1
|
Altea-Manzano P, Decker-Farrell A, Janowitz T, Erez A. Metabolic interplays between the tumour and the host shape the tumour macroenvironment. Nat Rev Cancer 2025; 25:274-292. [PMID: 39833533 DOI: 10.1038/s41568-024-00786-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
Metabolic reprogramming of cancer cells and the tumour microenvironment are pivotal characteristics of cancers, and studying these processes offer insights and avenues for cancer diagnostics and therapeutics. Recent advancements have underscored the impact of host systemic features, termed macroenvironment, on facilitating cancer progression. During tumorigenesis, these inherent features of the host, such as germline genetics, immune profile and the metabolic status, influence how the body responds to cancer. In parallel, as cancer grows, it induces systemic effects beyond the primary tumour site and affects the macroenvironment, for example, through inflammation, the metabolic end-stage syndrome of cachexia, and metabolic dysregulation. Therefore, understanding the intricate metabolic interplay between the tumour and the host is a growing frontier in advancing cancer diagnosis and therapy. In this Review, we explore the specific contribution of the metabolic fitness of the host to cancer initiation, progression and response to therapy. We then delineate the complex metabolic crosstalk between the tumour, the microenvironment and the host, which promotes disease progression to metastasis and cachexia. The metabolic relationships among the host, cancer pathogenesis and the consequent responsive systemic manifestations during cancer progression provide new perspectives for mechanistic cancer therapy and improved management of patients with cancer.
Collapse
Affiliation(s)
| | | | | | - Ayelet Erez
- Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
2
|
Won Y, Kim HH, Jeong SH, Bhosale PB, Abusaliya A, Heo JD, Seong JK, Ahn MJ, Kim HJ, Kim GS. The Effects of Iridin and Irigenin on Cancer: Comparison with Well-Known Isoflavones in Breast, Prostate, and Gastric Cancers. Int J Mol Sci 2025; 26:2390. [PMID: 40141034 PMCID: PMC11942201 DOI: 10.3390/ijms26062390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Cancer, a worldwide problem and one of the leading causes of death due to uncontrolled cell proliferation, can be caused by various factors, such as genetic and environmental factors. Apoptosis is a programmed cell death mechanism that eliminates abnormal cells or renews cells. There are two main apoptotic pathways: intrinsic and extrinsic pathways. These pathways can be affected by various signaling pathways in cancer, such as the PI3K/AKT, MAPK, Wnt, and JAK/STAT pathways. Numerous approaches to cancer treatment have been studied, and among them, natural compounds have been actively researched. Flavonoids are natural compounds from fruits and vegetables and have been studied for their anti-cancer effects. Isoflavones, one of the subclasses of flavonoids, are usually found in soy food or legumes and are effective in several bioactive functions. The well-known isoflavones are genistein, daidzein, and glycitein. Irigenin and iridin can be extracted from the Iris family. Both irigenin and iridin are currently being studied for anti-inflammation, antioxidant, and anti-cancer by inducing apoptosis. In this review, we summarized five isoflavones, genistein, daidzein, glycitein, irigenin, and iridin and their effects on three different cancers: breast cancer, prostate cancer, and gastric cancer.
Collapse
Affiliation(s)
- Yaeram Won
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (Y.W.); (H.-J.K.)
- Department of Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Hun-Hwan Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.-H.K.); (S.-H.J.); (P.B.B.); (A.A.)
| | - Se-Hyo Jeong
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.-H.K.); (S.-H.J.); (P.B.B.); (A.A.)
| | - Pritam Bhagwan Bhosale
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.-H.K.); (S.-H.J.); (P.B.B.); (A.A.)
| | - Abuyaseer Abusaliya
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.-H.K.); (S.-H.J.); (P.B.B.); (A.A.)
| | - Jeong-Doo Heo
- Biological Resources Research Group, Gyeongnam Department of Environment Toxicology and Chemistry, Korea Institute of Toxicology, 17 Jegok-gil, Jinju 52834, Republic of Korea;
| | - Je-Kyung Seong
- Laboratory of Developmental Biology and Goenomics, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea;
| | - Mee-Jung Ahn
- Department of Animal Science, College of Life Science, Sangji University, Wonju 26339, Republic of Korea;
| | - Hye-Jung Kim
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (Y.W.); (H.-J.K.)
- Department of Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Gon-Sup Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.-H.K.); (S.-H.J.); (P.B.B.); (A.A.)
| |
Collapse
|
3
|
Yang R, Zhang G, Meng Z, Wang L, Li Y, Li H, Yan S, Wei X, Wang S, Cui H. Glutamate dehydrogenase 1-catalytic glutaminolysis feedback activates EGFR/PI3K/AKT pathway and reprograms glioblastoma metabolism. Neuro Oncol 2025; 27:668-681. [PMID: 39446525 PMCID: PMC11889723 DOI: 10.1093/neuonc/noae222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Glutamine is an important nutrient for cancer cell growth that provides biological sources for nucleic acid and fatty acid synthesis, but the role of glutaminolysis in signal transduction and glioblastoma (GBM) progression remains little known. METHODS Knockdown and overexpression cells were obtained to explore the functional roles of glutamate dehydrogenase 1 (GDH1) in cell proliferation, tumor formation, and aerobic glycolysis. RNA-seq, Chromatin immunoprecipitation, luciferase assay, and western blot were performed to verify the regulation of the EGFR-AKT pathway by the GDH1 (also known as GLUD1) and KDM6A. Metabolite-level measurements and Seahorse Assay were performed to assess the functional role of GHD1 in reprogramming glycolysis. RESULTS Here, we report that GDH1 catalytic glutaminolysis is essential for GBM cell line proliferation and brain tumorigenesis even in high-glucose conditions. Glutamine is metabolized through glutaminolysis to produce α-ketoglutarate (α-KG). We demonstrate that glutamine in combination with leucine activates mammalian TORC1 by enhancing glutaminolysis and α-KG production. α-KG increases the transcription of PDPK1 by reducing the suppressive histone modification H3K27me3 and then promotes the activation of the PI3K/AKT/mTOR pathway. This transcriptional activation induced by α-KG requires histone demethylase KDM6A, which is a 2-oxoglutarate oxygenase that plays an important role in converting α-KG to succinate. Furthermore, we show that GDH1-catalytic glutaminolysis also increases the expression of HK2 and promotes glycolysis in high-glucose conditions dependent on KDM6A-mediated demethylation of H3K27. CONCLUSIONS These findings suggest a novel function of glutaminolysis in the regulation of signal transduction and metabolism reprogramming and provide further evidence for the unique role of glutaminolysis in GBM progression.
Collapse
Affiliation(s)
- Rui Yang
- Biomedical Laboratory, School of Medicine, Liaocheng University, Liaocheng, China
| | - Guanghui Zhang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhen Meng
- Biomedical Laboratory, School of Medicine, Liaocheng University, Liaocheng, China
| | - Li Wang
- Biomedical Laboratory, School of Medicine, Liaocheng University, Liaocheng, China
| | - Yanping Li
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Haibin Li
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Siyuan Yan
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Xiaonan Wei
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Shanshan Wang
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Hongjuan Cui
- Jinfeng Laboratory, Chongqing, China
- Medical Research Institute, State Key Laboratory of Resources Insects, Southwest University, Chongqing, China
| |
Collapse
|
4
|
Caprara G, Pallavi R, Sanyal S, Pelicci PG. Dietary Restrictions and Cancer Prevention: State of the Art. Nutrients 2025; 17:503. [PMID: 39940361 PMCID: PMC11820753 DOI: 10.3390/nu17030503] [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: 12/27/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Worldwide, almost 10 million cancer deaths occurred in 2022, a number that is expected to rise to 16.3 million by 2040. Primary prevention has long been acknowledged as a crucial approach to reducing cancer incidence. In fact, between 30 and 50 percent of all tumors are known to be preventable by eating a healthy diet, staying active, avoiding alcohol, smoking, and being overweight. Accordingly, many international organizations have created tumor prevention guidelines, which underlie the importance of following a diet that emphasizes eating plant-based foods while minimizing the consumption of red/processed meat, sugars, processed foods, and alcohol. However, further research is needed to define the relationship between the effect of specific diets or nutritional components on cancer prevention. Interestingly, reductions in food intake and dietetic restrictions can extend the lifespan of yeast, nematodes, flies, and rodents. Despite controversial results in humans, those approaches have the potential to ameliorate health via direct and indirect effects on specific signaling pathways involved in cancer onset. Here, we describe the latest knowledge on the cancer-preventive potential of dietary restrictions and the biochemical processes involved. Molecular, preclinical, and clinical studies evaluating the effects of different fasting strategies will also be reviewed.
Collapse
Affiliation(s)
- Greta Caprara
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
| | - Rani Pallavi
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
- Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad 500034, India
- The Operation Eyesight Universal Institute for Eye Cancer, L. V. Prasad Eye Institute, Hyderabad 500034, India; (R.P.); (S.S.)
| | - Shalini Sanyal
- Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad 500034, India
- The Operation Eyesight Universal Institute for Eye Cancer, L. V. Prasad Eye Institute, Hyderabad 500034, India; (R.P.); (S.S.)
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20139 Milan, Italy
| |
Collapse
|
5
|
Yang X, Yang D, Qi X, Luo X, Zhang G. Endocrine treatment mechanisms in triple-positive breast cancer: from targeted therapies to advances in precision medicine. Front Oncol 2025; 14:1467033. [PMID: 39845328 PMCID: PMC11753220 DOI: 10.3389/fonc.2024.1467033] [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: 07/19/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Triple-positive breast cancer (TPBC), defined by the co-expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), poses unique therapeutic challenges due to complex signaling interactions and resulting treatment resistance. This review summarizes key findings on the molecular mechanisms and cross-talk among ER, PR, and HER2 pathways, which drive tumor proliferation and resistance to conventional therapies. Current strategies in TPBC treatment, including endocrine and HER2-targeted therapies, are explored alongside emerging approaches such as immunotherapy and CRISPR/Cas9 gene editing. Additionally, we discuss the tumor microenvironment (TME) and its role in treatment resistance, highlighting promising avenues for intervention through combination therapies and predictive biomarkers. By addressing these interdependent pathways and optimizing therapeutic strategies, precision medicine holds significant potential for improving TPBC patient outcomes and advancing individualized cancer care.
Collapse
Affiliation(s)
| | | | | | | | - Guangmei Zhang
- Department of Medical Oncology, Third Division, Jilin City Second People’s Hospital, Jilin, China
| |
Collapse
|
6
|
Yu X, Zhou G, Zhang M, Zhang N. ABCA8 Elevation Predicts the Prognosis and Exerts the Anti-oncogenic Effects on the Malignancy of Non-small Cell Lung Cancer via TCF21-Mediated Inactivation of PI3K/AKT. Mol Biotechnol 2025; 67:226-236. [PMID: 38153664 DOI: 10.1007/s12033-023-00998-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 11/22/2023] [Indexed: 12/29/2023]
Abstract
The malignant growth and metastatic potential of non-small-cell lung cancer (NSCLC) are the major causes for its poor prognosis. ATP-binding cassette (ABC) subfamily A member 8 (ABCA8) exerts contradictive roles in the development of several cancers. Nevertheless, its role in NSCLC remains unclear. In this study, three GEO datasets and bioinformatics databases (GEPIA2 and UALCAN) revealed the obvious down-regulation of ABCA8 in NSCLC tissues and cells, and this expression was associated with cancer stages and lymph node metastasis. Low expression of ABCA8 predicted poor survival in NSCLC. ABCA8 elevation inhibited cell proliferation and induced cell apoptosis. Moreover, ABCA8 overexpression suppressed cancer cell invasion. Mechanistically, ABCA8 was associated with TCF21 in NSCLC specimens and its overexpression enhanced TCF21 expression. ABCA8 elevation inactivated the PI3K/AKT signaling, which was reversed after TCF21 knockdown. Additionally, targeting TCF21 overturned the anti-oncogenic effects of ABCA8 elevation on cell proliferation, apoptosis and invasion. Thus, the current findings highlight that ABCA8 may be a promising prognostic marker and may act as a suppressor gene to regulate the malignancy of NSCLC cells via TCF21-mediated inactivation of PI3K/AKT signaling, supporting a new promising target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Xin Yu
- Department of General Medicine, Honghui Hospital Affiliated to Xi'an Jiaotong University, No. 555 Youyi East Road, Xi'an, 710054, People's Republic of China
| | - Guoqiong Zhou
- Department of General Medicine, Honghui Hospital Affiliated to Xi'an Jiaotong University, No. 555 Youyi East Road, Xi'an, 710054, People's Republic of China
| | - Ming Zhang
- Department of General Medicine, Honghui Hospital Affiliated to Xi'an Jiaotong University, No. 555 Youyi East Road, Xi'an, 710054, People's Republic of China
| | - Nana Zhang
- Department of General Medicine, Honghui Hospital Affiliated to Xi'an Jiaotong University, No. 555 Youyi East Road, Xi'an, 710054, People's Republic of China.
