1
|
Wu X, Jin L, Ren D, Huang S, Meng X, Wu Z, Lv C, Ru J, Zhang H, Zhang S, Bao J, Wang O, Xia E. α-Hederin causes ferroptosis in triple-negative breast cancer through modulating IRF1 to suppress GPX4. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156611. [PMID: 40153970 DOI: 10.1016/j.phymed.2025.156611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/22/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Breast cancer ranks first in the global incidence rate of cancer among women. Triple-negative breast cancer (TNBC) is considered to be the most dangerous type because of the lack of specific therapeutic targets and rapid progression. The emergence of ferroptosis provides a new therapeutic perspective for TNBC. α-Hederin is a triterpenoid saponin derived from the traditional Chinese medicine Ivy, which has been proven to have anti-cancer effects on various cancers, but its efficacy and mechanism of inducing ferroptosis in TNBC remain to be further clarified. OBJECT To investigate the effect and mechanism of α-Hederin induced ferroptosis in TNBC. METHOD Cell viability was measured by CCK-8 assay, and cell proliferation and migration were evaluated by clone assay and scratch assay. The effect of α-Hederin on TNBC cell apoptosis was assessed by flow cytometry. Transcriptomics searches for critical pathways. Intracellular and lipid reactive oxygen species and Fe2+and Fe were detected by DCFH-DA probe, FerroOrange fluorescent probe and C11-BODIPY fluorescent probe, and the contents of malondialdehyde and reduced glutathione were detected by MDA and GSH kits. Erastin was used as a positive control for ferroptosis and Ferrrostatin-1(Fer-1) as an inhibitor. The relationship between α-Hederin and GPX4, IRF was analyzed by western blot and si-RNA, and the association was further confirmed by molecular simulation docking, external SPR experiments, and luciferase experiments. Constructing xenograft mouse models and human derived organoid models to evaluate the anti-TNBC efficacy of α-Hederin, and verifying the efficacy and ferroptosis mechanism of the drug in vivo through HE staining and IHC. RESULT α-Hederin significantly inhibited the progression of TNBC. In vitro, α-Hederin decreased cancer cell viability through ferroptosis, increased glutathione degradation and MDA production, and promoted intracellular Fe2+ and ROS production, whereas Fer-1, an ferroptosis inhibitor, reversed this effect. Mechanistically, molecular docking and SPR experiments showed binding of α-Hederin to the key regulator IRF1, and knockdown/overexpression of IRF1 significantly affected the expression of GPX4, a downstream target of the ferroptosis pathway. In vivo, α-Hederin prevented tumor growth in xenograft and organoid models via the IRF1/GPX4 axis. CONCLUSION We proved for the first time in this research that α-Hederin exerts anti-TNBC effects through a novel IRF1/GPX4 ferroptosis pathway.
Collapse
Affiliation(s)
- Xue Wu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Lingli Jin
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Zhejiang Provincial People's Hospital, Zhejiang 325000, China
| | - Disuo Ren
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shaolong Huang
- Graduate School of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xinyu Meng
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zhixuan Wu
- Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chaoyue Lv
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jiatong Ru
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Heyu Zhang
- Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shuwei Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jingxia Bao
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Graduate school of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Ouchen Wang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Erjie Xia
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
2
|
Reed VL, Lalu E, Yoon L, Fultang N, Peethambaran B. Uncovering a Novel Role of ROR1 in the Epigenetic Regulation of Tumor Suppressor Gene CREB3L1 in Triple-Negative Breast Cancer Cells. Biomolecules 2025; 15:734. [PMID: 40427627 PMCID: PMC12109183 DOI: 10.3390/biom15050734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/25/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
A characteristic of triple-negative breast cancer (TNBC) is the epigenetic regulation of tumor suppressor genes, leading to TNBC heterogeneity and treatment resistance in patients. TNBC exhibits high methylation rates, leading to the silencing of numerous tumor suppressor genes. DNA methyltransferase inhibitors (DNMTis) have shown limited clinical efficacy in TNBC treatment. This study aims to uncover a target that could be used to reverse the epigenetic silencing of tumor suppressor genes in TNBC. The Western blot analysis demonstrated that ROR1 knockdown, an oncofetal gene, reduced DNMT3A and DNMT3B protein expression in the TNBC cell lines MDA-MB-231 and HCC1806, as well as a non-malignant breast cell line, MCF10A. The reduced representation bisulfite sequencing (RRBS) analysis identified differential methylation of CREB3L1 when ROR1 is knocked down in TNBC cell lines. CREB3L1 is a transcription factor that plays tumor-suppressive roles in TNBC and is commonly epigenetically silenced in patients. This study shows that ROR1 requires pSTAT3 activation to upregulate DNMT3A and DNMT3B expression to induce CREB3L1 epigenetic silencing in TNBC. ROR1 knockdown resulted in the re-expression of CREB3L1 in TNBC cells. The data provide evidence that ROR1 inhibition, in combination with DNMTis, could enhance patient outcomes as a therapeutic approach for TNBC.
