1
|
Yan C, He X, Sun J. The Sex and Age-Associated Infiltration of B Cells May Result in the Dimorphic Behaviors Observed in Papillary Thyroid Carcinomas. Int J Gen Med 2024; 17:3057-3072. [PMID: 39055976 PMCID: PMC11269459 DOI: 10.2147/ijgm.s467704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Background and Purpose Sex and age show a dimorphism role in the pathogenesis, lymph node metastasis, and prognostic outcomes of papillary thyroid carcinoma. This investigation endeavors to elucidate the mechanisms underlying these disparities. Methods The clinicopathological characteristics and risk factors of lymph node metastasis were explored by analyzing the 2261 patients. The gene expression information of 497 samples from The Cancer Genome Atlas Thyroid Cancer database was used to explore the differentially expressed genes in different phenotypes. What's more, the single-cell RNA sequencing data obtained from the Gene Expression Omnibus database was used to explore the gene expression in specific cells. Results Multivariate logistic regression analysis showed that in male patients, a larger tumor size, extrathyroidal extension, younger age, and the presence of calcification emerged as significant predictors for lymph node metastasis (LNM)(p < 0.05). Conversely, female patients exhibited a different profile, with larger tumor size, extrathyroidal extension, younger age, calcification, and bilateral tumors being identified as key risk factors (p < 0.05). Further stratification by age demonstrated distinct patterns: among the younger cohort, a larger tumor size, extrathyroidal extension, male gender, calcification, multifocality, and the presence of Hashimoto's thyroiditis held statistical significance (p < 0.05). In contrast, the older subgroup was characterized by a larger tumor size, extrathyroidal extension, male gender, calcification, bilateral tumors, and unclear margins as salient indicators of risk (p < 0.05). In the bulk gene analysis, there were two sex-age-related differentially expressed genes with a contrary trend in tissue sources and LNM status: TCL1A and CR2. The analysis of single-cell RNA sequencing showed that the infiltration of TCL1A- and CR2-related B cells varied in different clinical subtypes. Conclusion Lymph node metastasis of papillary thyroid carcinoma in different sexes and ages may have distinct patterns, and the ages-sex-related B cell infiltration might explain the dimorphism biological behavior.
Collapse
Affiliation(s)
- Caigu Yan
- Department of General Surgery, the Second Hospital of Tianjin Medical University, Tianjin, People’s Republic of China
| | - Xianghui He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Jinjin Sun
- Department of General Surgery, the Second Hospital of Tianjin Medical University, Tianjin, People’s Republic of China
| |
Collapse
|
2
|
Ho MF, Zhang C, Moon I, Biernacka J, Coombes B, Ngo Q, Skillon C, Skime M, Oesterle T, Croarkin PE, Karpyak VM, Li H, Weinshilboum RM. Epigenetic regulation of GABA catabolism in iPSC-derived neurons: The molecular links between FGF21 and histone methylation. Mol Metab 2023; 77:101798. [PMID: 37689244 PMCID: PMC10514449 DOI: 10.1016/j.molmet.2023.101798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/27/2023] [Accepted: 09/03/2023] [Indexed: 09/11/2023] Open
Abstract
OBJECTIVE Fibroblast growth factor 21 (FGF21) analogs have been tested as potential therapeutics for substance use disorders. Prior research suggests that FGF21 administration might affect alcohol consumption and reward behaviors. Our recent report showed that plasma FGF21 levels were positively correlated with alcohol use in patients with alcohol use disorder (AUD). FGF21 has a short half-life (0.5-2 h) and crosses the blood-brain barrier. Therefore, we set out to identify molecular mechanisms for both the naïve form of FGF21 and a long-acting FGF21 molecule (PF-05231023) in induced pluripotent stem cell (iPSC)-derived forebrain neurons. METHODS We performed RNA-seq in iPSC-derived forebrain neurons treated with naïve FGF21 or PF-05231023 at physiologically relevant concentrations. We obtained plasma levels of FGF21 and GABA from our previous AUD clinical trial (n = 442). We performed ELISA for FGF21 in both iPSC-derived forebrain neurons and forebrain organoids. We determined protein interactions using co-immunoprecipitation. Finally, we applied ChIP assays to confirm the occupancy of REST, EZH2 and H3K27me3 by FGF21 using iPSC-derived forebrain neurons with and without drug exposure. RESULTS We identified 4701 and 1956 differentially expressed genes in response to naïve FGF21 or PF-05231023, respectively (FDR < 0.05). Notably, 974 differentially expressed genes overlapped between treatment with naïve FGF21 and PF-05231023. REST was the most important upstream regulator of differentially expressed genes. The GABAergic synapse pathway was the most significant pathway identified using the overlapping genes. We also observed a significant positive correlation between plasma FGF21 and GABA concentrations in AUD patients. In parallel, FGF21 and PF-05231023 significantly induced GABA levels in iPSC-derived neurons. Finally, functional genomics studies showed a drug-dependent occupancy of REST, EZH2, and H3K27me3 in the promoter regions of genes involved in GABA catabolism which resulted in transcriptional repression. CONCLUSIONS Our results highlight a significant role in the epigenetic regulation of genes involved in GABA catabolism related to FGF21 action. (The ClinicalTrials.gov Identifier: NCT00662571).
Collapse
Affiliation(s)
- Ming-Fen Ho
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
| | - Cheng Zhang
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Irene Moon
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Joanna Biernacka
- Division of Computational Biology, Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Brandon Coombes
- Division of Computational Biology, Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Quyen Ngo
- Hazelden Betty Ford Foundation, Center City, MN, USA
| | | | - Michelle Skime
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Tyler Oesterle
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Paul E Croarkin
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Victor M Karpyak
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Richard M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Ho MF, Zhang C, Moon I, Coombes BJ, Biernacka J, Skime M, Choi DS, Croarkin PE, Frye MA, Ngo Q, Skillon C, Oesterle TS, Karpyak VM, Li H, Weinshilboum RM. Plasma TNFSF10 levels associated with acamprosate treatment response in patients with alcohol use disorder. Front Pharmacol 2022; 13:986238. [PMID: 36120372 PMCID: PMC9475292 DOI: 10.3389/fphar.2022.986238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Acamprosate is an anti-craving drug used in alcohol use disorder (AUD) pharmacotherapy. However, only a subset of patients achieves optimal treatment outcomes. The identification of predictive biomarkers of acamprosate treatment response in patients with AUD would be a substantial advance in addiction medicine. We designed this study to use proteomics data as a quantitative biological trait as a step toward identifying inflammatory modulators that might be associated with acamprosate treatment outcomes. The NIAAA-funded Mayo Clinic Center for the Individualized Treatment of Alcoholism study had previously recruited 442 AUD patients who received 3 months of acamprosate treatment. However, only 267 subjects returned for the 3-month follow-up visit and, as a result, had treatment outcome information available. Baseline alcohol craving intensity was the most significant predictor of acamprosate treatment outcomes. We performed plasma proteomics using the Olink target 96 inflammation panel and identified that baseline plasma TNF superfamily member 10 (TNFSF10) concentration was associated with alcohol craving intensity and variation in acamprosate treatment outcomes among AUD patients. We also performed RNA sequencing using baseline peripheral blood mononuclear cells from AUD patients with known acamprosate treatment outcomes which revealed that inflammation-related pathways were highly associated with relapse to alcohol use during the 3 months of acamprosate treatment. These observations represent an important step toward advancing our understanding of the pathophysiology of AUD and molecular mechanisms associated with acamprosate treatment response. In conclusion, applying omics-based approaches may be a practical approach for identifying biologic markers that could potentially predict alcohol craving intensity and acamprosate treatment response.