| |
Collapse
|
7
|
Zhang X, Shi L, Xing M, Li C, Ma F, Ma Y, Ma Y. Interplay between lncRNAs and the PI3K/AKT signaling pathway in the progression of digestive system neoplasms (Review). Int J Mol Med 2025; 55:15. [PMID: 39513614 PMCID: PMC11573320 DOI: 10.3892/ijmm.2024.5456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
Long non‑coding RNA (lncRNA) is a class of non‑coding RNA molecules located in the cytoplasm or nucleus, which can regulate chromosome structure and function by interacting with DNA, RNA, proteins and other molecules; binding to mRNA bases in a complementary manner, affecting the splicing, stabilization, translation and degradation of mRNA; acting as competing endogenous RNA competitively binds to microRNAs to regulate gene expression and participate in the regulation of various vital activities of the body. The PI3K/AKT signalling pathway plays a key role in numerous biological and cellular processes, such as cell proliferation, invasion, migration and angiogenesis. It has been found that the lncRNA/PI3K/AKT axis regulates the expression of cancer‑related genes and thus tumour progression. The abnormal regulation of lncRNA expression in the lncRNA/PI3K/AKT axis is clearly associated with clinicopathological features and plays an important role in regulating biological functions. In the present review, the expression and biological functions of PI3K/AKT‑related lncRNAs both in vitro and in vivo over recent years, were comprehensively summarized and analyzed. Their correlation with clinicopathological features was also evaluated, with the objective of furnishing a solid theoretical foundation for clinical diagnosis and the monitoring of efficacy in digestive system neoplasms. The present review aimed to provide a comprehensive overview of the expression and biological functions of PI3K/AKT‑related lncRNAs in digestive system neoplasms and to assess their correlation with clinicopathological features. This endeavor seeks to establish a solid theoretical foundation for the clinical diagnosis and efficacy monitoring of digestive system tumors.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Lei Shi
- Key Laboratory of New Material Research Institute, Institute of Pharmaceutical Research, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Mengzhen Xing
- Key Laboratory of New Material Research Institute, Institute of Pharmaceutical Research, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Chunjing Li
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Fengjun Ma
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Yuning Ma
- Key Laboratory of New Material Research Institute, Institute of Pharmaceutical Research, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Yuxia Ma
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| |
Collapse
|
8
|
Zhang P, Liu H, Yu Y, Peng S, Zeng A, Song L. Terpenoids mediated cell apoptotsis in cervical cancer: Mechanisms, advances and prospects. Fitoterapia 2025; 180:106323. [PMID: 39631509 DOI: 10.1016/j.fitote.2024.106323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/14/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Cervical cancer remains one of the most common malignancies among women globally, causing hundreds of thousands of deaths annually. Despite widespread vaccination and screening programs, the incidence of cervical cancer remains high in developing countries. OBJECTIVE This review aims to systematically summarize the existing terpenoids effective in preventing cervical cancer, elucidate their potential mechanisms in the prophylaxis and treatment of cervical cancer, and assess the limitations of current studies. RESULTS Studies have shown that terpenoids can decrease the incidence of cervical cancer and promote apoptosis of cancer cells through various signaling pathways, including the PI3K/AKT pathway, the endoplasmic reticulum stress (ERS) pathway, and the mitochondria- and caspase-dependent cell death pathways. Furthermore, some terpenoids have been found to enhance the sensitivity to chemotherapy drugs, thus improving patients' quality of life. CONCLUSION Terpenoids play a significant role in inhibiting the progression of cervical cancer. However, due to their diversity and complex mechanisms of action, further research is necessary to investigate their specific targets and bioactivities to advance their clinical trials and applications.
Collapse
Affiliation(s)
- Peng Zhang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| | - Hong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| | - Yuan Yu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| | - Shiyang Peng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan 610041, PR China.
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China.
| |
Collapse
|
9
|
Zheng K, Gao Y, Xu J, Kang M, Chai R, Jin G, Kang Y. mTOR Inhibitor Everolimus Modulates Tumor Growth in Small-Cell Carcinoma of the Ovary, Hypercalcemic Type and Augments the Drug Sensitivity of Cancer Cells to Cisplatin. Biomedicines 2024; 13:1. [PMID: 39857585 PMCID: PMC11759183 DOI: 10.3390/biomedicines13010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Small-cell carcinoma of the ovary, hypercalcemic type (SCCOHT), is a rare and aggressive cancer with a poor prognosis and limited treatment options. Current chemotherapy regimens are predominantly platinum-based; however, the development of platinum resistance during treatment significantly worsens patient outcomes. Everolimus, an mTOR inhibitor, has been widely used in combination cancer therapies and has successfully enhanced the efficacy of platinum-based treatments. Method: In this study, we investigated the combined effects of everolimus and cisplatin on SCCOHT through both in vitro and in vivo experiments, complemented by RNA sequencing (RNA-seq) analyses to further elucidate the therapeutic impact. Result: Our findings revealed that everolimus significantly inhibits the proliferation of SCCOHT cells, induces cell cycle arrest, and accelerates apoptosis. When combined with cisplatin, everolimus notably enhances the therapeutic efficacy without increasing the toxicity typically associated with platinum-based drugs. RNA-seq analysis uncovered alterations in the expression of apoptosis-related genes, suggesting that the underlying mechanism involves autophagy regulation. Conclusions: Despite the current challenges in treating SCCOHT and the suboptimal efficacy of platinum-based therapies, the addition of everolimus significantly suppresses tumor growth. This indicates that everolimus enhances cisplatin efficacy by disrupting survival-promoting signaling cascades and inducing cell cycle arrest. Furthermore, it points to potential biomarkers for predicting therapeutic response.
Collapse
Affiliation(s)
- Kewei Zheng
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (K.Z.); (Y.G.); (J.X.); (M.K.); (R.C.); (G.J.)
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Yi Gao
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (K.Z.); (Y.G.); (J.X.); (M.K.); (R.C.); (G.J.)
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Jing Xu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (K.Z.); (Y.G.); (J.X.); (M.K.); (R.C.); (G.J.)
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Mingyi Kang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (K.Z.); (Y.G.); (J.X.); (M.K.); (R.C.); (G.J.)
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Ranran Chai
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (K.Z.); (Y.G.); (J.X.); (M.K.); (R.C.); (G.J.)
| | - Guanqin Jin
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (K.Z.); (Y.G.); (J.X.); (M.K.); (R.C.); (G.J.)
| | - Yu Kang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (K.Z.); (Y.G.); (J.X.); (M.K.); (R.C.); (G.J.)
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| |
Collapse
|
10
|
Gan M, Liu N, Li W, Chen M, Bai Z, Liu D, Liu S. Metabolic targeting of regulatory T cells in oral squamous cell carcinoma: new horizons in immunotherapy. Mol Cancer 2024; 23:273. [PMID: 39696340 DOI: 10.1186/s12943-024-02193-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a prevalent oral malignancy, which poses significant health risks with a high mortality rate. Regulatory T cells (Tregs), characterized by their immunosuppressive capabilities, are intricately linked to OSCC progression and patient outcomes. The metabolic reprogramming of Tregs within the OSCC tumor microenvironment (TME) underpins their function, with key pathways such as the tryptophan-kynurenine-aryl hydrocarbon receptor, PI3K-Akt-mTOR and nucleotide metabolism significantly contributing to their suppressive activities. Targeting these metabolic pathways offers a novel therapeutic approach to reduce Treg-mediated immunosuppression and enhance anti-tumor responses. This review explores the metabolic dependencies and pathways that sustain Treg function in OSCC, highlighting key metabolic adaptations such as glycolysis, fatty acid oxidation, amino acid metabolism and PI3K-Akt-mTOR signaling pathway that enable Tregs to thrive in the challenging conditions of the TME. Additionally, the review discusses the influence of the oral microbiome on Treg metabolism and evaluates potential therapeutic strategies targeting these metabolic pathways. Despite the promising potential of these interventions, challenges such as selectivity, toxicity, tumor heterogeneity, and resistance mechanisms remain. The review concludes with perspectives on personalized medicine and integrative approaches, emphasizing the need for continued research to translate these findings into effective clinical applications for OSCC treatment.
Collapse
Affiliation(s)
- Menglai Gan
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Nanshu Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Wenting Li
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Mingwei Chen
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Zhongyu Bai
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Dongjuan Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China.
| | - Sai Liu
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China.
| |
Collapse
|
11
|
Jaiswal R, Bhati G, Ahmed S, Siddiqi MI. iDCNNPred: an interpretable deep learning model for virtual screening and identification of PI3Ka inhibitors against triple-negative breast cancer. Mol Divers 2024:10.1007/s11030-024-11055-9. [PMID: 39648257 DOI: 10.1007/s11030-024-11055-9] [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: 10/01/2024] [Accepted: 11/12/2024] [Indexed: 12/10/2024]
Abstract
Triple-negative breast cancer (TNBC) lacks estrogen, progesterone, and HER2 expression, accounting for 15-20% of breast cancer cases. It is challenging due to low therapeutic response, heterogeneity, and aggressiveness. The PI3Ka isoform is a promising therapeutic target, often hyperactivated in TNBC, contributing to uncontrolled growth and cancer cell formation. We have proposed an interpretable deep convolutional neural network prediction (iDCNNPred) system using 2D molecular images to classify bioactivity and identify potential PI3Ka inhibitors. We built Custom-DCNN models and pre-trained models such as AlexNet, SqueezeNet, and VGG19 by using the Bayesian optimization algorithm, and found that our Custom-DCNN model performed better than a pre-trained model with lower complexity and memory usage. All top-performed models were screened with the Maybridge Chemical library to find predictive hit molecules. The screened molecules were further evaluated for protein-ligand interaction with molecular docking and finally 12 promising hits were shortlisted for biological validation using in-vitro PI3K inhibition studies. After biological evaluation, 4 potent molecules with different structural moieties were identified, and these molecules present new starting scaffolds for further improvement in terms of their potency and selectivity as PI3K inhibitors with the help of medicinal chemistry efforts. Furthermore, we also showed the significance of the interpretation and visualization of the model's predictions by the Grad-CAM technique, enhancing the robustness, transparency, and interpretability of the model's predictions. The data and script files and prediction run of models used for this study to reproduce the experiment are available in the GitHub repository at https://github.com/ravishankar1307/iDCNNPred.git .
Collapse
Affiliation(s)
- Ravishankar Jaiswal
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Girdhar Bhati
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Shakil Ahmed
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Mohammad Imran Siddiqi
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
12
|
Abd Elkarim AS, Mohamed SH, Ali NA, Elsayed GH, Aly MS, Elgamal AM, Elsayed WM, El-Newary SA. The Phytochemical Profile of the Petroleum Ether Extract of Purslane Leaves and Its Anticancer Effect on 4-(Methylnitrosamino)-1-(3-pyridyl)-1-buta-4 None (NNK)-Induced Lung Cancer in Rats. Int J Mol Sci 2024; 25:13024. [PMID: 39684736 DOI: 10.3390/ijms252313024] [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: 10/06/2024] [Revised: 11/13/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Lung cancer is a prevalent and very aggressive sickness that will likely claim 1.8 million lives by 2022, with an estimated 2.2 million additional cases expected worldwide. The goal of the current investigation was to determine whether petroleum ether extract of purslane leaf could be used to treat lung cancer induced by 4-(Methylnitrosamino)-1-(3-pyridyl)-1-buta-4 none (NNK) in rats. In the in vitro extract recorded, promising anticancer effects in A540 cell lines with IC50 were close to the reference drug, doxorubicin (14.3 and 13.8 μg/mL, respectively). A dose of 500 mg/kg/day orally for 20 weeks exhibited recovery effects on NNK-induced lung cancer with a good safety margin, where Intercellular Adhesion Molecule-1 (ICAM-1), the lung cancer biomarker, was significantly reduced by about 18.75% compared to cancer control. Purslane exhibited many anticancer mechanisms, including (i) anti-proliferation as a significant reduction in Ki67 level (20.42%), (ii) anti-angiogenesis as evident by a considerable decrease in Matrix metalloproteinase-9 (MMP-9) expression (79%), (iii) anti-inflammation as a remarked decline in Insulin-like growth factor 1 (IGF-1) expression (62%), (iv) pro-apoptotic effect as a significant activation in Forkhead box protein O1 (FOXO1) expression (262%), and (v) anti-oxidation as remarkable activation on antioxidant biomarkers either non-enzymatic or enzymatic concurrent with considerable depletion on oxidative stress biomarker, in comparison to cancer control. The histopathological examination revealed that Purslane extract showed markedly improved tissue structure and reduced pathological changes across all examined organs caused by NNK. The anti-lung cancer effect exhibited by the extract may be linked to the active ingredients of the extract that were characterized by LC-MS, such as α-linolenic acid, linoleic acid, palmitic acid, β-sitosterol, and alkaloids (berberine and magnoflorine).