Collapse
Affiliation(s)
- Victoria L. Reed
- Department of Biology, St. Joseph’s University, Philadelphia, PA 19131, USA; (V.L.R.); (E.L.); (L.Y.)
| | - Eric Lalu
- Department of Biology, St. Joseph’s University, Philadelphia, PA 19131, USA; (V.L.R.); (E.L.); (L.Y.)
| | - Leena Yoon
- Department of Biology, St. Joseph’s University, Philadelphia, PA 19131, USA; (V.L.R.); (E.L.); (L.Y.)
- Cancer Biology Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Norman Fultang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Bela Peethambaran
- Department of Biology, St. Joseph’s University, Philadelphia, PA 19131, USA; (V.L.R.); (E.L.); (L.Y.)
| |
Collapse
|
3
|
Kakimi K, Sugie T. Why combine and why neoadjuvant? Tumor immunological perspectives on chemoimmunotherapy in triple-negative breast cancer. Breast Cancer 2025:10.1007/s12282-025-01707-5. [PMID: 40327275 DOI: 10.1007/s12282-025-01707-5] [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: 02/24/2025] [Accepted: 04/18/2025] [Indexed: 05/07/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by limited targeted therapies and high recurrence rates. While immune checkpoint inhibitors (ICIs) have shown promise, their efficacy as monotherapy is limited. Clinically, ICIs demonstrate significant benefit primarily when combined with chemotherapy, particularly in the neoadjuvant setting for early-stage TNBC, which yields superior outcomes compared to adjuvant therapy. This review elucidates the tumor immunological principles underlying these observations. We discussed how the suppressive tumor microenvironment (TME), progressive T cell exhaustion, and associated epigenetic scarring constrain ICI monotherapy effectiveness. Crucially, we highlight the immunological advantages of the neoadjuvant approach: the presence of the primary tumor provides abundant antigens, and intact tumor-draining lymph nodes (TDLNs) act as critical sites for ICI-mediated priming and expansion of naïve and precursor exhausted T cells. This robust activation within TDLNs enhances systemic anti-tumor immunity and expands the T cell repertoire, a process less effectively achieved in the adjuvant setting after tumor resection. These mechanisms provide a strong rationale for the improved pathological complete response (pCR) rates and event-free survival observed with neoadjuvant chemoimmunotherapy, as demonstrated in trials like KEYNOTE-522. We further explore the implications for adjuvant therapy decisions based on treatment response, the challenges of ICI resistance, the need for predictive biomarkers, management of immune-related adverse events (irAEs), and future therapeutic directions. Understanding the dynamic interplay between chemotherapy, ICIs, T cells, and the TME, particularly the role of TDLNs in the neoadjuvant context, is essential for optimizing immunotherapy strategies and improving outcomes for patients with TNBC.
Collapse
Affiliation(s)
- Kazuhiro Kakimi
- Department of Immunology, Kindai University Faculty of Medicine, 377-2 Onohigashi, Osakasayama, Osaka, 589-8511, Japan.
- Chemotherapy Center, Kansai Medical University Kori Hospital, 8-45 Korihondori, Neyagawa, Osaka, 572-8551, Japan.
| | - Tomoharu Sugie
- Chemotherapy Center, Kansai Medical University Kori Hospital, 8-45 Korihondori, Neyagawa, Osaka, 572-8551, Japan.
