hand, had no significant impact on migration. RNA sequencing revealed that E2 regulated a transcriptome of around 800 genes via each receptor, but over half were specific for either ERα or ERβ (417 and 503 genes, respectively). Functional gene ontology enrichment analysis reinforced that E2 regulated cell proliferation in opposite directions depending on the ER, and that ERβ specifically impacted extracellular matrix organization. We corroborated that ERβ bound to cis-regulatory chromatin of its unique proposed migration-related direct targets ANXA9 and TFAP2C. In conclusion, we demonstrate that within the same cellular context, the two ERs regulate cell proliferation in the opposite manner, impact migration differently, and each receptor also regulates a distinct set of target genes in response to E2. The developed cell model provides a novel and valuable resource to further complement the mechanistic understanding of the two different ER isoforms.
Collapse
Affiliation(s)
- Dandan Song
- Clinical Medical Research Center for Women and Children Diseases, Maternal and Child Health Care Hospital of Shandong Province, Jinan, China
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Huan He
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- School of Public Health, Jilin University, Changchun, China
| | - Rajitha Indukuri
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Solna, Sweden
| | - Zhiqiang Huang
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Lina Stepanauskaite
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Solna, Sweden
| | - Indranil Sinha
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Lars-Arne Haldosén
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Chunyan Zhao
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Cecilia Williams
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Solna, Sweden
- *Correspondence: Cecilia Williams,
| |
Collapse
|
14
|
Estrogen Receptors in Colorectal Cancer: Facts, Novelties and Perspectives. Curr Oncol 2021; 28:4256-4263. [PMID: 34898546 PMCID: PMC8544350 DOI: 10.3390/curroncol28060361] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) is the second cause of cancer-related death in both sexes worldwide. As pre-menopausal women are less likely to develop CRC compared to age-matched men, a protective role for estrogens has been hypothesized. Indeed, two isoforms of nuclear estrogen receptors (ER) have been described: ERα and ERβ. While the binding of 17beta-estradiol to ERα activates anti-apoptotic pathways, the interaction with ERβ activates caspase-3, inducing apoptosis. In this regard, several pieces of evidence show that ERβ tends to be under-regulated in advanced adenomas and CRC, with an opposite trend for ERα. Furthermore, ERβ stimulation slows adenomatous polyp growth and modulates relevant CRC pathways. Based on such considerations, dietary modulation of ER is promising, particularly in subjects with genetic predisposition for CRC. Nevertheless, the main limitation is the lack of clinical trials on a large population scale.
Collapse
|
15
|
Kurmann L, Okoniewski M, Dubey RK. Estradiol Inhibits Human Brain Vascular Pericyte Migration Activity: A Functional and Transcriptomic Analysis. Cells 2021; 10:cells10092314. [PMID: 34571963 PMCID: PMC8472363 DOI: 10.3390/cells10092314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/24/2021] [Accepted: 09/01/2021] [Indexed: 12/24/2022] Open
Abstract
Stroke is the third leading cause of mortality in women and it kills twice as many women as breast cancer. A key role in the pathophysiology of stroke plays the disruption of the blood–brain barrier (BBB) within the neurovascular unit. While estrogen induces vascular protective actions, its influence on stroke remains unclear. Moreover, experiments assessing its impact on endothelial cells to induce barrier integrity are non-conclusive. Since pericytes play an active role in regulating BBB integrity and function, we hypothesize that estradiol may influence BBB by regulating their activity. In this study using human brain vascular pericytes (HBVPs) we investigated the impact of estradiol on key pericyte functions known to influence BBB integrity. HBVPs expressed estrogen receptors (ER-α, ER-β and GPER) and treatment with estradiol (10 nM) inhibited basal cell migration but not proliferation. Since pericyte migration is a hallmark for BBB disruption following injury, infection and inflammation, we investigated the effects of estradiol on TNFα-induced PC migration. Importantly, estradiol prevented TNFα-induced pericyte migration and this effect was mimicked by PPT (ER-α agonist) and DPN (ER-β agonist), but not by G1 (GPR30 agonist). The modulatory effects of estradiol were abrogated by MPP and PHTPP, selective ER-α and ER-β antagonists, respectively, confirming the role of ER-α and ER-β in mediating the anti-migratory actions of estrogen. To delineate the intracellular mechanisms mediating the inhibitory actions of estradiol on PC migration, we investigated the role of AKT and MAPK activation. While estradiol consistently reduced the TNFα-induced MAPK and Akt phosphorylation, only the inhibition of MAPK, but not Akt, significantly abrogated the migratory actions of TNFα. In transendothelial electrical resistance measurements, estradiol induced barrier function (TEER) in human brain microvascular endothelial cells co-cultured with pericytes, but not in HBMECs cultured alone. Importantly, transcriptomics analysis of genes modulated by estradiol in pericytes showed downregulation of genes known to increase cell migration and upregulation of genes known to inhibit cell migration. Taken together, our findings provide the first evidence that estradiol modulates pericyte activity and thereby improves endothelial integrity.
Collapse
Affiliation(s)
- Lisa Kurmann
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland;
| | | | - Raghvendra K. Dubey
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland;
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Correspondence:
| |
Collapse
|