Collapse
Affiliation(s)
- Asmaa S Abd Elkarim
- Chemistry of Tanning Materials and Leather Technology Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Safaa H Mohamed
- Hormones Department, National Research Centre, 33 El-Bouhoths St., Dokki, Giza 12622, Egypt
| | - Naglaa A Ali
- Hormones Department, National Research Centre, 33 El-Bouhoths St., Dokki, Giza 12622, Egypt
| | - Ghada H Elsayed
- Hormones Department, National Research Centre, 33 El-Bouhoths St., Dokki, Giza 12622, Egypt
- Stem Cells Lab, Centre of Excellence for Advanced Sciences, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Mohamed S Aly
- Department of Animal Reproduction and Artificial Insemination, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Abdelbaset M Elgamal
- Department of Chemistry of Microbial and Natural Products, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Wael M Elsayed
- Chemistry of Medicinal Plants Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Samah A El-Newary
- Medicinal and Aromatic Plants Research Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| |
Collapse
|
13
|
Luo Z, Zhou F, Tan C, Yin L, Bao M, He X, Li H, Yan J. Targeting PDGF-CC as a promising therapeutic strategy to inhibit cholangiocarcinoma progression. J Transl Med 2024; 22:1023. [PMID: 39543636 PMCID: PMC11566273 DOI: 10.1186/s12967-024-05857-6] [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: 08/29/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA) is an aggressive malignancy with limited treatment options and poor prognosis. Platelet-Derived Growth Factor CC (PDGF-CC) has been implicated in the progression of various tumors, but its specific role in CCA is not well understood. This study aims to investigate the expression and function of PDGF-CC in CCA and evaluate its potential as a therapeutic target. METHODS We conducted gene expression analysis using the GEPIA database to compare PDGF-CC mRNA levels in CCA tissues and normal tissues. Serum samples from CCA patients were analyzed for PDGF-CC protein levels, and immunohistochemistry was used to assess PDGF-CC expression in tissue samples. The impact of PDGF-CC on CCA cell behavior was examined by knocking out PDGF-CC in HuCCT1 and QBC939 cell lines, followed by assessments of cell proliferation, migration, invasion, and colony formation in vitro. Additionally, the effects of PDGF-CC knockout were evaluated in xenograft models. The therapeutic potential of PDGF-CC inhibition was further explored using pharmacological inhibitors and antibodies. RESULTS PDGF-CC mRNA and protein levels were significantly elevated in CCA tissues and patient sera compared to normal controls. Immunohistochemical analysis confirmed increased PDGF-CC expression in CCA tissues. High PDGF-CC expression correlated with poor overall survival in CCA patients, as shown by Kaplan-Meier analysis. Functional assays revealed that PDGF-CC knockout significantly reduced proliferation, migration, invasion, and colony formation in HuCCT1 and QBC939 cells, the lines with the highest PDGF-CC levels. In vivo, PDGF-CC knockout markedly decreased tumor growth in xenograft models. Pharmacological inhibition of PDGF-CC mirrored the effects of genetic knockout, suggesting it as a viable therapeutic strategy. CONCLUSIONS This study underscores the critical role of PDGF-CC in CCA progression and supports the potential of PDGF-CC inhibitors as a therapeutic approach. Given the association between high PDGF-CC expression and poor prognosis, targeting PDGF-CC may improve outcomes for CCA patients. Further clinical investigations are warranted to develop PDGF-CC-targeted therapies for CCA.
Collapse
Affiliation(s)
- Zhenchao Luo
- Department of General Surgery, the Third Affiliated Hospital of Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Fangfang Zhou
- Department of Hepatobiliary surgery, the Third Affiliated Hospital of Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Canliang Tan
- Department of General Surgery, the Third Affiliated Hospital of Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Liangchun Yin
- Department of General Surgery, the Third Affiliated Hospital of Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Man Bao
- Department of pathology, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Xiang He
- Department of Hepatobiliary surgery, the Third Affiliated Hospital of Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Haohui Li
- Department of General Surgery, the Third Affiliated Hospital of Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, Guangdong, China.
| | - Jian Yan
- Department of Hepatobiliary surgery, the Third Affiliated Hospital of Southern Medical University, 183 Zhongshan Avenue West, Tianhe District, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
14
|
Zasheva D, Mladenov P, Zapryanova S, Gospodinova Z, Georgieva M, Alexandar I, Velinov V, Djilianov D, Moyankova D, Simova-Stoilova L. Cytotoxic Effects of Plant Secondary Metabolites and Naturally Occurring Bioactive Peptides on Breast Cancer Model Systems: Molecular Mechanisms. Molecules 2024; 29:5275. [PMID: 39598664 PMCID: PMC11596968 DOI: 10.3390/molecules29225275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Breast cancer is the second leading cause of death among women, and the number of mortal cases in diagnosed patients is constantly increasing. The search for new plant compounds with antitumor effects is very important because of the side effects of conventional therapy and the development of drug resistance in cancer cells. The use of plant substances in medicine has been well known for centuries, but the exact mechanism of their action is far from being elucidated. The molecular mechanisms of cytotoxicity exerted by secondary metabolites and bioactive peptides of plant origin on breast cancer cell lines are the subject of this review.
Collapse
Affiliation(s)
- Diana Zasheva
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Tsarigradsko Shosse, 73, 1113 Sofia, Bulgaria; (D.Z.); (S.Z.)
| | - Petko Mladenov
- Agrobioinstitute, Agricultural Academy, bul. “Dragan Tsankov” 8, 1164 Sofia, Bulgaria; (P.M.); (D.D.); (D.M.)
| | - Silvina Zapryanova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Tsarigradsko Shosse, 73, 1113 Sofia, Bulgaria; (D.Z.); (S.Z.)
| | - Zlatina Gospodinova
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Science, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria; (Z.G.); (M.G.); (V.V.)
| | - Mariyana Georgieva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Science, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria; (Z.G.); (M.G.); (V.V.)
| | - Irina Alexandar
- Institute of Molecular Biology “Rumen Tzanev”, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria;
| | - Valentin Velinov
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Science, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria; (Z.G.); (M.G.); (V.V.)
| | - Dimitar Djilianov
- Agrobioinstitute, Agricultural Academy, bul. “Dragan Tsankov” 8, 1164 Sofia, Bulgaria; (P.M.); (D.D.); (D.M.)
| | - Daniela Moyankova
- Agrobioinstitute, Agricultural Academy, bul. “Dragan Tsankov” 8, 1164 Sofia, Bulgaria; (P.M.); (D.D.); (D.M.)
| | - Lyudmila Simova-Stoilova
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Science, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria; (Z.G.); (M.G.); (V.V.)
| |
Collapse
|
15
|
Lian S, Du Z, Chen Q, Xia Y, Miao X, Yu W, Sun Q, Feng C. From lab to clinic: The discovery and optimization journey of PI3K inhibitors. Eur J Med Chem 2024; 277:116786. [PMID: 39180946 DOI: 10.1016/j.ejmech.2024.116786] [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: 07/24/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
PI3K inhibitors have emerged as promising therapeutic agents due to their critical role in various cellular processes, particularly in cancer, where the PI3K pathway is frequently dysregulated. This review explores the evolutionary path of PI3K inhibitors from laboratory discovery to clinical application. The journey begins with early laboratory investigations into PI3K signaling and inhibitor development, highlighting fundamental discoveries that laid the foundation for subsequent advancements. Optimization strategies, including medicinal chemistry approaches and structural modifications, are scrutinized for their contributions to enhancing inhibitor potency, selectivity, and pharmacokinetic properties. The translation from preclinical studies to clinical trials is examined, emphasizing pivotal trials that evaluated efficacy and safety profiles. Challenges encountered during clinical development are critically assessed. Finally, the review discusses ongoing research directions and prospects for PI3K inhibitors, underscoring these agents' continuous evolution and therapeutic potential.
Collapse
Affiliation(s)
- Siyu Lian
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhenhua Du
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qingqing Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinxin Miao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weiwei Yu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Qian Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Chong Feng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
16
|
Waki Y, Morine Y, Saito Y, Teraoku H, Yamada S, Ikemoto T, Tominaga T, Shimada M. Lenvatinib-resistant hepatocellular carcinoma promotes malignant potential of tumor-associated macrophages via exosomal miR-301a-3p. Ann Gastroenterol Surg 2024; 8:1084-1095. [PMID: 39502738 PMCID: PMC11533007 DOI: 10.1002/ags3.12814] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 11/08/2024] Open
Abstract
Background The interactions between cancer cells and tumor-associated macrophages (TAMs) via microRNAs (miRNAs) play crucial roles in malignant potential and drug resistance. However, it remains unclear how lenvatinib-resistant hepatocellular carcinoma (LR HCC) promotes TAM tumor biology. Here we investigated the crosstalk between LR HCC cells and TAMs for cancer progression and lenvatinib resistance, focusing on an exosomal miRNA. Methods We used two bioinformatics software programs to identify miRNAs that target PTEN in gastrointestinal cancers, then investigated exosomal miRNA expression in LR HCC conditioned medium (CM). After modifying TAMs with LR HCC CM (LR TAM), macrophage phenotype and PTEN-Nrf2 signaling pathway component expression were analyzed in LR TAMs. The malignant potential and drug resistance were investigated in naïve HCC cells cultured with LR TAM CM. Results LR HCC cells highly induced M2-like properties in macrophages compared with naïve HCC cells. Exosomal miR-301a-3p expression was increased in LR HCC CM, with higher activation of the PTEN/PI3K/GSK3β/Nrf2 signaling pathway in LR TAMs. Naïve HCC cells were educated with LR TAM CM to promote malignant potential and lenvatinib resistance. Inhibition of exosomal miR-301a-3p prevented the malignant potential of LR TAMs. Activation of Nrf2 signaling by LR HCC cell-derived exosomal miR-301a-3p skewed the transformation of macrophages to the M2 phenotype. Conclusion Our study provides new findings on the role of miR-301a-3p, suggesting it is a promising therapeutic target to improve HCC lenvatinib resistance.
Collapse
Affiliation(s)
- Yuhei Waki
- Department of Digestive and Transplant SurgeryTokushima UniversityTokushimaJapan
| | - Yuji Morine
- Department of Digestive and Transplant SurgeryTokushima UniversityTokushimaJapan
| | - Yu Saito
- Department of Digestive and Transplant SurgeryTokushima UniversityTokushimaJapan
| | - Hiroki Teraoku
- Department of Digestive and Transplant SurgeryTokushima UniversityTokushimaJapan
| | - Shinichiro Yamada
- Department of Digestive and Transplant SurgeryTokushima UniversityTokushimaJapan
| | - Tetsuya Ikemoto
- Department of Digestive and Transplant SurgeryTokushima UniversityTokushimaJapan
| | - Tatsuya Tominaga
- Department of Bioanalytical TechnologyTokushima UniversityTokushimaJapan
| | - Mitsuo Shimada
- Department of Digestive and Transplant SurgeryTokushima UniversityTokushimaJapan
| |
Collapse
|
17
|
Xiong J, Zhou Z, Jiang Y, Li Q, Geng Z, Guo J, Yan C, Zhang J. Hypoxic stabilization of RIPOR3 mRNA via METTL3-mediated m 6A methylation drives breast cancer progression and metastasis. Oncogene 2024; 43:3426-3441. [PMID: 39341989 DOI: 10.1038/s41388-024-03180-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Dysregulated N6-methyladenosine (m6A) modification has been associated with breast cancer pathogenesis. Hypoxia which characterizes solid tumors is known to reprogram the m6A epitranscriptome, but the underlying mechanisms of how this process contributes to breast cancer progression remain poorly understood. Through integrative analyses of m6A-RIP sequencing and RNA sequencing databases, we reveal a cluster of mRNAs with upregulated m6A methylation and expression under hypoxia, that are enriched by many oncogenic pathways, including PI3K-Akt signaling. Furthermore, we identify the mRNA, RIPOR3, as a target of METTL3-mediated m6A methylation in response to hypoxia. We find that m6A methylation stabilizes RIPOR3, increasing its protein expression in a METTL3 catalytic activity-dependent manner, and consequently driving breast tumor growth and metastasis. RIPOR3 is found to be overexpressed in breast cancer cell lines and tumor tissues from breast cancer patients, in whom elevated RIPOR3 is associated with a worse prognosis. Mechanistically, we show that RIPOR3 interacts with EGFR and is essential for the PI3K-Akt pathway activation. In conclusion, we identify RIPOR3 as a hypoxia-stabilized oncogenic driver via METTL3-mediated m6A methylation, thus provide a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Jingjing Xiong
- Department of Urology, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Zirui Zhou
- Department of Urology, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yulong Jiang
- Department of Urology, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Qifang Li
- Department of Urology, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Zuhan Geng
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahao Guo
- Department of Urology, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Chaojun Yan
- Department of Urology, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China.