| |
Collapse
|
4
|
Zhu H, Ouyang L, Huang Y, Tan J, Liu C, Wang Q, Huang R, Wong WT, Liu X, Pan H, Liao Y. Dual-Gating Strategy: Ultrasound Activation of TRPV2 Channels and Borate-Glass-Induced Calcium Overload for Tumor Suppression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414676. [PMID: 40013983 PMCID: PMC12021067 DOI: 10.1002/advs.202414676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/05/2025] [Indexed: 02/28/2025]
Abstract
Effective and precise treatment of breast cancer, particularly with bone metastasis, remains a significant challenge. Here, a dual-gating strategy combining locally delivered borate glass (BG) and ultrasound (US) is developed for the precise and effective inhibition of breast cancer by targeting transient receptor potential vanilloid 2 (TRPV2). The results demonstrate that after local delivery of BG to the solid tumor, US effectively triggers calcium overload by activating the overexpressed TRPV2 channels, leading to mitochondrial autophagy and apoptosis in breast cancer cells, thereby inhibiting tumor growth with high precision. These effects are validated in subcutaneous, orthotopic, and TRPV2-overexpressing breast cancer mouse models. In the bone metastasis model, BG combined with US treatment simultaneously suppresses tumor growth and promotes bone regeneration. Overall, this dual-gating strategy offers a safe and efficient approach for the precise treatment of cancers with high TRPV2 expression and provides new insights into the design and clinical translation of calcium-overload-based cancer therapies.
Collapse
Affiliation(s)
- Haihong Zhu
- Department of Pharmacy, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
- Department of Pharmacy, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghai200336China
| | - Liping Ouyang
- Department of Pharmacy, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghai200336China
- Hongqiao International Institute of MedicineShanghai Jiao Tong University School of MedicineShanghai200336China
| | - Yangguang Huang
- Department of Pharmacy, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghai200336China
| | - Ji Tan
- State Key Laboratory of Advanced CeramicsShanghai Institute of CeramicsChinese Academy of SciencesShanghai200050China
| | - Chunyu Liu
- Shenzhen Key Laboratory of Marine Biomedical MaterialsCAS‐HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055China
| | - Qian Wang
- Department of Pharmacy, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghai200336China
| | - Rongkun Huang
- Department of Pharmacy, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghai200336China
| | - Wing Tak Wong
- School of Life SciencesFaculty of ScienceThe Chinese University of Hong KongHong Kong999077China
| | - Xuanyong Liu
- State Key Laboratory of Advanced CeramicsShanghai Institute of CeramicsChinese Academy of SciencesShanghai200050China
- School of Chemistry and Materials ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesSub‐Lane XiangshanHangzhou310024China
| | - Haobo Pan
- Shenzhen Key Laboratory of Marine Biomedical MaterialsCAS‐HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055China
| | - Yun Liao
- Department of Pharmacy, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
- Department of Pharmacy, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghai200336China
| |
Collapse
|
5
|
Wan H, Ling Z, Xie Y, Jiang H, Ruan Z, Yang D, Yang X, Pei J. Single-cell and transcriptome analyses revealed CTHRC1 a potential therapeutic target mediating invasion and tumor microenvironment in TNBC: experimental validation. Front Immunol 2025; 16:1534981. [PMID: 40134434 PMCID: PMC11933001 DOI: 10.3389/fimmu.2025.1534981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Background Investigating the pivotal role of CTHRC1 in the tumor microenvironment of triple-negative breast cancer (TNBC). Method The RNA transcriptomic data obtained from the Cancer Genome Atlas and single-cell sequencing data from TNBC in Gene Expression Omnibus (GEO) were acquired and subjected to analysis. A comprehensive investigation was conducted with a specific focus on characterizing CTHRC1 in TNBC and its correlation with invasive genes. Furthermore, additional analyses were performed to explore the relationship between CTHRC1, tumor immune cell infiltration, and immunotherapy in TNBC. The expression of CTHRC1 in the tumor microenvironment, cellular differentiation, and cellular communication was systematically analyzed using single-cell data from TNBC. Result The expression of CTHRC1 in patients with TNBC gradually increases concomitantly with the progression of tumor T-stage and N-stage. Simultaneously, there is a concurrent increase in the expression of most invasive gene sets. Furthermore, there is a significant augmentation in both infiltration abundance and activity of M2-type macrophages associated with elevated levels of CTHRC1 expression. Single-cell data reveal an upregulated expression of the invasive gene set in CTHRC1-positive cancer associated fibroblasts (CAFs), thereby modulating their interaction with M2-type macrophages. Multiple immunofluorescence analyses confirmed that CTHRC1 modulates immune cell infiltration and tumor cell invasion through the mediation of CAFs. Conclusion CTHRC1 was a molecule that exhibits characteristic expression in TNBC. CTHRC1 positive CAFs exert regulatory effects within the immunosuppressive microenvironment of TNBC by modulating M2-type macrophages.