Collapse
Affiliation(s)
- Ming-Fen Ho
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Irene Moon
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Brandon J. Coombes
- Division of Computational Biology, Quantitative Health Sciences, Rochester, MN, United States
| | - Joanna Biernacka
- Division of Computational Biology, Quantitative Health Sciences, Rochester, MN, United States
| | - Michelle Skime
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Paul E. Croarkin
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Mark A. Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Quyen Ngo
- Hazelden Betty Ford Foundation, Mayo Clinic, Center City, MN, United States
| | - Cedric Skillon
- Hazelden Betty Ford Foundation, Mayo Clinic, Center City, MN, United States
| | - Tyler S. Oesterle
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Victor M. Karpyak
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Richard M. Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
4
|
17β-estradiol binding to ERα promotes the progression of prolactinoma through estrogen-response element-induced CaBP-9k up-regulation. Biosci Rep 2020; 40:221738. [PMID: 31894842 PMCID: PMC6960063 DOI: 10.1042/bsr20191330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 08/19/2019] [Accepted: 09/03/2019] [Indexed: 12/26/2022] Open
Abstract
17β-estradiol (E2) is considered to be an important instigator of prolactinoma, and can positively regulate the expression of calbindin-D9k (CaBP-9k) which contains an estrogen responsive element (ERE) via estrogen receptors (ERs). However, the detailed mechanism of E2 in promoting CaBP-9k expression and their roles in prolactinoma progression remain unclear. Here, we aimed to characterize it. The luciferase gene reporter assay with luc-ERE transfection showed that E2 treatment significantly enhanced the transcriptional level of CaBP-9k, whereas CaBP-9k activity was reduced when GH3 and MMQ cells were treated with AZD9496, an antagonist of ERα. E2 treatment increased the protein expressions of CaBP-9k and ERα but not ERβ, whereas this effect was also abolished when cells were treated with AZD9496. Besides, immunoprecipitation (IP) and immunofluorescence assays demonstrated that CaBP-9k could directly interact with ERα not ERβ, and Chromatin IP (ChIP) assay showed that ERα could bind to ERE of the CaBP-9k promoter. Moreover, cell counting kit-8 (CCK-8) and flow cytometry assays showed that E2 treatment significantly enhanced cell viability and inhibited cell apoptosis, but these effects were all abolished when ERα was down-regulated by short hairpin RNA (shRNA) or inhibited by AZD9496, as well as CaBP-9K suppression in both GH3 and MMQ cell lines. Taken together, these findings indicated that E2 stimulation promoted prolactin cell proliferation and inhibited cell apoptosis through ERα-induced CaBP-9k up-regulation, which then accelerated the advanced progression of prolactinoma.
Collapse
|
5
|
Moradifard S, Hoseinbeyki M, Emam MM, Parchiniparchin F, Ebrahimi-Rad M. Association of the Sp1 binding site and -1997 promoter variations in COL1A1 with osteoporosis risk: The application of meta-analysis and bioinformatics approaches offers a new perspective for future research. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2020; 786:108339. [PMID: 33339581 DOI: 10.1016/j.mrrev.2020.108339] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 08/11/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022]
Abstract
As a complex disease, osteoporosis is influenced by several genetic markers. Many studies have examined the link between the Sp1 binding site +1245 G > T (rs1800012) and -1997 G > T (rs1107946) variations in the COL1A1 gene with osteoporosis risk. However, the findings of these studies have been contradictory; therefore, we performed a meta-analysis to aggregate additional information and obtain increased statistical power to more efficiently estimate this correlation. A meta-analysis was conducted with studies published between 1991-2020 that were identified by a systematic electronic search of the Scopus and Clarivate Analytics databases. Studies with bone mineral density (BMD) data and complete genotypes of the single-nucleotide variations (SNVs) for the overall and postmenopausal female population were included in this meta-analysis and analyzed using the R metaphor package. A relationship between rs1800012 and significantly decreased BMD values at the lumbar spine and femoral neck was found in individuals carrying the "ss" versus the "SS" genotype in the overall population according to a random effects model (p < 0.0001). Similar results were also found in the postmenopausal female population (p = 0.003 and 0.0002, respectively). Such findings might be an indication of increased osteoporosis risk in both studied groups in individuals with the "ss" genotype. Although no association was identified between the -1997 G > T and low BMD in the overall population, those individuals with the "GT" genotype showed a higher level of BMD than those with "GG" in the subgroup analysis (p = 0.007). To determine which transcription factor (TF) might bind to the -1997 G > T in COL1A1, 45 TFs were identified based on bioinformatics predictions. According to the GSE35958 microarray dataset, 16 of 45 TFs showed differential expression profiles in osteoporotic human mesenchymal stem cells relative to normal samples from elderly donors. By identifying candidate TFs for the -1997 G > T site, our study offers a new perspective for future research.
Collapse
Affiliation(s)
| | | | - Mohammad Mehdi Emam
- Rheumatology Ward, Loghman Hospital, Shahid Beheshti Medical University (SBMU), Tehran, Iran
| | | | | |
Collapse
|
6
|
Koch M, Reinartz S, Saggau J, Knittel G, Rosen N, Fedorchenko O, Thelen L, Barthel R, Reinart N, Seeger-Nukpezah T, Reinhardt HC, Hallek M, Nguyen PH. Meta-Analysis Reveals Significant Sex Differences in Chronic Lymphocytic Leukemia Progression in the Eµ-TCL1 Transgenic Mouse Model. Cancers (Basel) 2020; 12:cancers12071980. [PMID: 32698538 PMCID: PMC7409315 DOI: 10.3390/cancers12071980] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/08/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023] Open
Abstract
The Eµ-TCL1 transgenic mouse model represents the most widely and extensively used animal model for chronic lymphocytic leukemia (CLL). In this report, we performed a meta-analysis of leukemia progression in over 300 individual Eµ-TCL1 transgenic mice and discovered a significantly accelerated disease progression in females compared to males. This difference is also reflected in an aggressive CLL mouse model with additional deletion of Tp53 besides the TCL1 transgene. Moreover, after serial adoptive transplantation of murine CLL cells, female recipients also succumbed to CLL earlier than male recipients. This sex-related disparity in the murine models is markedly contradictory to the human CLL condition. Thus, due to our observation we urge both careful consideration in the experimental design and accurate description of the Eµ-TCL1 transgenic cohorts in future studies.
Collapse
Affiliation(s)
- Maximilian Koch
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
| | - Sebastian Reinartz
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
| | - Julia Saggau
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
| | - Gero Knittel
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
| | - Natascha Rosen
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
| | - Oleg Fedorchenko
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
| | - Lisa Thelen
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
| | - Romy Barthel
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
| | - Nina Reinart
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
| | - Tamina Seeger-Nukpezah
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
| | - Hans Christian Reinhardt
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
- Clinic for Hematology, West German Cancer Center, University Hospital Essen, Essen, German Cancer Consortium (DKTK), 45147 Essen, Germany
| | - Michael Hallek
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
| | - Phuong-Hien Nguyen
- University of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine Cologne, CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, 50931 Cologne, Germany; (M.K.); (S.R.); (J.S.); (G.K.); (N.R.); (O.F.); (L.T.); (R.B.); (N.R.); (T.S.-N.); (H.C.R.); (M.H.)