| | - Jing Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China.
| |
Collapse
|
18
|
Gupta I, Gaykalova DA. Unveiling the role of PIK3R1 in cancer: A comprehensive review of regulatory signaling and therapeutic implications. Semin Cancer Biol 2024; 106-107:58-86. [PMID: 39197810 DOI: 10.1016/j.semcancer.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/11/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Phosphoinositide 3-kinase (PI3K) is responsible for phosphorylating phosphoinositides to generate secondary signaling molecules crucial for regulating various cellular processes, including cell growth, survival, and metabolism. The PI3K is a heterodimeric enzyme complex comprising of a catalytic subunit (p110α, p110β, or p110δ) and a regulatory subunit (p85). The binding of the regulatory subunit, p85, with the catalytic subunit, p110, forms an integral component of the PI3K enzyme. PIK3R1 (phosphoinositide-3-kinase regulatory subunit 1) belongs to class IA of the PI3K family. PIK3R1 exhibits structural complexity due to alternative splicing, giving rise to distinct isoforms, prominently p85α and p55α. While the primary p85α isoform comprises multiple domains, including Src homology 3 (SH3) domains, a Breakpoint Cluster Region Homology (BH) domain, and Src homology 2 (SH2) domains (iSH2 and nSH2), the shorter isoform, p55α, lacks certain domains present in p85α. In this review, we will highlight the intricate regulatory mechanisms governing PI3K signaling along with the impact of PIK3R1 alterations on cellular processes. We will further delve into the clinical significance of PIK3R1 mutations in various cancer types and their implications for prognosis and treatment outcomes. Additionally, we will discuss the evolving landscape of targeted therapies aimed at modulating PI3K-associated pathways. Overall, this review will provide insights into the dynamic interplay of PIK3R1 in cancer, fostering advancements in precision medicine and the development of targeted interventions.
Collapse
Affiliation(s)
- Ishita Gupta
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Otorhinolaryngology-Head and Neck Surgery, Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
| | - Daria A Gaykalova
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Otorhinolaryngology-Head and Neck Surgery, Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
19
|
Li C, Luo P, Guo F, Jia X, Shen M, Zhang T, Wang S, Du T. Identification of HSPG2 as a bladder pro-tumor protein through NID1/AKT signaling. Cancer Cell Int 2024; 24:345. [PMID: 39438949 PMCID: PMC11515648 DOI: 10.1186/s12935-024-03527-7] [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: 06/25/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
PURPOSE Heparan sulfate proteoglycans (HSPGs) are complex molecules found on the cell membrane and within the extracellular matrix, increasingly recognized for their role in tumor progression. This study aimed to investigate the involvement of Heparan sulfate proteoglycan 2 (HSPG2) in the progression of bladder cancer. METHODS We identified HSPG2 as a promoter of bladder tumor progression using single-cell RNA sequencing and transcriptome analysis of sequencing data from seven patient samples obtained from the Gene Expression Omnibus (GEO) database (GSE135337). Transcript profiles of 28 normal tissues and 407 bladder tumor tissues were analyzed for HSPG2 expression and survival outcomes using the Sanger tools and cBioPortal databases. HSPG2-overexpressing T24 and Biu-87 cell lines were generated, and cell proliferation and migration were assessed using CCK-8 and Transwell assays. Western blotting and immunostaining were performed to evaluate the activation of Nidogen-1 (NID1)/protein kinase B (AKT) signaling. Mouse models with patient-derived tumor organoids (HSPG2high and HSPG2low) were established to assess anticancer effects. RESULTS Our results demonstrated a marked upregulation of HSPG2 in malignant bladder tumors, which correlated significantly with poor patient prognosis. HSPG2 overexpression consistently enhanced bladder tumor cell proliferation and conferred chemotherapy resistance, as shown in both in vitro and in vivo experiments. Mechanistically, HSPG2 upregulated NID1 expression, leading to the activation of the AKT pro-survival signaling pathway and promoting sustained tumor growth in bladder cancer. CONCLUSION This study highlights the critical pro-tumor role of HSPG2/NID1/AKT signaling in bladder cancer and suggests its potential as a therapeutic target in clinical treatment.
Collapse
Affiliation(s)
- Cong Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Pengwei Luo
- School of Clinical Medicine & The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Fengzhu Guo
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xu Jia
- School of Clinical Medicine & The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Min Shen
- School of Clinical Medicine & The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Ting Zhang
- School of Clinical Medicine & The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Shusen Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Ting Du
- Non-Coding RNA and Drug Discovery Key Laboratory of Sichuan Province, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, China.
| |
Collapse
|
20
|
Liu J, Wang Y, Tian M, Xia M, Zheng Y, Hao M, Qian Y, Shu H, Zhang W, Peng P, Zhao Z, Dong K, Peng W, Gao T, Li Z, Jin X, Wei M, Feng Y. O-GlcNAcylation of ATP-citrate lyase couples glucose supply to lipogenesis for rapid tumor cell proliferation. Proc Natl Acad Sci U S A 2024; 121:e2402674121. [PMID: 39388261 PMCID: PMC11494317 DOI: 10.1073/pnas.2402674121] [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: 02/15/2024] [Accepted: 08/26/2024] [Indexed: 10/12/2024] Open
Abstract
Elevated lipid synthesis is one of the best-characterized metabolic alterations in cancer and crucial for membrane expansion. As a key rate-limiting enzyme in de novo fatty acid synthesis, ATP-citrate lyase (ACLY) is frequently up-regulated in tumors and regulated by posttranslational modifications (PTMs). Despite emerging evidence showing O-GlcNAcylation on ACLY, its biological function still remains unknown. Here, we observed a significant upregulation of ACLY O-GlcNAcylation in various types of human tumor cells and tissues and identified S979 as a major O-GlcNAcylation site. Importantly, S979 O-GlcNAcylation is required for substrate CoA binding and crucial for ACLY enzymatic activity. Moreover, it is sensitive to glucose fluctuation and decisive for fatty acid synthesis as well as tumor cell proliferation. In response to EGF stimulation, both S979 O-GlcNAcylation and previously characterized S455 phosphorylation played indispensable role in the regulation of ACLY activity and cell proliferation; however, they functioned independently from each other. In vivo, streptozocin treatment- and EGFR overexpression-induced growth of xenograft tumors was mitigated once S979 was mutated. Collectively, this work helps comprehend how cells interrogate the nutrient enrichment for proliferation and suggests that although mammalian cell proliferation is controlled by mitogen signaling, the ancient nutrition-sensing mechanism is conserved and still efficacious in the cells of multicellular organisms.
Collapse
Affiliation(s)
- Jia Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Yang Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Miaomiao Tian
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Mingjie Xia
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Yi Zheng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Miao Hao
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin130033, People’s Republic Of China
| | - Yuqiang Qian
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun130062, China
| | - Hengyao Shu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Wenxia Zhang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Pinghui Peng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Zhexuan Zhao
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Kejian Dong
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Wanting Peng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Tian Gao
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Zhanjun Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun130062, China
| | - Xin Jin
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Min Wei
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| | - Yunpeng Feng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, Jilin130024, People’s Republic Of China
| |
Collapse
|
21
|
Golla U, Patel S, Shah N, Talamo S, Bhalodia R, Claxton D, Dovat S, Sharma A. From Deworming to Cancer Therapy: Benzimidazoles in Hematological Malignancies. Cancers (Basel) 2024; 16:3454. [PMID: 39456548 PMCID: PMC11506385 DOI: 10.3390/cancers16203454] [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: 08/16/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Drug repurposing is a strategy to discover new therapeutic uses for existing drugs, which have well-established toxicity profiles and are often more affordable. This approach has gained significant attention in recent years due to the high costs and low success rates associated with traditional drug development. Drug repositioning offers a more time- and cost-effective path for identifying new treatments. Several FDA-approved non-chemotherapy drugs have been investigated for their anticancer potential. Among these, anthelmintic benzimidazoles (such as albendazole, mebendazole, and flubendazole) have garnered interest due to their effects on microtubules and oncogenic signaling pathways. Blood cancers, which frequently develop resistance and have high mortality rates, present a critical need for effective therapies. This review highlights the recent advances in repurposing benzimidazoles for blood malignancies. These compounds induce cell cycle arrest, differentiation, tubulin depolymerization, loss of heterozygosity, proteasomal degradation, and inhibit oncogenic signaling to exert their anticancer effects. We also discuss current limitations and strategies to overcome them, emphasizing the potential of combining benzimidazoles with standard therapies for improved treatment of hematological cancers.
Collapse
Affiliation(s)
- Upendarrao Golla
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (D.C.)
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Satyam Patel
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| | - Nyah Shah
- Department of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada;
| | - Stella Talamo
- Department of Medicine, Liberty University College of Osteopathic Medicine, Lynchburg, VA 24502, USA;
| | - Riya Bhalodia
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (R.B.); (S.D.)
| | - David Claxton
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (D.C.)
| | - Sinisa Dovat
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (R.B.); (S.D.)
| | - Arati Sharma
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| |
Collapse
|
22
|
Ding Q, Cai J, Jin L, Hu W, Song W, Rose P, Tang Z, Zhan Y, Bao L, Lei W, Zhu YZ. A novel small molecule ZYZ384 targeting SMYD3 for hepatocellular carcinoma via reducing H3K4 trimethylation of the Rac1 promoter. MedComm (Beijing) 2024; 5:e711. [PMID: 39286779 PMCID: PMC11401973 DOI: 10.1002/mco2.711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/19/2024] [Accepted: 06/04/2024] [Indexed: 09/19/2024] Open
Abstract
SMYD3 (SET and MYND domain-containing 3) is a histone lysine methyltransferase highly expressed in different types of cancer(s) and is a promising epigenetic target for developing novel antitumor therapeutics. No selective inhibitors for this protein have been developed for cancer treatment. Therefore, the current study describes developing and characterizing a novel small molecule ZYZ384 screened and synthesized based on SMYD3 structure. Virtual screening was initially used to identify a lead compound and followed up by modification to get the novel molecules. Several technologies were used to facilitate compound screening about these novel molecules' binding affinities and inhibition activities with SMYD3 protein; the antitumor activity has been assessed in vitro using various cancer cell lines. In addition, a tumor-bearing nude mice model was established, and the activity of the selected molecule was determined in vivo. Both RNA-seq and chip-seq were performed to explore the antitumor mechanism. This work identified a novel small molecule ZYZ384 targeting SMYD3 with antitumor activity and impaired hepatocellular carcinoma tumor growth by reducing H3K4 trimethylation of the Rac1 promoter triggering the tumor cell cycle arrest through the AKT pathway.
Collapse
Affiliation(s)
- Qian Ding
- State Key Laboratory of Quality Research in Chinese Medicine & Laboratory of Drug Discovery from Natural Resources and Industrialization & School of Pharmacy Macau University of Science and Technology Macau SAR China
- Affiliated Hospital of Guangdong Medical University Zhanjiang China
- Joint Laboratory of TCM Innovation (Transformation) of Guizhou and Macau Guizhou University of Traditional Chinese Medicine Guiyang China
| | - Jianghong Cai
- State Key Laboratory of Quality Research in Chinese Medicine & Laboratory of Drug Discovery from Natural Resources and Industrialization & School of Pharmacy Macau University of Science and Technology Macau SAR China
| | - Li Jin
- State Key Laboratory of Quality Research in Chinese Medicine & Laboratory of Drug Discovery from Natural Resources and Industrialization & School of Pharmacy Macau University of Science and Technology Macau SAR China
| | - Wei Hu
- State Key Laboratory of Quality Research in Chinese Medicine & Laboratory of Drug Discovery from Natural Resources and Industrialization & School of Pharmacy Macau University of Science and Technology Macau SAR China
| | - Wu Song
- State Key Laboratory of Quality Research in Chinese Medicine & Laboratory of Drug Discovery from Natural Resources and Industrialization & School of Pharmacy Macau University of Science and Technology Macau SAR China
| | - Peter Rose
- School of Biosciences University of Nottingham Loughborough UK
| | - Zhiyuan Tang
- Department of Pharmacy Affiliated Hospital of Nantong University & Medical School of Nantong University Nantong China
| | - Yangyang Zhan
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital Navy Military Medical University Shanghai China
| | - Leilei Bao
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital Navy Military Medical University Shanghai China
| | - Wei Lei
- Affiliated Hospital of Guangdong Medical University Zhanjiang China
| | - Yi Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine & Laboratory of Drug Discovery from Natural Resources and Industrialization & School of Pharmacy Macau University of Science and Technology Macau SAR China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy Fudan University Shanghai China
| |
Collapse
|
23
|
Krymchenko R, Coşar Kutluoğlu G, van Hout N, Manikowski D, Doberenz C, van Kuppevelt TH, Daamen WF. Elastogenesis in Focus: Navigating Elastic Fibers Synthesis for Advanced Dermal Biomaterial Formulation. Adv Healthc Mater 2024; 13:e2400484. [PMID: 38989717 DOI: 10.1002/adhm.202400484] [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: 02/07/2024] [Revised: 05/31/2024] [Indexed: 07/12/2024]
Abstract
Elastin, a fibrous extracellular matrix (ECM) protein, is the main component of elastic fibers that are involved in tissues' elasticity and resilience, enabling them to undergo reversible extensibility and to endure repetitive mechanical stress. After wounding, it is challenging to regenerate elastic fibers and biomaterials developed thus far have struggled to induce its biosynthesis. This review provides a comprehensive summary of elastic fibers synthesis at the cellular level and its implications for biomaterial formulation, with a particular focus on dermal substitutes. The review delves into the intricate process of elastogenesis by cells and investigates potential triggers for elastogenesis encompassing elastin-related compounds, ECM components, and other molecules for their potential role in inducing elastin formation. Understanding of the elastogenic processes is essential for developing biomaterials that trigger not only the synthesis of the elastin protein, but also the formation of a functional and branched elastic fiber network.