Collapse
Affiliation(s)
- Hong Wan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zichen Ling
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuwei Xie
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Han Jiang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhifan Ruan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dashuai Yang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaowei Yang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Pei
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
6
|
Wang Y, Haase S, Whitman A, Beltran A, Spanheimer PM, Brunk E. A Multimodal Framework to Uncover Drug-Responsive Subpopulations in Triple-Negative Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638274. [PMID: 40027670 PMCID: PMC11870422 DOI: 10.1101/2025.02.14.638274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Understanding how individual cancer cells adapt to drug treatment is a fundamental challenge limiting precision medicine cancer therapy strategies. While single-cell technologies have advanced our understanding of cellular heterogeneity, efforts to connect the behavior of individual cells to broader tumor drug responses and uncover global trends across diverse systems remain limited. There is a growing availability of single-cell and bulk omics data, but a lack of centralized tools and repositories makes it difficult to study drug response globally, especially at the level of single-cell adaptation. To address this, we present a multimodal framework that integrates bulk and single-cell treated and untreated transcriptomics data to identify drug responsive cell populations in triple-negative breast cancer (TNBC). Our framework leverages population-scale bulk transcriptomics data from TNBC samples to define seven main "identities", each representing unique combinations of biologically relevant genes. These identities are dynamic and trackable, allowing us to map them onto single cells and uncover global patterns of how cell populations respond to drug treatment. Unlike static classifications, this approach captures the evolving nature of cellular states, revealing that a select few identities dominate and drive population-level responses during treatment. Crucially, our ability to decode these trends through the inherent noise of single-cell data provides a clearer picture of how heterogeneous cell populations adapt to therapy. By identifying the dominant identities and their dynamics, we can better predict how entire tumors respond to treatment. This insight is essential for designing precise combination therapies tailored to the unique heterogeneity of patient tumors, addressing the single-cell variations that ultimately determine therapeutic outcomes.
Collapse
|
7
|
Xu L, Xu P, Wang J, Ji H, Zhang L, Tang Z. Advancements in clinical research and emerging therapies for triple-negative breast cancer treatment. Eur J Pharmacol 2025; 988:177202. [PMID: 39675457 DOI: 10.1016/j.ejphar.2024.177202] [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: 08/02/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
Triple-negative breast cancer (TNBC), defined by the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor 2 (HER2) expression, is acknowledged as the most aggressive form of breast cancer (BC), comprising 15%-20% of all primary cases. Despite the prevalence of TNBC, effective and well-tolerated targeted therapies remain limited, with chemotherapy continuing to be the mainstay of treatment. However, the horizon is brightened by recent advancements in immunotherapy and antibody-drug conjugates (ADCs), which have garnered the U.S. Food and Drug Administration (FDA) approval for various stages of TNBC. Poly (ADP-ribose) polymerase inhibitors (PARPi), particularly for TNBC with BRCA mutations, present a promising avenue, albeit with the challenge of resistance that must be addressed. The success of phosphoinositide-3 kinase (PI3K) pathway inhibitors in hormone receptor (HR)-positive BC suggests potential applicability in TNBC, spurring optimism within the research community. This review endeavors to offer a comprehensive synthesis of both established and cutting-edge targeted therapies for TNBC. We delve into the specifics of PARPi, androgen receptor (AR) inhibitors, Cancer stem cells (CSCs), PI3K/Protein Kinase B (AKT)/mammalian target of rapamycin (mTOR), the transforming growth factor-beta (TGF-β), Ntoch, Wnt/β-catenin, hedgehog (Hh) pathway inhibitors, Epigenetic target-mediated drug delivery, ADCs, immune checkpoint inhibitors (ICIs)and novel immunotherapeutic solutions, contextualizing TNBC within current treatment paradigms. By elucidating the mechanisms of these drugs and their prospective clinical applications, we aim to shed light on the challenges and underscore the beacon of hope that translational research and innovative therapies represent for the oncology field.