- Correspondence: ; Tel.: +49-221-478-84120; Fax: +49-221-478-84115
| |
Collapse
|
7
|
Ishikawa A, Wada T, Nishimura S, Ito T, Okekawa A, Onogi Y, Watanabe E, Sameshima A, Tanaka T, Tsuneki H, Saito S, Sasaoka T. Estrogen regulates sex-specific localization of regulatory T cells in adipose tissue of obese female mice. PLoS One 2020; 15:e0230885. [PMID: 32240221 PMCID: PMC7117686 DOI: 10.1371/journal.pone.0230885] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 03/10/2020] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cells (Treg) play essential roles in maintaining immune homeostasis. Resident Treg in visceral adipose tissue (VAT-Treg) decrease in male obese mice, which leads to the development of obesity-associated chronic inflammations and insulin resistance. Although gender differences in immune responses have been reported, the effects of the difference in metabolic environment on VAT-Treg are unclear. We investigated the localization of VAT-Treg in female mice in comparison with that in male mice. On a high-fat diet (HFD), VAT-Treg decreased in male mice but increased in female mice. The increase was abolished in ovariectomized and HFD-fed mice, but was restored by estrogen supplementation. The IL33 receptor ST2, which is important for the localization and maturation of VAT-Treg in males, was reduced in CD4+CD25+ T cells isolated from gonadal fat of obese mice of both genders, suggesting that a different system exists for VAT-Treg localization in females. Extensive analysis of chemokine expression in gonadal fat and adipose CD4+CD25+T cells revealed several chemokine signals related to female-specific VAT-Treg accumulation such as CCL24, CCR6, and CXCR3. Taken together, the current study demonstrated sexual dimorphism in VAT-Treg localization in obese mice. Estrogen may attenuate obesity-associated chronic inflammation partly through altering chemokine-related VAT-Treg localization in females.
Collapse
Affiliation(s)
- Akari Ishikawa
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Tsutomu Wada
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Sanshiro Nishimura
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Tetsuo Ito
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Akira Okekawa
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Yasuhiro Onogi
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Eri Watanabe
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Azusa Sameshima
- Department of Obstetrics and Gynecology, University of Toyama, Toyama, Japan
| | - Tomoko Tanaka
- Department of Obstetrics and Gynecology, University of Toyama, Toyama, Japan
| | - Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Shigeru Saito
- Department of Obstetrics and Gynecology, University of Toyama, Toyama, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| |
Collapse
|
8
|
Genetic predictors to acupuncture response for hot flashes: an exploratory study of breast cancer survivors. ACTA ACUST UNITED AC 2020; 27:913-917. [PMID: 32217888 DOI: 10.1097/gme.0000000000001545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Because hot flashes are a common symptom experienced by women with breast cancer, we sought to explore genetic predictors associated with response to acupuncture for the treatment of hot flashes. METHODS Using data from our completed randomized controlled trial (Clinicaltrials.gov identifier: NCT01005108) on hot flashes among breast cancer survivors who provided biomarker collection (N = 108), we extracted and assayed DNA for single nucleotide polymorphisms in genes involved in neurotransmission, thermoregulation, and inflammation (ADORA1, COMT, TCL1A, and TRPV1). For our primary outcome we classified individuals with a 50% or more reduction in their hot flash composite score at the end of treatment as responders. We used Fisher exact test to identify individual and combined single nucleotide polymorphisms associated with treatment response. RESULTS Among women (N = 57) who received acupuncture treatment (electro or sham), we found that women who were carriers of at least one of these six genotypes (ADORA1 rs41264025-GA or rs16851029-GG or rs12744240-GT, COMT rs6269-GA, TCL1A rs2369049-GG, and TRPV1 rs8065080-TT) were more likely to respond to acupuncture for hot flashes than noncarriers (70.3% vs 37.5%, P = 0.035). These six genotypes were not associated with response in women (N = 51) who received pharmacological hot flash treatment (gabapentin or placebo pill; 37.5% vs 37.5%, P = 1.0). CONCLUSIONS In this exploratory, proof of concept study, we identified six genotypes that may predict response to acupuncture for hot flashes in breast cancer survivors. If confirmed by future studies, these findings may inform the development of personalized acupuncture for managing hot flashes.
Collapse
|
9
|
Romero SAD, Su HI, Satagopan J, Li QS, Seluzicki CM, Dries A, DeMichele AM, Mao JJ. Clinical and genetic risk factors for aromatase inhibitor-associated arthralgia in breast cancer survivors. Breast 2019; 49:48-54. [PMID: 31678641 PMCID: PMC7375589 DOI: 10.1016/j.breast.2019.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/08/2019] [Accepted: 10/16/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Arthralgia is a common and debilitating toxicity of aromatase inhibitors (AI) that leads to premature drug discontinuation. We sought to evaluate the clinical and genetic risk factors associated with AI-associated arthralgia (AIAA). METHODS We performed a cross-sectional study among postmenopausal women with stage 0-III breast cancer who were prescribed a third-generation AI for adjuvant therapy. The primary outcome was patient-reported AIAA occurrence. We extracted and assayed germline DNA for single nucleotide polymorphisms (SNPs) of genes implicated in estrogen and inflammation pathways. Multivariable logistic regression models examined the association between demographic, clinical, and genetic factors and AIAA. Analyses were restricted to White participants. RESULTS Among 1049 White participants, 543 (52%) reported AIAA. In multivariable analyses, women who had a college education [Adjusted Odds Ratio (AOR) 1.49, 95% Confidence Interval (CI) 1.00-2.20], had a more recent transition into menopause (<10 years) (5-10 years AOR 1.55, 95% CI 1.09-2.22; <5 years AOR 1.78, 95% CI 1.18-2.67), were within one year of starting AIs (AOR 1.61, 95% CI 1.08-2.40), and those who received chemotherapy (AOR 1.38, 95% CI 1.02-1.88) were significantly more likely to report AIAA. Additionally, SNP rs11648233 (HSD17B2) was significantly associated with higher odds of AIAA (AOR 2.21, 95% CI 1.55-3.16). CONCLUSIONS Time since menopause and start of AIs, prior chemotherapy, and SNP rs11648233 within the HSD17B2 gene in the estrogen pathway were significantly associated with patient-reported AIAA. These findings suggest that clinical and genetic factors involved in estrogen withdrawal increase the risk of AIAA in postmenopausal breast cancer survivors.