Collapse
Affiliation(s)
- Roman Krymchenko
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Gizem Coşar Kutluoğlu
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
- MedSkin Solutions Dr. Suwelack AG, 48727, Billerbeck, Germany
| | - Noor van Hout
- Department of Dermatology, Radboud university medical center, Nijmegen, 6525 GA, The Netherlands
| | | | | | - Toin H van Kuppevelt
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Willeke F Daamen
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| |
Collapse
|
24
|
Şeker Karatoprak G, Dumlupınar B, Celep E, Kurt Celep I, Küpeli Akkol E, Sobarzo-Sánchez E. A comprehensive review on the potential of coumarin and related derivatives as multi-target therapeutic agents in the management of gynecological cancers. Front Pharmacol 2024; 15:1423480. [PMID: 39364049 PMCID: PMC11447453 DOI: 10.3389/fphar.2024.1423480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/16/2024] [Indexed: 10/05/2024] Open
Abstract
Current treatments for gynecological cancers include surgery, radiotherapy, and chemotherapy. However, these treatments often have significant side effects. Phytochemicals, natural compounds derived from plants, offer promising anticancer properties. Coumarins, a class of benzopyrone compounds found in various plants like tonka beans, exhibit notable antitumor effects. These compounds induce cell apoptosis, target PI3K/Akt/mTOR signaling pathways, inhibit carbonic anhydrase, and disrupt microtubules. Additionally, they inhibit tumor multidrug resistance and angiogenesis and regulate reactive oxygen species. Specific coumarin derivatives, such as auraptene, praeruptorin, osthole, and scopoletin, show anti-invasive, anti-migratory, and antiproliferative activities by arresting the cell cycle and inducing apoptosis. They also inhibit metalloproteinases-2 and -9, reducing tumor cell migration, invasion, and metastasis. These compounds can sensitize tumor cells to radiotherapy and chemotherapy. Synthetic coumarin derivatives also demonstrate potent antitumor and anticancer activities with minimal side effects. Given their diverse mechanisms of action and minimal side effects, coumarin-class phytochemicals hold significant potential as therapeutic agents in gynecological cancers, potentially improving treatment outcomes and reducing side effects. This review will aid in the synthesis and development of novel coumarin-based drugs for these cancers.
Collapse
Affiliation(s)
| | - Berrak Dumlupınar
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Istanbul Okan University, İstanbul, Türkiye
| | - Engin Celep
- Department of Pharmacognosy, Faculty of Pharmacy, Acıbadem Mehmet Ali Aydinlar University, Atasehir, Istanbul, Türkiye
| | - Inci Kurt Celep
- Department of Biotechnology, Faculty of Pharmacy, Istanbul Okan University, Istanbul, Türkiye
| | - Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
| | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación y Postgrado Facultad de Ciencias de la Salud Universidad Central de Chile, Santiago, Chile
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
25
|
Hossain MA. Targeting the RAS upstream and downstream signaling pathway for cancer treatment. Eur J Pharmacol 2024; 979:176727. [PMID: 38866361 DOI: 10.1016/j.ejphar.2024.176727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Cancer often involves the overactivation of RAS/RAF/MEK/ERK (MAPK) and PI3K-Akt-mTOR pathways due to mutations in genes like RAS, RAF, PTEN, and PIK3CA. Various strategies are employed to address the overactivation of these pathways, among which targeted therapy emerges as a promising approach. Directly targeting specific proteins, leads to encouraging results in cancer treatment. For instance, RTK inhibitors such as imatinib and afatinib selectively target these receptors, hindering ligand binding and reducing signaling initiation. These inhibitors have shown potent efficacy against Non-Small Cell Lung Cancer. Other inhibitors, like lonafarnib targeting Farnesyltransferase and GGTI 2418 targeting geranylgeranyl Transferase, disrupt post-translational modifications of proteins. Additionally, inhibition of proteins like SOS, SH2 domain, and Ras demonstrate promising anti-tumor activity both in vivo and in vitro. Targeting downstream components with RAF inhibitors such as vemurafenib, dabrafenib, and sorafenib, along with MEK inhibitors like trametinib and binimetinib, has shown promising outcomes in treating cancers with BRAF-V600E mutations, including myeloma, colorectal, and thyroid cancers. Furthermore, inhibitors of PI3K (e.g., apitolisib, copanlisib), AKT (e.g., ipatasertib, perifosine), and mTOR (e.g., sirolimus, temsirolimus) exhibit promising efficacy against various cancers such as Invasive Breast Cancer, Lymphoma, Neoplasms, and Hematological malignancies. This review offers an overview of small molecule inhibitors targeting specific proteins within the RAS upstream and downstream signaling pathways in cancer.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
26
|
Yang S, Li D. Role of microRNAs in triple‑negative breast cancer and new therapeutic concepts (Review). Oncol Lett 2024; 28:431. [PMID: 39049985 PMCID: PMC11268089 DOI: 10.3892/ol.2024.14565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Breast cancer has surpassed lung cancer as the most prevalent malignancy affecting women worldwide. Triple-negative breast cancer (TNBC) is the type of breast cancer with the worst prognosis. As a heterogeneous disease, TNBC has a pathogenesis that involves multiple oncogenic pathways, including involvement of gene mutations and alterations in signaling pathways. MicroRNAs (miRNAs) are small endogenous, single-stranded non-coding RNAs that bind to the 3' untranslated region of target cell mRNAs to negatively regulate the gene expression of these specific mRNAs. Therefore, miRNAs are involved in cell growth, development, division and differentiation stages. miRNAs are also involved in gene targeting in tumorigenesis, tumor growth and the regulation of metastasis, including in breast cancer. Meanwhile, miRNAs also regulate components of signaling pathways. In this review, the role of miRNAs in the TNBC signaling pathway discovered in recent years is described in detail. The new concept of bi-targeted therapy for breast cancer using miRNA and artificial intelligence is also discussed.
Collapse
Affiliation(s)
- Shaofeng Yang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Donghai Li
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| |
Collapse
|
27
|
Liu X, Zhou C, Cheng B, Xiong Y, Zhou Q, Wan E, He Y. Genipin promotes the apoptosis and autophagy of neuroblastoma cells by suppressing the PI3K/AKT/mTOR pathway. Sci Rep 2024; 14:20231. [PMID: 39215133 PMCID: PMC11364629 DOI: 10.1038/s41598-024-71123-w] [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: 12/11/2023] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
This study investigated the underlying function and mechanism of genipin in neuroblastoma (NB). Using flow cytometry analysis and cytotoxicity tests, in vitro studies were conducted to assess the effects of genipin on the SK-N-SH cell line. The mechanism of action of genipin was explored through immunofluorescence staining, Western blotting, and caspase-3 activity assays. In addition, we also created a xenograft tumour model to investigate the effects of genipin in vivo. This research confirmed that genipin suppressed cell viability, induced apoptosis, and promoted autophagy, processes that are likely linked to the inhibition of the PI3K/AKT/mTOR signalling pathway. Autophagy inhibition increases the sensitivity of SK-N-SH cells to genipin. Furthermore, combination treatment with a PI3K inhibitor enhanced the therapeutic efficacy of genipin. These results highlight the potential of genipin as a candidate drug for the treatment of NB.
Collapse
Affiliation(s)
- Xinying Liu
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
- Science and Technology Innovation Centre, North Sichuan Medical College, Shunqing District, Nanchong, 637000, Sichuan, China
- Institute of Hepatobiliary Research, North Sichuan Medical College, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Can Zhou
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Boli Cheng
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Yan Xiong
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Qin Zhou
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Enyu Wan
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China
| | - Yun He
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Shunqing District, Nanchong, 637000, Sichuan, China.
| |
Collapse
|
28
|
Zajec Ž, Dernovšek J, Cingl J, Ogris I, Gedgaudas M, Zubrienė A, Mitrović A, Golič Grdadolnik S, Gobec M, Tomašič T. New Class of Hsp90 C-Terminal Domain Inhibitors with Anti-tumor Properties against Triple-Negative Breast Cancer. J Med Chem 2024; 67:12984-13018. [PMID: 39042910 PMCID: PMC11320583 DOI: 10.1021/acs.jmedchem.4c00932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/22/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
Triple-negative breast cancer (TNBC) remains a treatment challenge and requires innovative therapies. Hsp90, crucial for the stability of numerous oncogenic proteins, has emerged as a promising therapeutic target. In this study, we present the optimization of the Hsp90 C-terminal domain (CTD) inhibitor TVS21. Biochemical methods, NMR binding studies, and molecular modeling were employed to investigate the binding of representative analogs to Hsp90. The newly synthesized analogs showed increased antiproliferative activity in breast cancer cell lines, including the MDA-MB-231 TNBC cell line. Compounds 89 and 104 proved to be the most effective, inducing apoptosis, slowing proliferation, and degrading key oncogenic proteins without inducing a heat shock response. In vivo, compound 89 showed comparable efficacy to the clinical candidate AUY922 and a better safety profile in a TNBC xenograft model. These results highlight the promise of Hsp90 CTD inhibitors for TNBC therapy, potentially filling a significant treatment gap.
Collapse
Affiliation(s)
- Živa Zajec
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Jaka Dernovšek
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Jernej Cingl
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Iza Ogris
- Laboratory
for Molecular Structural Dynamics, Theory Department, National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Marius Gedgaudas
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania
| | - Asta Zubrienė
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania
| | - Ana Mitrović
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
- Department
of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Simona Golič Grdadolnik
- Laboratory
for Molecular Structural Dynamics, Theory Department, National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Martina Gobec
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Tihomir Tomašič
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
29
|
Hua R, Zhao K, Xu Z, Zheng Y, Wu C, Zhang L, Teng Y, Wang J, Wang M, Hu J, Chen L, Yuan D, Dong W, Cheng X, Xia Y. Stratifin-mediated activation of AKT signaling and therapeutic targetability in hepatocellular carcinoma progression. CELL INSIGHT 2024; 3:100178. [PMID: 39027058 PMCID: PMC11254524 DOI: 10.1016/j.cellin.2024.100178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 07/20/2024]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide and presents a significant threat to human health. Despite its prevalence, the underlying regulatory mechanisms of HCC remain unclear. In this study, we integrated RNA-seq datasets, proteome dataset and survival analysis and unveiled Stratifin (SFN) as a potential prognostic biomarker for HCC. SFN knockdown inhibited HCC progression in cell cultures and mouse models. Conversely, ectopic expression of Sfn in primary mouse HCC model accelerated HCC progression. Mechanistically, SFN acted as an adaptor protein, activating AKT1 signaling by fostering the interaction between PDK1 and AKT1, with the R56 and R129 sites on SFN proving to be crucial for this binding. In the syngeneic implantation model, the R56A/R129A mutant of SFN inhibited Akt signaling activation and impeded HCC growth. Additionally, peptide inhibitors designed based on the binding motif of AKT1 to SFN significantly inhibited HCC progression. In summary, our findings establish that SFN promotes HCC progression by activating AKT signaling through the R56 and R129 binding sites. This discovery opens new avenues for a promising therapeutic strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Rong Hua
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Kaitao Zhao
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Zaichao Xu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Yingcheng Zheng
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Chuanjian Wu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Lu Zhang
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Yan Teng
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Jingjing Wang
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Mengfei Wang
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Jiayu Hu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Lang Chen
- Department of Immunology, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Detian Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Xiaoming Cheng
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
- Wuhan University Center for Pathology and Molecular Diagnostics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| | - Yuchen Xia
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
- Pingyuan Laboratory, Henan, China
| |
Collapse
|
30
|
Bartkowiak-Wieczorek J, Malesza M, Malesza I, Hadada T, Winkler-Galicki J, Grzelak T, Mądry E. Methylsulfinyl Hexyl Isothiocyanate (6-MSITC) from Wasabi Is a Promising Candidate for the Treatment of Cancer, Alzheimer's Disease, and Obesity. Nutrients 2024; 16:2509. [PMID: 39125389 PMCID: PMC11313713 DOI: 10.3390/nu16152509] [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: 07/02/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Methylsulfinyl hexyl isothiocyanate (6-MSITC) isolated from Eutrema japonicum is a promising candidate for the treatment of breast cancer, colorectal and stomach cancer, metabolic syndrome, heart diseases, diabetes, and obesity due to its anti-inflammatory and antioxidant properties. Also, its neuroprotective properties, improving cognitive function and protecting dopaminergic neurons, make it an excellent candidate for treating neurodegenerative diseases like dementia, Alzheimer's, and Parkinson's disease. 6-MSITC acts on many signaling pathways, such as PPAR, AMPK, PI3K/AKT/mTOR, Nrf2/Keap1-ARE, ERK1/2-ELK1/CHOP/DR5, and MAPK. However, despite the very promising results of in vitro and in vivo animal studies and a few human studies, the molecule has not yet been thoroughly tested in the human population. Nonetheless, wasabi should be classified as a "superfood" for the primary and secondary prevention of human diseases. This article reviews the current state-of-the-art research on 6-MSITC and its potential clinical uses, discussing in detail the signaling pathways activated by the molecule and their interactions.