Collapse
Affiliation(s)
- Lili Xu
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Pengtao Xu
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Jingsong Wang
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, 628000, China
| | - Hui Ji
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Lin Zhang
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China.
| |
Collapse
|
8
|
Alalhareth IS, Alyami SM, Alshareef AH, Ajeibi AO, Al Munjem MF, Elfifi AA, Alsharif MM, Alzahrani SA, Alqaad MA, Bakir MB, Abdel-Wahab BA. Cellular Epigenetic Targets and Epidrugs in Breast Cancer Therapy: Mechanisms, Challenges, and Future Perspectives. Pharmaceuticals (Basel) 2025; 18:207. [PMID: 40006021 PMCID: PMC11858621 DOI: 10.3390/ph18020207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/31/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Breast cancer is the most common malignancy affecting women, manifesting as a heterogeneous disease with diverse molecular characteristics and clinical presentations. Recent studies have elucidated the role of epigenetic modifications in the pathogenesis of breast cancer, including drug resistance and efflux characteristics, offering potential new diagnostic and prognostic markers, treatment efficacy predictors, and therapeutic agents. Key modifications include DNA cytosine methylation and the covalent modification of histone proteins. Unlike genetic mutations, reprogramming the epigenetic landscape of the cancer epigenome is a promising targeted therapy for the treatment and reversal of drug resistance. Epidrugs, which target DNA methylation and histone modifications, can provide novel options for the treatment of breast cancer by reversing the acquired resistance to treatment. Currently, the most promising approach involves combination therapies consisting of epidrugs with immune checkpoint inhibitors. This review examines the aberrant epigenetic regulation of breast cancer initiation and progression, focusing on modifications related to estrogen signaling, drug resistance, cancer progression, and the epithelial-mesenchymal transition (EMT). It examines existing epigenetic drugs for treating breast cancer, including agents that modify DNA, inhibitors of histone acetyltransferases, histone deacetylases, histone methyltransferases, and histone demethyltransferases. It also delves into ongoing studies on combining epidrugs with other therapies and addresses the upcoming obstacles in this field.
Collapse
Affiliation(s)
- Ibrahim S. Alalhareth
- College of Pharmacy, Najran University, Najran 66256, Saudi Arabia; (I.S.A.); (S.M.A.)
| | - Saleh M. Alyami
- College of Pharmacy, Najran University, Najran 66256, Saudi Arabia; (I.S.A.); (S.M.A.)
| | - Ali H. Alshareef
- Department of Pharmaceuticals Care, Ministry of Defense, Najran 66281, Saudi Arabia; (A.H.A.); (A.O.A.); (A.A.E.); (M.M.A.)
| | - Ahmed O. Ajeibi
- Department of Pharmaceuticals Care, Ministry of Defense, Najran 66281, Saudi Arabia; (A.H.A.); (A.O.A.); (A.A.E.); (M.M.A.)
| | - Manea F. Al Munjem
- King Khaled Hospital -Najran Health Cluster, Najran 66261, Saudi Arabia;
| | - Ahmad A. Elfifi
- Department of Pharmaceuticals Care, Ministry of Defense, Najran 66281, Saudi Arabia; (A.H.A.); (A.O.A.); (A.A.E.); (M.M.A.)
| | - Meshal M. Alsharif
- Department of Pharmaceuticals Care, Ministry of Defense, Najran 66281, Saudi Arabia; (A.H.A.); (A.O.A.); (A.A.E.); (M.M.A.)