Collapse
Affiliation(s)
- Sally A D Romero
- Memorial Sloan Kettering Cancer Center, Bendheim Integrative Medicine Center, 1429 First Avenue, New York, NY, 10021, USA.
| | - H Irene Su
- Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive, La Jolla, CA, 92093, USA.
| | - Jaya Satagopan
- Rutgers School of Public Health, 683 Hoes Lane West, Piscataway, NJ, 08854, USA.
| | - Q Susan Li
- Memorial Sloan Kettering Cancer Center, Bendheim Integrative Medicine Center, 1429 First Avenue, New York, NY, 10021, USA.
| | - Christina M Seluzicki
- Memorial Sloan Kettering Cancer Center, Bendheim Integrative Medicine Center, 1429 First Avenue, New York, NY, 10021, USA.
| | - Annika Dries
- Stanford University School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA.
| | - Angela M DeMichele
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
| | - Jun J Mao
- Memorial Sloan Kettering Cancer Center, Bendheim Integrative Medicine Center, 1429 First Avenue, New York, NY, 10021, USA.
| |
Collapse
|
10
|
Genovese TJ, Mao JJ. Genetic Predictors of Response to Acupuncture for Aromatase Inhibitor-Associated Arthralgia Among Breast Cancer Survivors. PAIN MEDICINE 2019; 20:191-194. [PMID: 29912452 DOI: 10.1093/pm/pny067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective To evaluate the associations between polymorphisms in two genes, catechol-O-methyltransferase and T-cell leukemia/lymphoma 1 A, and acupuncture-mediated pain reduction among breast cancer survivors with aromatase inhibitor-associated arthralgia. Design, Setting, and Subjects Biospecimens were obtained from 38 patients enrolled in a clinical trial of acupuncture for aromatase inhibitor-associated arthralgia in postmenopausal hormone receptor-positive breast cancer survivors. Methods We used polymerase chain reaction to genotype the rs4680 (Val158Met) and rs4633 (His62His) variants in the catechol-O-methyltransferase gene and rs2369049 (A > G) and rs7158782 (A > G) variants in the T-cell leukemia/lymphoma 1 A gene. Response to acupuncture was defined by 30% reduction in end-of-treatment average pain, measured by the Brief Pain Inventory. We used Fisher exact tests to evaluate associations between genotype and treatment response. Results Among participants, all six (15.8%) subjects who expressed AA in locus rs4680 responded to acupuncture. In a combined analysis, the 18 (47.4%) subjects with the responder genotype at either rs4680 (AA) or rs2369049 (GG or AG) were significantly more likely to respond to acupuncture than those without (77.8% vs 45.0%, P = 0.039). Conclusions Specific genetic variations at loci rs4680 and rs2369049 are associated with response to acupuncture-type intervention for management of arthralgia. These results serve as a proof of concept for applying a precision medicine framework to the study of cancer pain management.
Collapse
Affiliation(s)
- Timothy J Genovese
- The Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Jun J Mao
- Department of Integrative Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
11
|
Neavin DR, Liu D, Ray B, Weinshilboum RM. The Role of the Aryl Hydrocarbon Receptor (AHR) in Immune and Inflammatory Diseases. Int J Mol Sci 2018; 19:ijms19123851. [PMID: 30513921 PMCID: PMC6321643 DOI: 10.3390/ijms19123851] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 12/17/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a nuclear receptor that modulates the response to environmental stimuli. It was recognized historically for its role in toxicology but, in recent decades, it has been increasingly recognized as an important modulator of disease—especially for its role in modulating immune and inflammatory responses. AHR has been implicated in many diseases that are driven by immune/inflammatory processes, including major depressive disorder, multiple sclerosis, rheumatoid arthritis, asthma, and allergic responses, among others. The mechanisms by which AHR has been suggested to impact immune/inflammatory diseases include targeted gene expression and altered immune differentiation. It has been suggested that single nucleotide polymorphisms (SNPs) that are near AHR-regulated genes may contribute to AHR-dependent disease mechanisms/pathways. Further, we have found that SNPs that are outside of nuclear receptor binding sites (i.e., outside of AHR response elements (AHREs)) may contribute to AHR-dependent gene regulation in a SNP- and ligand-dependent manner. This review will discuss the evidence and mechanisms of AHR contributions to immune/inflammatory diseases and will consider the possibility that SNPs that are outside of AHR binding sites might contribute to AHR ligand-dependent inter-individual variation in disease pathophysiology and response to pharmacotherapeutics.
Collapse
Affiliation(s)
- Drew R Neavin
- Mayo Clinic Graduate School of Biomedical Sciences, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA.
| | - Duan Liu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA.
| | - Balmiki Ray
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA.
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA.
| |
Collapse
|
12
|
Merkulov VM, Leberfarb EY, Merkulova TI. Regulatory SNPs and their widespread effects on the transcriptome. J Biosci 2018; 43:1069-1075. [PMID: 30541964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Currently, it is generally accepted that the cis-acting effects of noncoding variants on gene expression are a major factor for phenotypic variation in complex traits and disease susceptibility. Meanwhile, the protein products of many target genes for the identified cis-regulatory variants (rSNPs) are regulatory molecules themselves (transcription factors, effectors, components of signal transduction pathways, etc.), which implies dramatic downstream effects of these variations on complex gene networks. Here, we brief the results of recent most comprehensive studies on the role of rSNPs in transcriptional regulation across the genome.
Collapse
Affiliation(s)
- Vasily M Merkulov
- Laboratory of Gene Expression Regulation, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | | | | |
Collapse
|
13
|
Merkulov VM, Leberfarb EY, Merkulova TI. Regulatory SNPs and their widespread effects on the transcriptome. J Biosci 2018. [DOI: 10.1007/s12038-018-9817-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
14
|
Liu D, Qin S, Ray B, Kalari KR, Wang L, Weinshilboum RM. Single Nucleotide Polymorphisms (SNPs) Distant from Xenobiotic Response Elements Can Modulate Aryl Hydrocarbon Receptor Function: SNP-Dependent CYP1A1 Induction. Drug Metab Dispos 2018; 46:1372-1381. [PMID: 29980579 PMCID: PMC6090174 DOI: 10.1124/dmd.118.082164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 06/28/2018] [Indexed: 12/20/2022] Open
Abstract
CYP1A1 expression can be upregulated by the ligand-activated aryl hydrocarbon receptor (AHR). Based on prior observations with estrogen receptors and estrogen response elements, we tested the hypothesis that single-nucleotide polymorphisms (SNPs) mapping hundreds of base pairs (bp) from xenobiotic response elements (XREs) might influence AHR binding and subsequent gene expression. Specifically, we analyzed DNA sequences 5 kb upstream and downstream of the CYP1A1 gene for putative XREs. SNPs located ±500 bp of these putative XREs were studied using a genomic data-rich human lymphoblastoid cell line (LCL) model system. CYP1A1 mRNA levels were determined after treatment with varying concentrations of 3-methylcholanthrene (3MC). The rs2470893 (-1694G>A) SNP, located 196 bp from an XRE in the CYP1A1 promoter, was associated with 2-fold variation in AHR-XRE binding in a SNP-dependent fashion. LCLs with the AA genotype displayed significantly higher AHR-XRE binding and CYP1A1 mRNA expression after 3MC treatment than did those with the GG genotype. Electrophoretic mobility shift assay (EMSA) showed that oligonucleotides with the AA genotype displayed higher LCL nuclear extract binding after 3MC treatment than did those with the GG genotype, and mass spectrometric analysis of EMSA protein-DNA complex bands identified three candidate proteins, two of which were co-immunoprecipitated with AHR. In conclusion, we have demonstrated that the rs2470893 SNP, which maps 196 bp from a CYP1A1 promoter XRE, is associated with variations in 3MC-dependent AHR binding and CYP1A1 expression. Similar "distant SNP effects" on AHR binding to an XRE motif and subsequent gene expression might occur for additional AHR-regulated genes.