Collapse
Affiliation(s)
- Joanna Bartkowiak-Wieczorek
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| | - Michał Malesza
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| | - Ida Malesza
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Tomasz Hadada
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| | - Jakub Winkler-Galicki
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| | - Teresa Grzelak
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| | - Edyta Mądry
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| |
Collapse
|
31
|
Low ZY, Yip AJW, Chan AML, Choo WS. 14-3-3 Family of Proteins: Biological Implications, Molecular Interactions, and Potential Intervention in Cancer, Virus and Neurodegeneration Disorders. J Cell Biochem 2024; 125:e30624. [PMID: 38946063 DOI: 10.1002/jcb.30624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024]
Abstract
The 14-3-3 family of proteins are highly conserved acidic eukaryotic proteins (25-32 kDa) abundantly present in the body. Through numerous binding partners, the 14-3-3 is responsible for many essential cellular pathways, such as cell cycle regulation and gene transcription control. Hence, its dysregulation has been linked to the onset of critical illnesses such as cancers, neurodegenerative diseases and viral infections. Interestingly, explorative studies have revealed an inverse correlation of 14-3-3 protein in cancer and neurodegenerative diseases, and the direct manipulation of 14-3-3 by virus to enhance infection capacity has dramatically extended its significance. Of these, COVID-19 has been linked to the 14-3-3 proteins by the interference of the SARS-CoV-2 nucleocapsid (N) protein during virion assembly. Given its predisposition towards multiple essential host signalling pathways, it is vital to understand the holistic interactions between the 14-3-3 protein to unravel its potential therapeutic unit in the future. As such, the general structure and properties of the 14-3-3 family of proteins, as well as their known biological functions and implications in cancer, neurodegeneration, and viruses, were covered in this review. Furthermore, the potential therapeutic target of 14-3-3 proteins in the associated diseases was discussed.
Collapse
Affiliation(s)
- Zheng Yao Low
- School of Science, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Ashley Jia Wen Yip
- School of Science, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Alvin Man Lung Chan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Wee Sim Choo
- School of Science, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
32
|
Hamwi MN, Elsayed E, Dabash H, Abuawad A, Aweer NA, Al Zeir F, Pedersen S, Al-Mansoori L, Burgon PG. MLIP and Its Potential Influence on Key Oncogenic Pathways. Cells 2024; 13:1109. [PMID: 38994962 PMCID: PMC11240681 DOI: 10.3390/cells13131109] [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: 01/26/2024] [Revised: 05/27/2024] [Accepted: 06/19/2024] [Indexed: 07/13/2024] Open
Abstract
Muscle-enriched A-type lamin-interacting protein (MLIP) is an emerging protein involved in cellular homeostasis and stress adaptation. Eukaryotic cells regulate various cellular processes, including metabolism, DNA repair, and cell cycle progression, to maintain cellular homeostasis. Disruptions in this homeostasis can lead to diseases such as cancer, characterized by uncontrolled cell growth and division. This review aims to explore for the first time the unique role MLIP may play in cancer development and progression, given its interactions with the PI3K/Akt/mTOR pathway, p53, MAPK9, and FOXO transcription factors, all critical regulators of cellular homeostasis and tumor suppression. We discuss the current understanding of MLIP's involvement in pro-survival pathways and its potential implications in cancer cells' metabolic remodeling and dysregulated homeostasis. Additionally, we examine the potential of MLIP as a novel therapeutic target for cancer treatment. This review aims to shed light on MLIP's potential impact on cancer biology and contribute to developing innovative therapeutic strategies.
Collapse
Affiliation(s)
- Mahmoud N. Hamwi
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar; (M.N.H.); (E.E.); (N.A.A.); (F.A.Z.); (S.P.)
| | - Engy Elsayed
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar; (M.N.H.); (E.E.); (N.A.A.); (F.A.Z.); (S.P.)
| | - Hanan Dabash
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar; (H.D.); (A.A.)
| | - Amani Abuawad
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar; (H.D.); (A.A.)
| | - Noor A. Aweer
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar; (M.N.H.); (E.E.); (N.A.A.); (F.A.Z.); (S.P.)
| | - Faissal Al Zeir
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar; (M.N.H.); (E.E.); (N.A.A.); (F.A.Z.); (S.P.)
| | - Shona Pedersen
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar; (M.N.H.); (E.E.); (N.A.A.); (F.A.Z.); (S.P.)
| | - Layla Al-Mansoori
- Biomedical Research Centre, Qatar University, Doha P.O. Box 2713, Qatar
| | - Patrick G. Burgon
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar; (H.D.); (A.A.)
| |
Collapse
|
33
|
Shang S, Zhang L, Liu K, Lv M, Zhang J, Ju D, Wei D, Sun Z, Wang P, Yuan J, Zhu Z. Landscape of targeted therapies for advanced urothelial carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:641-677. [PMID: 38966172 PMCID: PMC11220318 DOI: 10.37349/etat.2024.00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/06/2024] [Indexed: 07/06/2024] Open
Abstract
Bladder cancer (BC) is the tenth most common malignancy globally. Urothelial carcinoma (UC) is a major type of BC, and advanced UC (aUC) is associated with poor clinical outcomes and limited survival rates. Current options for aUC treatment mainly include chemotherapy and immunotherapy. These options have moderate efficacy and modest impact on overall survival and thus highlight the need for novel therapeutic approaches. aUC patients harbor a high tumor mutation burden and abundant molecular alterations, which are the basis for targeted therapies. Erdafitinib is currently the only Food and Drug Administration (FDA)-approved targeted therapy for aUC. Many potential targeted therapeutics aiming at other molecular alterations are under investigation. This review summarizes the current understanding of molecular alterations associated with aUC targeted therapy. It also comprehensively discusses the related interventions for treatment in clinical research and the potential of using novel targeted drugs in combination therapy.
Collapse
Affiliation(s)
- Shihao Shang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Lei Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Kepu Liu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Maoxin Lv
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming 65000, Yunnan, China
| | - Jie Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
- College of Life Sciences, Northwest University, Xi’an 710068, Shaanxi, China
| | - Dongen Ju
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Di Wei
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Zelong Sun
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Pinxiao Wang
- School of Clinical Medicine, Xi’an Medical University, Xi’an 710021, Shaanxi, China
| | - Jianlin Yuan
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Zheng Zhu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| |
Collapse
|
34
|
Shrestha P, Ghanwatkar Y, Mahto S, Pramanik N, Mahato RI. Gemcitabine-Lipid Conjugate and ONC201 Combination Therapy Effectively Treats Orthotopic Pancreatic Tumor-Bearing Mice. ACS APPLIED MATERIALS & INTERFACES 2024; 16:29686-29698. [PMID: 38813771 PMCID: PMC11600442 DOI: 10.1021/acsami.4c02626] [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] [Indexed: 05/31/2024]
Abstract
Gemcitabine (GEM) is a nucleoside analogue approved as a first line of therapy for pancreatic ductal adenocarcinoma (PDAC). However, rapid metabolism by plasma cytidine deaminase leading to the short half-life, intricate intracellular metabolism, ineffective cell uptake, and swift development of chemoresistance downgrades the clinical efficacy of GEM. ONC201 is a small molecule that inhibits the Akt and ERK pathways and upregulates the TNF-related apoptosis-inducing ligand (TRAIL), which leads to the reversal of both intrinsic and acquired GEM resistance in PDAC treatment. Moreover, the pancreatic cancer cells that were able to bypass apoptosis after treatment of ONC201 get arrested in the G1-phase, which makes them highly sensitive to GEM. To enhance the in vivo stability of GEM, we first synthesized a disulfide bond containing stearate conjugated GEM (lipid-GEM), which makes it sensitive to the redox tumor microenvironment (TME) comprising high glutathione levels. In addition, with the help of colipids 1,2-dioleoyl-glycero-3-phosphocholine (DOPC), cholesterol, and 1,2-distearoyl-glycero-3-phosphoethanolamine-poly(ethylene glycol)-2000 (DSPE-PEG 2000), we were able to synthesize the lipid-GEM conjugate and ONC201 releasing liposomes. A cumulative drug release study confirmed that both ONC201 and GEM showed sustained release from the formulation. Since MUC1 is highly expressed in 70-90% PDAC, we conjugated a MUC1 binding peptide in the liposomes which showed higher cytotoxicity, apoptosis, and cellular internalization by MIA PaCa-2 cells. A biodistribution study further confirmed that the systemic delivery of the liposomes through the tail vein resulted in a higher accumulation of drugs in orthotopic PDAC tumors in NSG mice. The IHC of the excised tumor grafts further confirmed the higher apoptosis and lower metastasis and cell proliferation. Thus, our MUC1 targeting binary drug-releasing liposomal formulation showed higher drug payload, enhanced plasma stability, and accumulation of drugs in the pancreatic orthotopic tumor and thus is a promising therapeutic alternative for the treatment of PDAC.
Collapse
Affiliation(s)
- Prakash Shrestha
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Yashwardhan Ghanwatkar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Sohan Mahto
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Nilkamal Pramanik
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
35
|
Sekihara K, Himuro H, Toda S, Saito N, Hirayama R, Suganuma N, Sasada T, Hoshino D. Recent Trends and Potential of Radiotherapy in the Treatment of Anaplastic Thyroid Cancer. Biomedicines 2024; 12:1286. [PMID: 38927493 PMCID: PMC11201408 DOI: 10.3390/biomedicines12061286] [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: 04/26/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Anaplastic thyroid cancer (ATC) is a rare but highly aggressive malignancy characterized by advanced disease at diagnosis and a poor prognosis. Despite multimodal therapeutic approaches that include surgery, radiotherapy, and chemotherapy, an optimal treatment strategy remains elusive. Current developments in targeted therapies and immunotherapy offer promising avenues for improved outcomes, particularly for BRAF-mutant patients. However, challenges remain regarding overcoming drug resistance and developing effective treatments for BRAF-wild-type tumors. This comprehensive review examines the clinical and biological features of ATC, outlines the current standards of care, and discusses recent developments with a focus on the evolving role of radiotherapy. Moreover, it emphasizes the necessity of a multidisciplinary approach and highlights the urgent need for further research to better understand ATC pathogenesis and identify new therapeutic targets. Collaborative efforts, including large-scale clinical trials, are essential for translating these findings into improved patient outcomes.
Collapse
Affiliation(s)
- Kazumasa Sekihara
- Cancer Biology Division, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (K.S.); (S.T.); (N.S.)
- Biospecimen Center, Kanagawa Cancer Center, Yokohama 2418515, Japan
| | - Hidetomo Himuro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (H.H.); (T.S.)
- Department of Radiation Oncology, Kanagawa Cancer Center, Yokohama 2418515, Japan
| | - Soji Toda
- Cancer Biology Division, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (K.S.); (S.T.); (N.S.)
- Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Yokohama 2320024, Japan
| | - Nao Saito
- Cancer Biology Division, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (K.S.); (S.T.); (N.S.)