| | - Seham A. Alzahrani
- Pharmacy Department, Khamis Mushait General Hospital, King Khalid Rd, Al Shifa, Khamis Mushait 62433, Saudi Arabia;
| | - Mohammed A. Alqaad
- Department of Pharmaceutical Care Services, Al Noor Specialized Hospital, Makkah Health, Cluster, Makkah 24241, Saudi Arabia;
| | - Marwa B. Bakir
- Department of Medical Education, College of Medicine, Najran University, Najran 1988, Saudi Arabia;
| | - Basel A. Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran 1988, Saudi Arabia
| |
Collapse
|
9
|
Djamgoz MBA. Stemness of Cancer: A Study of Triple-negative Breast Cancer From a Neuroscience Perspective. Stem Cell Rev Rep 2025; 21:337-350. [PMID: 39531198 PMCID: PMC11872763 DOI: 10.1007/s12015-024-10809-0] [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] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Stemness, giving cancer cells massive plasticity enabling them to survive in dynamic (e.g. hypoxic) environments and become resistant to treatment, especially chemotherapy, is an important property of aggressive tumours. Here, we review some essentials of cancer stemness focusing on triple-negative breast cancer (TNBC), the most aggressive form of all breast cancers. TNBC cells express a range of genes and mechanisms associated with stemness, including the fundamental four "Yamanaka factors". Most of the evidence concerns the transcription factor / oncogene c-Myc and an interesting case is the expression of the neonatal splice variant of voltage-gated sodium channel subtype Nav1.5. On the whole, measures that reduce the stemness make cancer cells less aggressive, reducing their invasive/metastatic potential and increasing/restoring their chemosensitivity. Such measures include gene silencing techniques, epigenetic therapies as well as novel approaches like optogenetics aiming to modulate the plasma membrane voltage. Indeed, simply hyperpolarizing their membrane potential can make stem cells differentiate. Finally, we give an overview of the clinical aspects and exploitation of cancer/TNBC stemness, including diagnostics and therapeutics. In particular, personalised mRNA-based therapies and mechanistically meaningful combinations are promising and the emerging discipline of 'cancer neuroscience' is providing novel insights to both fundamental issues and clinical applications.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
10
|
Kung CP, Terzich ND, Ilagen MXG, Prinsen MJ, Kaushal M, Kladney RD, Weber JH, Mabry AR, Torres LS, Bramel ER, Freeman EC, Sabloak T, Cottrell KA, Ryu S, Weber WM, Maggi L, Shriver LP, Patti GJ, Weber JD. ADAR1 Regulates Lipid Remodeling through MDM2 to Dictate Ferroptosis Sensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633410. [PMID: 39896528 PMCID: PMC11785053 DOI: 10.1101/2025.01.16.633410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Triple-negative breast cancer (TNBC), lacking expression of estrogen, progesterone, and HER2 receptors, is aggressive and lacks targeted treatment options. An RNA editing enzyme, adenosine deaminase acting on RNA 1 (ADAR1), has been shown to play important roles in TNBC tumorigenesis. We posit that ADAR1 functions as a homeostatic factor protecting TNBC from internal and external pressure, including metabolic stress. We tested the hypothesis that the iron- dependent cell death pathway, ferroptosis, is a ADAR1-protected metabolic vulnerability in TNBC by showing that ADAR1 knockdown sensitizes TNBC cells to GPX4 inhibitors. By performing single-reaction monitoring-based liquid chromatography coupled to mass spectrometry (LC-MS) to measure intracellular lipid contents, we showed that ADAR1 loss increased the abundance of polyunsaturated fatty acid phospholipids (PUFA-PL), of which peroxidation is the primary driver of ferroptosis. Transcriptomic analyses led to the discovery of the proto-oncogene MDM2 contributing to the lipid remodeling in TNBC upon ADAR1 loss. A phenotypic drug screen using a ferroptosis-focused library was performed to identify FDA- approved cobimetinib as a drug-repurposing candidate to synergize with ADAR1 loss to suppress TNBC tumorigenesis. By demonstrating that ADAR1 regulates the metabolic fitness of TNBC through desensitizing ferroptosis, we aim to leverage this metabolic vulnerability to inform basic, pre-clinical, and clinical studies to develop novel therapeutic strategies for TNBC.
Collapse
|
11
|
Shan Y, He D, Yan F, Huang W. Understanding the tumor microenvironment for personalized immunotherapy in early-onset head and neck squamous cell carcinoma. Front Immunol 2025; 15:1522820. [PMID: 39830511 PMCID: PMC11739722 DOI: 10.3389/fimmu.2024.1522820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Early-onset head and neck squamous cell carcinoma (HNSCC) has been increasingly observed in recent years, exhibiting distinct tumor behavior and a unique tumor microenvironment (TME) compared to older age groups. Studies suggest that early-onset HNSCC is associated with specific risk factors and prognostic outcomes, while the underlying mechanisms driving these age-related differences remain unclear. In this review, we systematically examined original studies involving young HNSCC patient samples, focusing on the characteristics of the TME and potential for personalized immunotherapy. While further evidence is needed, our findings indicate that the TME in early-onset HNSCC often exhibits higher aggressiveness and immune suppression. Consequently, tailored immunotherapy may offer a promising therapeutic strategy for this distinct patient population.