Collapse
Affiliation(s)
- Duan Liu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (D.L., S.Q., B.R., L.W., R.M.W.) and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota
| | - Sisi Qin
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (D.L., S.Q., B.R., L.W., R.M.W.) and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota
| | - Balmiki Ray
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (D.L., S.Q., B.R., L.W., R.M.W.) and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota
| | - Krishna R Kalari
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (D.L., S.Q., B.R., L.W., R.M.W.) and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (D.L., S.Q., B.R., L.W., R.M.W.) and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (D.L., S.Q., B.R., L.W., R.M.W.) and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
15
|
Ho MF, Lummertz da Rocha E, Zhang C, Ingle JN, Goss PE, Shepherd LE, Kubo M, Wang L, Li H, Weinshilboum RM. TCL1A, a Novel Transcription Factor and a Coregulator of Nuclear Factor κB p65: Single Nucleotide Polymorphism and Estrogen Dependence. J Pharmacol Exp Ther 2018; 365:700-710. [PMID: 29592948 DOI: 10.1124/jpet.118.247718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/19/2018] [Indexed: 01/10/2023] Open
Abstract
T-cell leukemia 1A (TCL1A) single-nucleotide polymorphisms (SNPs) have been associated with aromatase inhibitor-induced musculoskeletal adverse events. We previously demonstrated that TCL1A is inducible by estradiol (E2) and plays a critical role in the regulation of cytokines, chemokines, and Toll-like receptors in a TCL1A SNP genotype and estrogen-dependent fashion. Furthermore, TCLIA SNP-dependent expression phenotypes can be "reversed" by exposure to selective estrogen receptor modulators such as 4-hydroxytamoxifen (4OH-TAM). The present study was designed to comprehensively characterize the role of TCL1A in transcriptional regulation across the genome by performing RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) assays with lymphoblastoid cell lines. RNA-seq identified 357 genes that were regulated in a TCL1A SNP- and E2-dependent fashion with expression patterns that were 4OH-TAM reversible. ChIP-seq for the same cells identified 57 TCL1A binding sites that could be regulated by E2 in a SNP-dependent fashion. Even more striking, nuclear factor-κB (NF-κB) p65 bound to those same DNA regions. In summary, TCL1A is a novel transcription factor with expression that is regulated in a SNP- and E2-dependent fashion-a pattern of expression that can be reversed by 4OH-TAM. Integrated RNA-seq and ChIP-seq results suggest that TCL1A also acts as a transcriptional coregulator with NF-κB p65, an important immune system transcription factor.
Collapse
Affiliation(s)
- Ming-Fen Ho
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.H., E.L.d.R., C.Z., L.W., H.L., R.M.W.), and Division of Medical Oncology, Department of Oncology (J.N.I.), Mayo Clinic, Rochester, Minnesota; Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); Canadian Cancer Trials Group, Kingston, Ontario, Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Yokohama, Japan (M.K.)
| | - Edroaldo Lummertz da Rocha
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.H., E.L.d.R., C.Z., L.W., H.L., R.M.W.), and Division of Medical Oncology, Department of Oncology (J.N.I.), Mayo Clinic, Rochester, Minnesota; Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); Canadian Cancer Trials Group, Kingston, Ontario, Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Yokohama, Japan (M.K.)
| | - Cheng Zhang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.H., E.L.d.R., C.Z., L.W., H.L., R.M.W.), and Division of Medical Oncology, Department of Oncology (J.N.I.), Mayo Clinic, Rochester, Minnesota; Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); Canadian Cancer Trials Group, Kingston, Ontario, Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Yokohama, Japan (M.K.)
| | - James N Ingle
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.H., E.L.d.R., C.Z., L.W., H.L., R.M.W.), and Division of Medical Oncology, Department of Oncology (J.N.I.), Mayo Clinic, Rochester, Minnesota; Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); Canadian Cancer Trials Group, Kingston, Ontario, Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Yokohama, Japan (M.K.)
| | - Paul E Goss
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.H., E.L.d.R., C.Z., L.W., H.L., R.M.W.), and Division of Medical Oncology, Department of Oncology (J.N.I.), Mayo Clinic, Rochester, Minnesota; Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); Canadian Cancer Trials Group, Kingston, Ontario, Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Yokohama, Japan (M.K.)
| | - Lois E Shepherd
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.H., E.L.d.R., C.Z., L.W., H.L., R.M.W.), and Division of Medical Oncology, Department of Oncology (J.N.I.), Mayo Clinic, Rochester, Minnesota; Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); Canadian Cancer Trials Group, Kingston, Ontario, Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Yokohama, Japan (M.K.)
| | - Michiaki Kubo
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.H., E.L.d.R., C.Z., L.W., H.L., R.M.W.), and Division of Medical Oncology, Department of Oncology (J.N.I.), Mayo Clinic, Rochester, Minnesota; Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); Canadian Cancer Trials Group, Kingston, Ontario, Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Yokohama, Japan (M.K.)
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.H., E.L.d.R., C.Z., L.W., H.L., R.M.W.), and Division of Medical Oncology, Department of Oncology (J.N.I.), Mayo Clinic, Rochester, Minnesota; Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); Canadian Cancer Trials Group, Kingston, Ontario, Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Yokohama, Japan (M.K.)
| | - Hu Li
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.H., E.L.d.R., C.Z., L.W., H.L., R.M.W.), and Division of Medical Oncology, Department of Oncology (J.N.I.), Mayo Clinic, Rochester, Minnesota; Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); Canadian Cancer Trials Group, Kingston, Ontario, Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Yokohama, Japan (M.K.)
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.H., E.L.d.R., C.Z., L.W., H.L., R.M.W.), and Division of Medical Oncology, Department of Oncology (J.N.I.), Mayo Clinic, Rochester, Minnesota; Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); Canadian Cancer Trials Group, Kingston, Ontario, Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Yokohama, Japan (M.K.)
| |
Collapse
|
16
|
Wang L, Ingle J, Weinshilboum R. Pharmacogenomic Discovery to Function and Mechanism: Breast Cancer as a Case Study. Clin Pharmacol Ther 2017; 103:243-252. [PMID: 29052219 PMCID: PMC5760458 DOI: 10.1002/cpt.915] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/03/2017] [Accepted: 10/07/2017] [Indexed: 12/22/2022]
Abstract
Biomedical research is undergoing rapid change, with the development of a series of analytical omics techniques that are capable of generating Biomedical Big Data. These developments provide an unprecedented opportunity to gain novel insight into disease pathophysiology and mechanisms of drug action and response-but they also present significant challenges. Pharmacogenomics is a discipline within Clinical Pharmacology that has been at the forefront in defining, taking advantage of, and dealing with the opportunities and challenges of this aspect of the Post-Genome Project world. This overview will describe the evolution of germline pharmacogenomic research strategies as we have moved from an era of candidate genes to agnostic genome-wide association studies (GWAS) coupled with the functional and mechanistic pursuit of GWAS signals. Germline pharmacogenomic studies of breast cancer endocrine therapy will be used to illustrate research strategies that are being applied broadly to omics studies of drug response phenotypes.
Collapse
Affiliation(s)
- Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - James Ingle
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Richard Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
17
|
Weinshilboum RM, Wang L. Pharmacogenomics: Precision Medicine and Drug Response. Mayo Clin Proc 2017; 92:1711-1722. [PMID: 29101939 PMCID: PMC5682947 DOI: 10.1016/j.mayocp.2017.09.001] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/17/2017] [Accepted: 09/05/2017] [Indexed: 11/21/2022]
Abstract
Pharmacogenomics is the use of genomic and other "omic" information to individualize drug selection and drug use to avoid adverse drug reactions and to maximize drug efficacy. The science underlying pharmacogenomics has evolved rapidly over the 50 years since it was first suggested that genetics might influence drug response phenotypes. That process has occurred in parallel with advances in DNA sequencing and other molecular technologies, with striking increases in our understanding of the human genome. There are now many validated examples of the clinical utility of pharmacogenomics, and this type of clinical genomic information is increasingly being generated in clinical laboratories, incorporated into electronic health records, and used to "tailor" or individualize drug therapy. This review will survey the origins and development of pharmacogenomics; it will address some of the challenges associated with the clinical implementation of pharmacogenomics; and it will attempt to foresee future advances in this important genomic discipline, one that almost certainly will be among the earliest and most widely adopted aspects of clinical genomics.