- Biospecimen Center, Kanagawa Cancer Center, Yokohama 2418515, Japan
| | - Ryoichi Hirayama
- Department of Charged Particle Therapy Research, QST Hospital, National Institutes for Quantum Science and Technology, Chiba 2638555, Japan;
| | - Nobuyasu Suganuma
- Department of Surgery, Yokohama City University, Yokohama 2360004, Japan;
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (H.H.); (T.S.)
| | - Daisuke Hoshino
- Cancer Biology Division, Kanagawa Cancer Center Research Institute, Yokohama 2418515, Japan; (K.S.); (S.T.); (N.S.)
- Biospecimen Center, Kanagawa Cancer Center, Yokohama 2418515, Japan
| |
Collapse
|
36
|
Bennett JJR, Stern AD, Zhang X, Birtwistle MR, Pandey G. Low-frequency ERK and Akt activity dynamics are predictive of stochastic cell division events. NPJ Syst Biol Appl 2024; 10:65. [PMID: 38834572 PMCID: PMC11150372 DOI: 10.1038/s41540-024-00389-7] [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: 01/29/2024] [Accepted: 05/20/2024] [Indexed: 06/06/2024] Open
Abstract
Understanding the dynamics of intracellular signaling pathways, such as ERK1/2 (ERK) and Akt1/2 (Akt), in the context of cell fate decisions is important for advancing our knowledge of cellular processes and diseases, particularly cancer. While previous studies have established associations between ERK and Akt activities and proliferative cell fate, the heterogeneity of single-cell responses adds complexity to this understanding. This study employed a data-driven approach to address this challenge, developing machine learning models trained on a dataset of growth factor-induced ERK and Akt activity time courses in single cells, to predict cell division events. The most predictive models were developed by applying discrete wavelet transforms (DWTs) to extract low-frequency features from the time courses, followed by using Ensemble Integration, a data integration and predictive modeling framework. The results demonstrated that these models effectively predicted cell division events in MCF10A cells (F-measure=0.524, AUC=0.726). ERK dynamics were found to be more predictive than Akt, but the combination of both measurements further enhanced predictive performance. The ERK model`s performance also generalized to predicting division events in RPE cells, indicating the potential applicability of these models and our data-driven methodology for predicting cell division across different biological contexts. Interpretation of these models suggested that ERK dynamics throughout the cell cycle, rather than immediately after growth factor stimulation, were associated with the likelihood of cell division. Overall, this work contributes insights into the predictive power of intra-cellular signaling dynamics for cell fate decisions, and highlights the potential of machine learning approaches in unraveling complex cellular behaviors.
Collapse
Affiliation(s)
- Jamie J R Bennett
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alan D Stern
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xiang Zhang
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
| | - Marc R Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA.
| | - Gaurav Pandey
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
37
|
Yuan J, Hou B, Guo K, Zhu J, Xiao H. Tumor-derived exosomal hyaluronidase 1 induced M2 macrophage polarization and promoted esophageal cancer progression. Exp Cell Res 2024; 439:113963. [PMID: 38382806 DOI: 10.1016/j.yexcr.2024.113963] [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: 10/25/2023] [Revised: 01/18/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024]
Abstract
The communication between tumor-derived exosomes and macrophages plays an important role in facilitating the progression of tumors. However, the regulatory mechanisms by which exosomes regulate tumor progression in esophageal squamous cell carcinoma (ESCC) have not been fully elucidated. We constructed a coculture system containing an ESCC cell line and macrophages using a Transwell chamber. We isolated exosomes from the conditioned medium of cancer cells, and characterized them with transmission electron microscopy and western blotting and used then to treat macrophages. We used co-immunoprecipitation to evaluate the interaction between hyaluronidase 1 (HYAL1) and Aurora B kinase (AURKB). We evaluated HYAL1 and AURKB expression in tissues and cells with quantitative reverse-transcription polymerase chain reaction (RT-qPCR) and western blotting. We used RT-qPCR, enzyme-linked immunosorbent assay (ELISA) and flow cytometry to detect macrophage polarization. We assessed cell viability, invasion and migration with the cell counting kit-8 (CCK-8), Transwell and wound healing assays. HYAL1 was highly expressed in ESCC tissues and cells and cancer cell-derived exosomes, and exosomes can be delivered to macrophages through the cancer cell-derived exosomes. The exosomes extracted from HYAL1-overexpressed ESCC cells suppressed M1 macrophage polarization and induced M2 macrophage polarization, thereby promoting ESCC cell viability, invasion and migration. HYAL1 silencing in ESCC cells produced the opposite effects on macrophage polarization and cancer cell functions. We found that HYAL1 interacted with AURKB and further activated the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway in macrophages. In conclusion, ESCC-derived exosomes containing HYAL1 facilitate M2 macrophage polarization by targeting AURKB to active the PI3K/AKT signaling pathway, which in turn promotes ESCC progression.
Collapse
Affiliation(s)
- Jinyan Yuan
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China
| | - Bin Hou
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China
| | - Kai Guo
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China
| | - Jianfei Zhu
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China
| | - Haimin Xiao
- Department 1 of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China.
| |
Collapse
|
38
|
Yang F, Yan L, Ji J, Lou Y, Zhu J. HER2 puzzle pieces: Non-Coding RNAs as keys to mechanisms, chemoresistance, and clinical outcomes in Ovarian cancer. Pathol Res Pract 2024; 258:155335. [PMID: 38723327 DOI: 10.1016/j.prp.2024.155335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/23/2024] [Accepted: 04/27/2024] [Indexed: 05/29/2024]
Abstract
Ovarian cancer (OC) presents significant challenges, characterized by limited treatment options and therapy resistance often attributed to dysregulation of the HER2 signaling pathway. Non-coding RNAs (ncRNAs) have emerged as key players in regulating gene expression in OC. This comprehensive review underscores the pivotal role of ncRNAs in modulating HER2 signaling, with a specific focus on their mechanisms, impact on chemoresistance, and prognostic/diagnostic implications. MicroRNAs, long non-coding RNAs, and circular RNAs have been identified as essential regulators in the modulation of the HER2 pathway. By directly targeting key components of the HER2 axis, these ncRNAs influence its activation and downstream signaling cascades. Dysregulated ncRNAs have been closely associated with chemoresistance, leading to treatment failures and disease progression in OC. Furthermore, distinct expression profiles of ncRNAs hold promise as reliable prognostic and diagnostic markers, facilitating personalized treatment strategies and enhancing disease outcome assessments. A comprehensive understanding of how ncRNAs intricately modulate HER2 signaling is imperative for the development of targeted therapies and the improvement of patient outcomes. The integration of ncRNA profiles into clinical practice has the potential to enhance prognostic and diagnostic accuracy in the management of ovarian cancer. Further research efforts are essential to validate the clinical utility of ncRNAs and elucidate their precise roles in the regulation of HER2 signaling. In conclusion, ncRNAs play a crucial role in governing HER2 signaling in ovarian cancer, impacting chemoresistance and providing valuable prognostic and diagnostic insights. The exploration of ncRNA-mediated HER2 modulation offers promising avenues for the development of personalized treatment approaches, ultimately advancing patient care and outcomes in OC.
Collapse
Affiliation(s)
- Fangwei Yang
- Obstetrical Department, Yiwu Central Hospital, Yiwu, Zhejiang 322000, China.
| | - Lixiang Yan
- Obstetrical Department, Yiwu Central Hospital, Yiwu, Zhejiang 322000, China
| | - Junnan Ji
- Obstetrical Department, Yiwu Central Hospital, Yiwu, Zhejiang 322000, China
| | - Yunxia Lou
- Obstetrical Department, Yiwu Central Hospital, Yiwu, Zhejiang 322000, China
| | - Jinlu Zhu
- Obstetrical Department, Yiwu Central Hospital, Yiwu, Zhejiang 322000, China
| |
Collapse
|
39
|
Ahator SD, Hegstad K, Lentz CS, Johannessen M. Deciphering Staphylococcus aureus-host dynamics using dual activity-based protein profiling of ATP-interacting proteins. mSystems 2024; 9:e0017924. [PMID: 38656122 PMCID: PMC11097646 DOI: 10.1128/msystems.00179-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
The utilization of ATP within cells plays a fundamental role in cellular processes that are essential for the regulation of host-pathogen dynamics and the subsequent immune response. This study focuses on ATP-binding proteins to dissect the complex interplay between Staphylococcus aureus and human cells, particularly macrophages (THP-1) and keratinocytes (HaCaT), during an intracellular infection. A snapshot of the various protein activity and function is provided using a desthiobiotin-ATP probe, which targets ATP-interacting proteins. In S. aureus, we observe enrichment in pathways required for nutrient acquisition, biosynthesis and metabolism of amino acids, and energy metabolism when located inside human cells. Additionally, the direct profiling of the protein activity revealed specific adaptations of S. aureus to the keratinocytes and macrophages. Mapping the differentially activated proteins to biochemical pathways in the human cells with intracellular bacteria revealed cell-type-specific adaptations to bacterial challenges where THP-1 cells prioritized immune defenses, autophagic cell death, and inflammation. In contrast, HaCaT cells emphasized barrier integrity and immune activation. We also observe bacterial modulation of host processes and metabolic shifts. These findings offer valuable insights into the dynamics of S. aureus-host cell interactions, shedding light on modulating host immune responses to S. aureus, which could involve developing immunomodulatory therapies. IMPORTANCE This study uses a chemoproteomic approach to target active ATP-interacting proteins and examines the dynamic proteomic interactions between Staphylococcus aureus and human cell lines THP-1 and HaCaT. It uncovers the distinct responses of macrophages and keratinocytes during bacterial infection. S. aureus demonstrated a tailored response to the intracellular environment of each cell type and adaptation during exposure to professional and non-professional phagocytes. It also highlights strategies employed by S. aureus to persist within host cells. This study offers significant insights into the human cell response to S. aureus infection, illuminating the complex proteomic shifts that underlie the defense mechanisms of macrophages and keratinocytes. Notably, the study underscores the nuanced interplay between the host's metabolic reprogramming and immune strategy, suggesting potential therapeutic targets for enhancing host defense and inhibiting bacterial survival. The findings enhance our understanding of host-pathogen interactions and can inform the development of targeted therapies against S. aureus infections.
Collapse
Affiliation(s)
- Stephen Dela Ahator
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Kristin Hegstad
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | - Christian S. Lentz
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Mona Johannessen
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
40
|
Yonesi A, Tomihara K, Takatsuka D, Tachinami H, Yamazaki M, Jadidi ARY, Takaichi M, Imaue S, Fujiwara K, Yamada SI, Tanuma JI, Noguchi M. Rapamycin Induces Phenotypic Alterations in Oral Cancer Cells That May Facilitate Antitumor T Cell Responses. Biomedicines 2024; 12:1078. [PMID: 38791040 PMCID: PMC11117524 DOI: 10.3390/biomedicines12051078] [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: 04/06/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
OBJECTIVES In this study, we investigated the antitumor immunomodulatory effects of rapamycin in oral cancer. STUDY DESIGN We examined the proliferation, apoptosis, and migration of cancer cells and investigated the cell surface expression levels of immune accessory molecules and T cell immune responses in vitro. We investigated the effect of in vivo administration of rapamycin on immune cell distribution and T cell immune responses in oral tumor-bearing mice. RESULTS Rapamycin treatment significantly inhibited OSCC cell proliferation and migration, increased apoptotic cell death, and upregulated cell surface expression of several immune accessory and adhesion molecules, including CD40, CD83, PD-L1, PD-L2, MHC class I, P-selectin, and VCAM-1. These cancer cells augmented T cell proliferation. In vivo rapamycin administration significantly attenuated mouse tumor growth with an increased proportion of immune cells, including CD4+ T cells, CD8+ T cells, and dendritic cells (DCs); decreased the proportion of immune suppressive cells, such as myeloid-derived suppressor cells and regulatory T cells; enhanced DC maturation and upregulated the surface expression of CD40, CD86, and ICAM-1. CONCLUSIONS Our results suggest that the therapeutic effect of mTOR inhibition in oral cancer can cause direct antitumor and immunomodulatory effects.