Collapse
Affiliation(s)
| | | | | | - Weijia Huang
- Department of Oral & Maxillofacial Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
12
|
Guo Z, Zhu Z, Lin X, Wang S, Wen Y, Wang L, Zhi L, Zhou J. Tumor microenvironment and immunotherapy for triple-negative breast cancer. Biomark Res 2024; 12:166. [PMID: 39741315 DOI: 10.1186/s40364-024-00714-6] [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: 10/16/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer known for its high aggressiveness and poor prognosis. Conventional treatment of TNBC is challenging due to its heterogeneity and lack of clear targets. Recent advancements in immunotherapy have shown promise in treating TNBC, with immune checkpoint therapy playing a significant role in comprehensive treatment plans. The tumor microenvironment (TME), comprising immune cells, stromal cells, and various cytokines, plays a crucial role in TNBC progression and response to immunotherapy. The high presence of tumor-infiltrating lymphocytes and immune checkpoint proteins in TNBC indicates the potential of immunotherapeutic strategies. However, the complexity of the TME, while offering therapeutic targets, requires further exploration of its multiple roles in immunotherapy. In this review, we discuss the interaction mechanism between TME and TNBC immunotherapy based on the characteristics and composition of TME, and elaborate on and analyze the effect of TME on immunotherapy, the potential of TME as an immune target, and the ability of TME as a biomarker. Understanding these dynamics will offer new insights for enhancing therapeutic approaches and investigating stratification and prognostic markers for TNBC patients.
Collapse
Affiliation(s)
- Zijie Guo
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Ziyu Zhu
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Xixi Lin
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Shenkangle Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Yihong Wen
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Linbo Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Lili Zhi
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Jichun Zhou
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
13
|
Neagu AN, Josan CL, Jayaweera TM, Morrissiey H, Johnson KR, Darie CC. Bio-Pathological Functions of Posttranslational Modifications of Histological Biomarkers in Breast Cancer. Molecules 2024; 29:4156. [PMID: 39275004 PMCID: PMC11397409 DOI: 10.3390/molecules29174156] [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: 08/12/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Proteins are the most common types of biomarkers used in breast cancer (BC) theranostics and management. By definition, a biomarker must be a relevant, objective, stable, and quantifiable biomolecule or other parameter, but proteins are known to exhibit the most variate and profound structural and functional variation. Thus, the proteome is highly dynamic and permanently reshaped and readapted, according to changing microenvironments, to maintain the local cell and tissue homeostasis. It is known that protein posttranslational modifications (PTMs) can affect all aspects of protein function. In this review, we focused our analysis on the different types of PTMs of histological biomarkers in BC. Thus, we analyzed the most common PTMs, including phosphorylation, acetylation, methylation, ubiquitination, SUMOylation, neddylation, palmitoylation, myristoylation, and glycosylation/sialylation/fucosylation of transcription factors, proliferation marker Ki-67, plasma membrane proteins, and histone modifications. Most of these PTMs occur in the presence of cellular stress. We emphasized that these PTMs interfere with these biomarkers maintenance, turnover and lifespan, nuclear or subcellular localization, structure and function, stabilization or inactivation, initiation or silencing of genomic and non-genomic pathways, including transcriptional activities or signaling pathways, mitosis, proteostasis, cell-cell and cell-extracellular matrix (ECM) interactions, membrane trafficking, and PPIs. Moreover, PTMs of these biomarkers orchestrate all hallmark pathways that are dysregulated in BC, playing both pro- and/or antitumoral and context-specific roles in DNA damage, repair and genomic stability, inactivation/activation of tumor-suppressor genes and oncogenes, phenotypic plasticity, epigenetic regulation of gene expression and non-mutational reprogramming, proliferative signaling, endocytosis, cell death, dysregulated TME, invasion and metastasis, including epithelial-mesenchymal/mesenchymal-epithelial transition (EMT/MET), and resistance to therapy or reversal of multidrug therapy resistance. PTMs occur in the nucleus but also at the plasma membrane and cytoplasmic level and induce biomarker translocation with opposite effects. Analysis of protein PTMs allows for the discovery and validation of new biomarkers in BC, mainly for early diagnosis, like extracellular vesicle glycosylation, which may be considered as a potential source of circulating cancer biomarkers.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania;
| | - Claudiu-Laurentiu Josan
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania;
| | - Taniya M. Jayaweera
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (T.M.J.); (H.M.); (K.R.J.)
| | - Hailey Morrissiey
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (T.M.J.); (H.M.); (K.R.J.)
| | - Kaya R. Johnson
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (T.M.J.); (H.M.); (K.R.J.)