Collapse
Affiliation(s)
- Richard M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN.
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| |
Collapse
|
18
|
Liu D, Ho MF, Schaid DJ, Scherer SE, Kalari K, Liu M, Biernacka J, Yee V, Evans J, Carlson E, Goetz MP, Kubo M, Wickerham DL, Wang L, Ingle JN, Weinshilboum RM. Breast cancer chemoprevention pharmacogenomics: Deep sequencing and functional genomics of the ZNF423 and CTSO genes. NPJ Breast Cancer 2017; 3:30. [PMID: 28856246 PMCID: PMC5566425 DOI: 10.1038/s41523-017-0036-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 07/01/2017] [Accepted: 07/12/2017] [Indexed: 12/14/2022] Open
Abstract
Our previous GWAS using samples from the NSABP P-1 and P-2 selective estrogen receptor modulator (SERM) breast cancer prevention trials identified SNPs in ZNF423 and near CTSO that were associated with breast cancer risk during SERM chemoprevention. We have now performed Next Generation DNA sequencing to identify additional SNPs that might contribute to breast cancer risk and to extend our observation that SNPs located hundreds of bp from estrogen response elements (EREs) can alter estrogen receptor alpha (ERα) binding in a SERM-dependent fashion. Our study utilized a nested case-control cohort selected from patients enrolled in the original GWAS, with 199 cases who developed breast cancer during SERM therapy and 201 matched controls who did not. We resequenced approximately 500 kb across both ZNF423 and CTSO, followed by functional genomic studies. We identified 4079 SNPs across ZNF423 and 3876 across CTSO, with 9 SNPs in ZNF423 and 12 in CTSO with p < 1E-02 that were within 500 bp of an ERE motif. The rs746157 (p = 8.44E-04) and rs12918288 SNPs (p = 3.43E-03) in intron 5 of ZNF423, were in linkage equilibrium and were associated with alterations in ER-binding to an ERE motif distant from these SNPs. We also studied all nonsynonymous SNPs in both genes and observed that one nsSNP in ZNF423 displayed decreased protein expression. In conclusion, we identified additional functional SNPs in ZNF423 that were associated with SNP and SERM-dependent alternations in ER binding and transcriptional regulation for an ERE at a distance from the SNPs, thus providing novel insight into mechanisms of SERM effect.
Collapse
Affiliation(s)
- Duan Liu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA
| | - Ming-Fen Ho
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA
| | - Daniel J Schaid
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN USA
| | - Steven E Scherer
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX USA
| | - Krishna Kalari
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN USA
| | - Mohan Liu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA
| | - Joanna Biernacka
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN USA
| | - Vivien Yee
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH USA
| | - Jared Evans
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN USA
| | - Erin Carlson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN USA
| | - Matthew P Goetz
- Division of Medical Oncology, Mayo Clinic, Rochester, MN USA
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Science, Yokohama, Japan
| | - D Lawrence Wickerham
- Section of Cancer Genetics and Prevention, Allegheny General Hospital and the National Surgical Adjuvant Breast and Bowel Project (NSABP), Pittsburgh, PA USA
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA
| | - James N Ingle
- Division of Medical Oncology, Mayo Clinic, Rochester, MN USA
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
19
|
Qin S, Ingle JN, Liu M, Yu J, Wickerham DL, Kubo M, Weinshilboum RM, Wang L. Calmodulin-like protein 3 is an estrogen receptor alpha coregulator for gene expression and drug response in a SNP, estrogen, and SERM-dependent fashion. Breast Cancer Res 2017; 19:95. [PMID: 28821270 PMCID: PMC5562991 DOI: 10.1186/s13058-017-0890-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/04/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND We previously performed a case-control genome-wide association study in women treated with selective estrogen receptor modulators (SERMs) for breast cancer prevention and identified single nucleotide polymorphisms (SNPs) in ZNF423 as potential biomarkers for response to SERM therapy. The ZNF423rs9940645 SNP, which is approximately 200 bp away from the estrogen response elements, resulted in the SNP, estrogen, and SERM-dependent regulation of ZNF423 expression and, "downstream", that of BRCA1. METHODS Electrophoretic mobility shift assay-mass spectrometry was performed to identify proteins binding to the ZNF423 SNP and coordinating with estrogen receptor alpha (ERα). Clustered, regularly interspaced short palindromic repeats (CRISPR)/Cas9 genome editing was applied to generate ZR75-1 breast cancer cells with different ZNF423 SNP genotypes. Both cultured cells and mouse xenograft models with different ZNF423 SNP genotypes were used to study the cellular responses to SERMs and poly(ADP-ribose) polymerase (PARP) inhibitors. RESULTS We identified calmodulin-like protein 3 (CALML3) as a key sensor of this SNP and a coregulator of ERα, which contributes to differential gene transcription regulation in an estrogen and SERM-dependent fashion. Furthermore, using CRISPR/Cas9-engineered ZR75-1 breast cancer cells with different ZNF423 SNP genotypes, striking differences in cellular responses to SERMs and PARP inhibitors, alone or in combination, were observed not only in cells but also in a mouse xenograft model. CONCLUSIONS Our results have demonstrated the mechanism by which the ZNF423 rs9940645 SNP might regulate gene expression and drug response as well as its potential role in achieving more highly individualized breast cancer therapy.
Collapse
Affiliation(s)
- Sisi Qin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - James N Ingle
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Mohan Liu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Jia Yu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - D Lawrence Wickerham
- Section of Cancer Genetics and Prevention, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA.,National Surgical Adjuvant Breast and Bowel Project (NRG Oncology), Pittsburgh, PA, USA
| | - Michiaki Kubo
- Laboratory for Genotyping Development, Center for Genomic Medicine, RIKEN, Yokohama, Japan
| | - Richard M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
20
|
Ho MF, Weinshilboum RM. Immune Mediator Pharmacogenomics: TCL1A SNPs and Estrogen-Dependent Regulation of Inflammation. JOURNAL OF NATURE AND SCIENCE 2017; 3:e416. [PMID: 28868359 PMCID: PMC5578609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
This review describes the important functional implications of TCL1A single nucleotide polymorphisms (SNPs) discovered during pharmacogenomic studies of aromatase inhibitor-induced musculoskeletal adverse events that were subsequently shown to influence the expression of cytokines, chemokines, toll-like receptors (TLR), and NF-κB in a SNP and estrogen-dependent fashion. Functional genomic studies of these SNPs led to the discovery of novel mechanisms that may contribute to disease pathophysiology and which may also increase our understanding of pharmacogenomic aspects of regulation of the expression of inflammatory mediators. Specifically, TCL1A expression was induced by estrogens in a SNP-dependent fashion, resulting in downstream effects on the expression of immune mediators that included IL17RA, IL17A, CCR6, CCL20 TLR2, TLR7, TLR9, TLR10 and NF-κB. These observations have potential implications for inflammatory diseases such as rheumatoid arthritis-a disease for which two thirds of patients are women. Strikingly, this genomic phenomenon could be "reversed" by estrogen receptor antagonist treatment-once again in a SNP-dependent, i.e., in a pharmacogenomic fashion. Specifically, differential SNP-dependent effects on estrogen receptor binding to estrogen response elements before and after estrogen receptor blockade might be associated with mechanisms underlying the SNP genotype and estrogen-dependent regulation of TCL1A and the expression of downstream immune mediators. Furthermore, this SNP and estrogen-dependent phenotypic response could be "reversed" by SERM treatment. These observations could potentially open the way to understand, predict and even pharmacologically manipulate the expression of selected immune mediators in a SNP-dependent fashion.