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (T.M.J.); (H.M.); (K.R.J.)
| |
Collapse
|
14
|
Huang M, Jiang Z, Xu Y, Wu C, Wei D, Meng X, Qian D. Methylation modification of non-histone proteins in breast cancer: an emerging targeted therapeutic strategy. Pharmacol Res 2024; 208:107354. [PMID: 39154671 DOI: 10.1016/j.phrs.2024.107354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Breast cancer is a major public health concern worldwide, being the most commonly diagnosed cancer among women and a leading cause of cancer-related deaths. Recent studies have highlighted the significance of non-histone methylation in breast cancer, which modulates the activity, interaction, localization, and stability of target proteins. This regulation affects critical processes such as oncogenesis, tumor growth, proliferation, invasion, migration, and immune responses. This review delves into the enzymes responsible for non-histone methylation, such as protein arginine methyltransferases (PRMTs), lysine methyltransferases (KMTs), and demethylases, and explores their roles in breast cancer. By elucidating the molecular mechanisms and functional consequences of non-histone methylation, this review aims to provide insights into novel therapeutic strategies targeting these pathways. The therapeutic potential of targeting non-histone methylation to overcome drug resistance and enhance treatment efficacy in breast cancer is also discussed, highlighting promising avenues for future research and clinical applications.
Collapse
Affiliation(s)
- Mingyao Huang
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350011, China
| | - Zirong Jiang
- Department of Thyroid and Breast Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde 352100, China
| | - Yadan Xu
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Chaoshen Wu
- Central Laboratory, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu 215500, China
| | - Ding Wei
- Department of Human Resources, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Xuli Meng
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China.
| | - Da Qian
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Central Laboratory, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu 215500, China; Department of Plastic Surgery-Hand Surgery, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu 215500, China.
| |
Collapse
|
15
|
Manna PR, Molehin D, Ahmed AU, Yang S, Reddy PH. Acetylation of Steroidogenic Acute Regulatory Protein Sensitizes 17β-Estradiol Regulation in Hormone-Sensitive Breast Cancer Cells. Int J Mol Sci 2024; 25:8732. [PMID: 39201419 PMCID: PMC11354777 DOI: 10.3390/ijms25168732] [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: 07/18/2024] [Revised: 08/08/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
An imbalance in estrogen signaling is a critical event in breast tumorigenesis. The majority of breast cancers (BCs) are hormone-sensitive; they majorly express the estrogen receptor (ER+) and are activated by 17β-estradiol (E2). The steroidogenic acute regulatory protein (StAR) mediates the rate-limiting step in steroid biosynthesis. The dysregulation of the epigenetic machinery, modulating E2 levels, is a primary occurrence for promoting breast tumorigenesis. StAR expression, concomitant with E2 synthesis, was reported to be aberrantly high in human and mouse hormone-dependent BC cells compared with their non-cancerous counterparts. However, the mechanism of action of StAR remains poorly understood. We discovered StAR as an acetylated protein and have identified a number of lysine (K) residues that are putatively acetylated in malignant and non-malignant breast cells, using LC-MS/MS (liquid chromatography-tandem mass spectrometry), suggesting they differently influence E2 synthesis in mammary tissue. The treatment of hormone-sensitive MCF7 cells with a variety of histone deacetylase inhibitors (HDACIs), at therapeutically and clinically relevant doses, identified a few additional StAR acetylated lysine residues. Among a total of fourteen StAR acetylomes undergoing acetylation and deacetylation, K111 and K253 were frequently recognized either endogenously or in response to HDACIs. Site-directed mutagenesis studies of these two StAR acetylomes, pertaining to K111Q and K253Q acetylation mimetic states, resulted in increases in E2 levels in ER+ MCF7 and triple negative MB-231 BC cells, compared with their values seen with human StAR. Conversely, these cells carrying K111R and K253R deacetylation mutants diminished E2 biosynthesis. These findings provide novel and mechanistic insights into intra-tumoral E2 regulation by elucidating the functional importance of this uncovered StAR post-translational modification (PTM), involving acetylation and deacetylation events, underscoring the potential of StAR as a therapeutic target for hormone-sensitive BC.
Collapse
Affiliation(s)
- Pulak R. Manna
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Deborah Molehin
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA;
| | - Ahsen U. Ahmed
- Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA;
| | - Shengping Yang
- Department of Biostatistics, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA;
| | - P. Hemachandra Reddy
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
- Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|