Collapse
Affiliation(s)
- Ming-Fen Ho
- Correspondence and reprint request to Ming-Fen Ho, Ph.D. Mayo Clinic 200 First Street S.W., Rochester, MN 55905, USA.
| | | |
Collapse
|
21
|
Ho MF, Ingle JN, Bongartz T, Kalari KR, Goss PE, Shepherd LE, Mushiroda T, Kubo M, Wang L, Weinshilboum RM. TCL1A Single-Nucleotide Polymorphisms and Estrogen-Mediated Toll-Like Receptor-MYD88-Dependent Nuclear Factor- κB Activation: Single-Nucleotide Polymorphism- and Selective Estrogen Receptor Modulator-Dependent Modification of Inflammation and Immune Response. Mol Pharmacol 2017; 92:175-184. [PMID: 28615284 DOI: 10.1124/mol.117.108340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/30/2017] [Indexed: 12/15/2022] Open
Abstract
In a previous genome-wide association study (GWAS) for musculoskeletal adverse events during aromatase inhibitor therapy for breast cancer, we reported that single nucleotide polymorphisms (SNPs) near the TCL1A gene were associated with this adverse drug reaction. Functional genomic studies showed that TCL1A expression was induced by estradiol, but only in cells with the variant sequence for the top GWAS SNP (rs11849538), a SNP that created a functional estrogen response element. In addition, TCL1A genotype influenced the downstream expression of a series of cytokines and chemokines and had a striking effect on nuclear factor κB (NF-κB) transcriptional activity. Furthermore, this SNP-dependent regulation could be reversed by selective ER modulators (SERMs). The present study was designed to pursue mechanisms underlying TCL1A SNP-mediated, estrogen-dependent NF-κB activation. Functional genomic studies were performed using a panel of 300 lymphoblastoid cell lines for which we had generated genome-wide SNP and gene expression data. It is known that toll-like receptors (TLRs) can regulate NF-κB signaling by a process that requires the adaptor protein MYD88. We found that TLR2, TLR7, TLR9, and TLR10 expression, as well as that of MYD88, could be modulated by TCL1A in a SNP and estrogen-dependent fashion and that these effects were reversed in the presence of SERMs. Furthermore, MYD88 inhibition blocked the TCL1A SNP and estrogen-dependent NF-κB activation, as well as protein-protein interaction between TCL1A and MYD88. These observations greatly expand the range of pathways influenced by TCL1A genotype and raise the possibility that this estrogen- and SNP-dependent regulation might be altered pharmacologically by SERMs.
Collapse
Affiliation(s)
- Ming-Fen Ho
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.-F.H., L.W., R.M.W.), Division of Medical Oncology, Department of Oncology (J.N.I.), and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota; Department of Emergency Medicine, Vanderbilt Medical Center, Nashville, Tennessee (T.B.); Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); NCIC Clinical Trials Group, Kingston, Ontario Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Tsurumi-ku, Yokohama, Japan (T.M., M.K.)
| | - James N Ingle
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.-F.H., L.W., R.M.W.), Division of Medical Oncology, Department of Oncology (J.N.I.), and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota; Department of Emergency Medicine, Vanderbilt Medical Center, Nashville, Tennessee (T.B.); Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); NCIC Clinical Trials Group, Kingston, Ontario Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Tsurumi-ku, Yokohama, Japan (T.M., M.K.)
| | - Tim Bongartz
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.-F.H., L.W., R.M.W.), Division of Medical Oncology, Department of Oncology (J.N.I.), and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota; Department of Emergency Medicine, Vanderbilt Medical Center, Nashville, Tennessee (T.B.); Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); NCIC Clinical Trials Group, Kingston, Ontario Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Tsurumi-ku, Yokohama, Japan (T.M., M.K.)
| | - Krishna R Kalari
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.-F.H., L.W., R.M.W.), Division of Medical Oncology, Department of Oncology (J.N.I.), and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota; Department of Emergency Medicine, Vanderbilt Medical Center, Nashville, Tennessee (T.B.); Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); NCIC Clinical Trials Group, Kingston, Ontario Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Tsurumi-ku, Yokohama, Japan (T.M., M.K.)
| | - Paul E Goss
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.-F.H., L.W., R.M.W.), Division of Medical Oncology, Department of Oncology (J.N.I.), and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota; Department of Emergency Medicine, Vanderbilt Medical Center, Nashville, Tennessee (T.B.); Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); NCIC Clinical Trials Group, Kingston, Ontario Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Tsurumi-ku, Yokohama, Japan (T.M., M.K.)
| | - Lois E Shepherd
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.-F.H., L.W., R.M.W.), Division of Medical Oncology, Department of Oncology (J.N.I.), and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota; Department of Emergency Medicine, Vanderbilt Medical Center, Nashville, Tennessee (T.B.); Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); NCIC Clinical Trials Group, Kingston, Ontario Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Tsurumi-ku, Yokohama, Japan (T.M., M.K.)
| | - Taisei Mushiroda
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.-F.H., L.W., R.M.W.), Division of Medical Oncology, Department of Oncology (J.N.I.), and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota; Department of Emergency Medicine, Vanderbilt Medical Center, Nashville, Tennessee (T.B.); Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); NCIC Clinical Trials Group, Kingston, Ontario Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Tsurumi-ku, Yokohama, Japan (T.M., M.K.)
| | - Michiaki Kubo
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.-F.H., L.W., R.M.W.), Division of Medical Oncology, Department of Oncology (J.N.I.), and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota; Department of Emergency Medicine, Vanderbilt Medical Center, Nashville, Tennessee (T.B.); Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); NCIC Clinical Trials Group, Kingston, Ontario Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Tsurumi-ku, Yokohama, Japan (T.M., M.K.)
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.-F.H., L.W., R.M.W.), Division of Medical Oncology, Department of Oncology (J.N.I.), and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota; Department of Emergency Medicine, Vanderbilt Medical Center, Nashville, Tennessee (T.B.); Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); NCIC Clinical Trials Group, Kingston, Ontario Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Tsurumi-ku, Yokohama, Japan (T.M., M.K.)
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (M.-F.H., L.W., R.M.W.), Division of Medical Oncology, Department of Oncology (J.N.I.), and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota; Department of Emergency Medicine, Vanderbilt Medical Center, Nashville, Tennessee (T.B.); Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts (P.E.G.); NCIC Clinical Trials Group, Kingston, Ontario Canada (L.E.S.); and RIKEN Center for Integrative Medical Science, Tsurumi-ku, Yokohama, Japan (T.M., M.K.)
| |
Collapse
|
22
|
Hertz DL, Henry NL, Rae JM. Germline genetic predictors of aromatase inhibitor concentrations, estrogen suppression and drug efficacy and toxicity in breast cancer patients. Pharmacogenomics 2017; 18:481-499. [PMID: 28346074 PMCID: PMC6219438 DOI: 10.2217/pgs-2016-0205] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 02/07/2023] Open
Abstract
The third-generation aromatase inhibitors (AIs), anastrozole, letrozole and exemestane, are highly effective for the treatment of estrogen receptor-positive breast cancer in postmenopausal women. AIs inhibit the aromatase (CYP19A1)-mediated production of estrogens. Most patients taking AIs achieve undetectable blood estrogen concentrations resulting in drug efficacy with tolerable side effects. However, some patients have suboptimal outcomes, which may be due, in part, to inherited germline genetic variants. This review summarizes published germline genetic associations with AI treatment outcomes including systemic AI concentrations, estrogenic response to AIs, AI treatment efficacy and AI treatment toxicities. Significant associations are highlighted with commentary about prioritization for future validation to identify pharmacogenetic predictors of AI treatment outcomes that can be used to inform personalized treatment decisions in patients with estrogen receptor-positive breast cancer.
Collapse
Affiliation(s)
- Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109-1065, USA
| | - N Lynn Henry
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84103, USA
| | - James M Rae
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109-1065, USA
| |
Collapse
|
23
|
Ingle JN, Xie F, Ellis MJ, Goss PE, Shepherd LE, Chapman JAW, Chen BE, Kubo M, Furukawa Y, Momozawa Y, Stearns V, Pritchard KI, Barman P, Carlson EE, Goetz MP, Weinshilboum RM, Kalari KR, Wang L. Genetic Polymorphisms in the Long Noncoding RNA MIR2052HG Offer a Pharmacogenomic Basis for the Response of Breast Cancer Patients to Aromatase Inhibitor Therapy. Cancer Res 2016; 76:7012-7023. [PMID: 27758888 DOI: 10.1158/0008-5472.can-16-1371] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 09/12/2016] [Accepted: 09/28/2016] [Indexed: 02/07/2023]
Abstract
Genetic risks in breast cancer remain only partly understood. Here, we report the results of a genome-wide association study of germline DNA from 4,658 women, including 252 women experiencing a breast cancer recurrence, who were entered on the MA.27 adjuvant trial comparing the aromatase inhibitors (AI) anastrozole and exemestane. Single-nucleotide polymorphisms (SNP) of top significance were identified in the gene encoding MIR2052HG, a long noncoding RNA of unknown function. Heterozygous or homozygous individuals for variant alleles exhibited a ∼40% or ∼63% decrease, respectively, in the hazard of breast cancer recurrence relative to homozygous wild-type individuals. Functional genomic studies in lymphoblastoid cell lines and ERα-positive breast cancer cell lines showed that expression from MIR2052HG and the ESR1 gene encoding estrogen receptor-α (ERα) was induced by estrogen and AI in a SNP-dependent manner. Variant SNP genotypes exhibited increased ERα binding to estrogen response elements, relative to wild-type genotypes, a pattern that was reversed by AI treatment. Further, variant SNPs were associated with lower expression of MIR2052HG and ERα. RNAi-mediated silencing of MIR2052HG in breast cancer cell lines decreased ERα expression, cell proliferation, and anchorage-independent colony formation. Mechanistic investigations revealed that MIR2052HG sustained ERα levels both by promoting AKT/FOXO3-mediated ESR1 transcription and by limiting ubiquitin-mediated, proteasome-dependent degradation of ERα. Taken together, our results define MIR2052HS as a functionally polymorphic gene that affects risks of breast cancer recurrence in women treated with AI. More broadly, our results offer a pharmacogenomic basis to understand differences in the response of breast cancer patients to AI therapy. Cancer Res; 76(23); 7012-23. ©2016 AACR.
Collapse
Affiliation(s)
- James N Ingle
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Fang Xie
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | | | - Paul E Goss
- Massachusetts General Hospital Cancer Center, Harvard University, Boston, Massachusetts
| | | | | | | | - Michiaki Kubo
- RIKEN Center for Integrative Medical Science, Yokohama, Japan
| | | | | | - Vered Stearns
- Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Kathleen I Pritchard
- Sunnybrook Odette Regional Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Poulami Barman
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Erin E Carlson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Matthew P Goetz
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Krishna R Kalari
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
24
|
Identification of Claudin 1 Transcript Variants in Human Invasive Breast Cancer. PLoS One 2016; 11:e0163387. [PMID: 27649506 PMCID: PMC5029943 DOI: 10.1371/journal.pone.0163387] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/06/2016] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The claudin 1 tight junction protein, solely responsible for the barrier function of epithelial cells, is frequently down regulated in invasive human breast cancer. The underlying mechanism is largely unknown, and no obvious mutations in the claudin 1 gene (CLDN1) have been identified to date in breast cancer. Since many genes have been shown to undergo deregulation through splicing and mis-splicing events in cancer, the current study was undertaken to investigate the occurrence of transcript variants for CLDN1 in human invasive breast cancer. METHODS RT-PCR analysis of CLDN1 transcripts was conducted on RNA isolated from 12 human invasive breast tumors. The PCR products from each tumor were resolved by agarose gel electrophoresis, cloned and sequenced. Genomic DNA was also isolated from each of the 12 tumors and amplified using PCR CLDN1 specific primers. Sanger sequencing and single nucleotide polymorphism (SNP) analyses were conducted. RESULTS A number of CLDN1 transcript variants were identified in these breast tumors. All variants were shorter than the classical CLDN1 transcript. Sequence analysis of the PCR products revealed several splice variants, primarily in exon 1 of CLDN1; resulting in truncated proteins. One variant, V1, resulted in a premature stop codon and thus likely led to nonsense mediated decay. Interestingly, another transcript variant, V2, was not detected in normal breast tissue samples. Further, sequence analysis of the tumor genomic DNA revealed SNPs in 3 of the 4 coding exons, including a rare missense SNP (rs140846629) in exon 2 which represents an Ala124Thr substitution. To our knowledge this is the first report of CLDN1 transcript variants in human invasive breast cancer. These studies suggest that alternate splicing may also be a mechanism by which claudin 1 is down regulated at both the mRNA and protein levels in invasive breast cancer and may provide novel insights into how CLDN1 is reduced or silenced in human breast cancer.
Collapse
|
25
|
Yee SW, Momozawa Y, Kamatani Y, Tyndale RF, Weinshilboum RM, Ratain MJ, Giacomini KM, Kubo M. Genomewide Association Studies in Pharmacogenomics: Meeting Report of the NIH Pharmacogenomics Research Network-RIKEN (PGRN-RIKEN) Collaboration. Clin Pharmacol Ther 2016; 100:423-426. [PMID: 27256705 DOI: 10.1002/cpt.405] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 02/06/2023]
Abstract
Genomewide association studies (GWAS) have resulted in the identification of many heritable genetic factors that underlie risk for human disease or variation in physiologic traits. In contrast, there are fewer GWAS of drug response phenotypes, despite extensive unexplained interindividual variability. To address this urgent need, the NIH Pharmacogenomics Research Network (PGRN) and the Center for Integrative Medical Sciences (IMS) at RIKEN support a collaboration, PGRN-RIKEN, with the goal of accelerating GWAS of drug response phenotypes.
Collapse
Affiliation(s)
- S W Yee
- University of California San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, California, USA
| | - Y Momozawa
- University of California San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, California, USA
| | - Y Kamatani
- University of California San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, California, USA
| | - R F Tyndale
- University of California San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, California, USA
| | - R M Weinshilboum
- University of California San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, California, USA
| | - M J Ratain
- University of California San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, California, USA
| | - K M Giacomini
- University of California San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, California, USA.
| | - M Kubo
- University of California San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, California, USA.
| |
Collapse
|