Collapse
Affiliation(s)
- Amirmoezz Yonesi
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan (A.R.Y.J.); (M.N.)
| | - Kei Tomihara
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Danki Takatsuka
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan (A.R.Y.J.); (M.N.)
| | - Hidetake Tachinami
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan (A.R.Y.J.); (M.N.)
| | - Manabu Yamazaki
- Division of Oral Pathology, Faculty of Dentistry, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan (J.-I.T.)
| | - Amir Reza Younesi Jadidi
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan (A.R.Y.J.); (M.N.)
| | - Mayu Takaichi
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan (A.R.Y.J.); (M.N.)
| | - Shuichi Imaue
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan (A.R.Y.J.); (M.N.)
| | - Kumiko Fujiwara
- Department of Dentistry and Oral Surgery, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Japan;
| | - Shin-Ichi Yamada
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan (A.R.Y.J.); (M.N.)
| | - Jun-Ichi Tanuma
- Division of Oral Pathology, Faculty of Dentistry, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan (J.-I.T.)
| | - Makoto Noguchi
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama 930-0194, Japan (A.R.Y.J.); (M.N.)
| |
Collapse
|
41
|
Boxiang Q, Liping S, Tong Q. Cuscuta chinensis Lam. Flavonoids (CCLF) alleviate the symptoms of sepsis-associated encephalopathy via PI3K/Nrf2 pathway. Behav Brain Res 2024; 465:114887. [PMID: 38499156 DOI: 10.1016/j.bbr.2024.114887] [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: 07/26/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 03/20/2024]
Abstract
Sepsis-associated encephalopathy (SAE) frequently encounters patients who are in intensive care units and ∼70% of patients with severe systemic infection. However, due to the unclear pathological mechanisms of SAE, the desease-modifying drug is still lack. Here, we aimed to explore whether the flavonoid components extracted from CCL (CCLF) seeds possess protective effects on SAE animals, and systematically evaluate the transcriptomic alteration (in the hippocampus) after CCLF treatment on SAE animals employing RNA sequencing. We observed that CCLF improved the brain's learning and memory abilities and the structural integrity of BBB using cecal ligation and puncture (CLP)-induced SAE animal models, evaluated by behavioral test and tissue examination of animals respectively. RNA sequencing results showed that CCLF treatment reverses SAE-induced transcriptomic alteration in the hippocampus. Moreover, CCLF also dramatically relieved inflammatory (such as TNF-α, IL-2, and IL-6) and oxidative (MDA and SOD activity) stresses, and inhibited SAE-induced neuron apoptosis in brain tissues. More importantly, CCLF restored the PI3K/AKT signaling pathway and then induced the Nrf2 nuclear translocation to drive HO-1 expression both in vitro and in vivo. LY294002, an inhibitor of PI3K, obviously blocked CCLF's functions on anti-apoptosis, anti-inflammation, and anti-oxidation in vivo, demonstrating that CCLF achieves its bioactivities in a PI3K/AKT signaling dependent manner. Altogether, CCLF exhibits remarkable neuro-protective function and may be a promising candidate for further clinical trials for SAE treatment.
Collapse
Affiliation(s)
- Qi Boxiang
- Medicine Intensive Care Unit, Nantong University Affiliated Maternal and Child Health Hospital/ Nantong Children Hospital, Jiangsu, 226000, PR China.
| | - Sheng Liping
- Medicine Intensive Care Unit, Xuzhou Children Hospital, Xuzhou, Jiangsu, 221000, PR China
| | - Qian Tong
- Medicine Intensive Care Unit, Xuzhou Children Hospital, Xuzhou, Jiangsu, 221000, PR China
| |
Collapse
|
42
|
Malakar S, Chatterjee S, Das M, Pal DK. Genetic variants in the mTOR pathway with renal cancer risk and subtypes in East Indian population. Urologia 2024; 91:270-275. [PMID: 38205940 DOI: 10.1177/03915603231222081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
INTRODUCTION Renal Cell Carcinoma (RCC), which accounts for 2%-3% of all adult malignant neoplasms with a male-to-female predominance of 1.9 to 1 with typical presentation between 55 and 75 years. The phosphoinositide-3-kinase-protein kinase B/Akt (PI3KPKB/Akt) pathway is a main pathway in control of cell growth. mTOR pathway plays a key role in the pathogenesis of RCC. MATERIAL AND METHODS Its a prospective observational study. Tissue samples were collected and processed and DNA isolation and sequencing was done to see for any association and expression. RESULTS AND ANALYSIS Polymorphism analysis of the sequence of three genes MTOR, AKT1, and PIK3CA done and found an intronic variant of the MTOR gene (rs3737611) and AKT1 gene (rs2498797) to be significantly associated with clear cell Renal Cell Carcinoma tumor samples. DISCUSSION This study will help to understand the pathogenesis better and the information can be used to develop new drugs and personalized treatment strategies that are tailored to an individual's genetic makeup. The study identify individuals who are at heightened risk for developing renal cancer and could benefit from targeted screening or preventative measures. Some sample size and definite geographical sample pool remains the main limitation of the study which may not be externally validate the study results.
Collapse
Affiliation(s)
- Subhajit Malakar
- Department of Urology, IPGME&R and SSKM Hospital, Kolkata, West Bengal, India
| | | | - Madhusudhan Das
- Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Dilip Kumar Pal
- Department of Urology, IPGME&R and SSKM Hospital, Kolkata, West Bengal, India
| |
Collapse
|
43
|
Wang S, He Y, Wang J, Luo E. Re-exploration of immunotherapy targeting EMT of hepatocellular carcinoma: Starting from the NF-κB pathway. Biomed Pharmacother 2024; 174:116566. [PMID: 38631143 DOI: 10.1016/j.biopha.2024.116566] [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: 11/22/2023] [Revised: 01/15/2024] [Accepted: 04/04/2024] [Indexed: 04/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common malignancies worldwide, and its high morbidity and mortality have brought a heavy burden to the global public health system. Due to the concealment of its onset, the limitation of treatment, the acquisition of multi-drug resistance and radiation resistance, the treatment of HCC cannot achieve satisfactory results. Epithelial mesenchymal transformation (EMT) is a key process that induces progression, distant metastasis, and therapeutic resistance to a variety of malignant tumors, including HCC. Therefore, targeting EMT has become a promising tumor immunotherapy method for HCC. The NF-κB pathway is a key regulatory pathway for EMT. Targeting this pathway has shown potential to inhibit HCC infiltration, invasion, distant metastasis, and therapeutic resistance. At present, there are still some controversies about this pathway and new ideas of combined therapy, which need to be further explored. This article reviews the progress of immunotherapy in improving EMT development in HCC cells by exploring the mechanism of regulating EMT.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, PR China
| | - Yan He
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China
| | - Jun Wang
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, PR China
| | - En Luo
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, PR China.
| |
Collapse
|
44
|
Meuten TK, Dean GA, Thamm DH. Review: The PI3K-AKT-mTOR signal transduction pathway in canine cancer. Vet Pathol 2024; 61:339-356. [PMID: 37905509 DOI: 10.1177/03009858231207021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Tumors in dogs and humans share many similar molecular and genetic features, incentivizing a better understanding of canine neoplasms not only for the purpose of treating companion animals, but also to facilitate research of spontaneously developing tumors with similar biologic behavior and treatment approaches in an immunologically competent animal model. Multiple tumor types of both species have similar dysregulation of signal transduction through phosphatidylinositol 3-kinase (PI3K), protein kinase B (PKB; AKT), and mechanistic target of rapamycin (mTOR), collectively known as the PI3K-AKT-mTOR pathway. This review aims to delineate the pertinent aspects of the PI3K-AKT-mTOR signaling pathway in health and in tumor development. It will then present a synopsis of current understanding of PI3K-AKT-mTOR signaling in important canine cancers and advancements in targeted inhibitors of this pathway.
Collapse
|
45
|
Ren X, Deng D, Xiang S, Feng J. Promoter hypomethylated PDZK1 acts as a tumorigenic gene in glioma by interacting with AKT1. Aging (Albany NY) 2024; 16:7174-7187. [PMID: 38669103 PMCID: PMC11087087 DOI: 10.18632/aging.205750] [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: 11/15/2023] [Accepted: 03/25/2024] [Indexed: 04/28/2024]
Abstract
Glioma is the most frequently diagnosed primary brain tumor and typically has a poor prognosis because of malignant proliferation and invasion. It is urgent to elucidate the mechanisms driving glioma tumorigenesis and develop novel treatments to address this deadly disease. Here, we first revealed that PDZK1 is expressed at high levels in gliomas. Promoter hypomethylation may cause high expression of PDZK1 in glioma. Knockdown of PDZK1 inhibits glioma cell proliferation and invasion in vitro. Mechanistically, further investigations revealed that the loss of PDZK1 expression by siRNA inhibited the activation of the AKT/mTOR signaling pathway, leading to cell cycle arrest and apoptosis. Clinically, high expression of PDZK1 predicts a poorer prognosis for glioma patients than low expression of PDZK1. Overall, our study revealed that PDZK1 acts as a novel oncogene in glioma by binding to AKT1 and maintaining the activation of the AKT/mTOR signaling pathway. Thus, PDZK1 may be a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Xing Ren
- Clinical Laboratory Medicine Center, The First Affiliated Hospital of University of South China, Hengyang 421001, Hunan, P.R. China
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, P.R. China
| | - Dan Deng
- Clinical Laboratory Medicine Center, The First Affiliated Hospital of University of South China, Hengyang 421001, Hunan, P.R. China
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, P.R. China
| | - Shasha Xiang
- Clinical Laboratory Medicine Center, The First Affiliated Hospital of University of South China, Hengyang 421001, Hunan, P.R. China
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, P.R. China
| | - Jianbo Feng
- Clinical Laboratory Medicine Center, The First Affiliated Hospital of University of South China, Hengyang 421001, Hunan, P.R. China
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, P.R. China
| |
Collapse
|
46
|
Göttig L, Jummer S, Staehler L, Groitl P, Karimi M, Blanchette P, Kosulin K, Branton PE, Schreiner S. The human adenovirus PI3K-Akt activator E4orf1 is targeted by the tumor suppressor p53. J Virol 2024; 98:e0170123. [PMID: 38451084 PMCID: PMC11019960 DOI: 10.1128/jvi.01701-23] [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: 11/20/2023] [Accepted: 01/13/2024] [Indexed: 03/08/2024] Open
Abstract
Human adenoviruses (HAdV) are classified as DNA tumor viruses due to their potential to mediate oncogenic transformation in non-permissive mammalian cells and certain human stem cells. To achieve transformation, the viral early proteins of the E1 and E4 regions must block apoptosis and activate proliferation: the former predominantly through modulating the cellular tumor suppressor p53 and the latter by activating cellular pro-survival and pro-metabolism protein cascades, such as the phosphoinositide 3-kinase (PI3K-Akt) pathway, which is activated by HAdV E4orf1. Focusing on HAdV-C5, we show that E4orf1 is necessary and sufficient to stimulate Akt activation through phosphorylation in H1299 cells, which is not only hindered but repressed during HAdV-C5 infection with a loss of E4orf1 function in p53-positive A549 cells. Contrary to other research, E4orf1 localized not only in the common, cytoplasmic PI3K-Akt-containing compartment, but also in distinct nuclear aggregates. We identified a novel inhibitory mechanism, where p53 selectively targeted E4orf1 to destabilize it, also stalling E4orf1-dependent Akt phosphorylation. Co-IP and immunofluorescence studies showed that p53 and E4orf1 interact, and since p53 is bound by the HAdV-C5 E3 ubiquitin ligase complex, we also identified E4orf1 as a novel factor interacting with E1B-55K and E4orf6 during infection; overexpression of E4orf1 led to less-efficient E3 ubiquitin ligase-mediated proteasomal degradation of p53. We hypothesize that p53 specifically subverts the pro-survival function of E4orf1-mediated PI3K-Akt activation to protect the cell from metabolic hyper-activation or even transformation.IMPORTANCEHuman adenoviruses (HAdV) are nearly ubiquitous pathogens comprising numerous subtypes that infect various tissues and organs. Among many encoded proteins that facilitate viral replication and subversion of host cellular processes, the viral E4orf1 protein has emerged as an intriguing yet under-investigated player in the complex interplay between the virus and its host. Nonetheless, E4orf1 has gained attention as a metabolism activator and oncogenic agent, while recent research is showing that E4orf1 may play a more important role in modulating the cellular pathways such as phosphoinositide 3-kinase-Akt-mTOR. Our study reveals a novel and general impact of E4orf1 on host mechanisms, providing a novel basis for innovative antiviral strategies in future therapeutic settings. Ongoing investigations of the cellular pathways modulated by HAdV are of great interest, particularly since adenovirus-based vectors actually serve as vaccine or gene vectors. HAdV constitute an ideal model system to analyze the underlying molecular principles of virus-induced tumorigenesis.
Collapse
Affiliation(s)
- Lilian Göttig
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Simone Jummer
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Luisa Staehler
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Peter Groitl
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Maryam Karimi
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Paola Blanchette
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Karin Kosulin
- Molecular Microbiology, Children’s Cancer Research Institute, Vienna, Austria
| | - Philip E. Branton
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Sabrina Schreiner
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (Resolving Infection Susceptibility; EXC 2155), Freiburg, Germany
- Institute of Virology, Medical Center—University of Freiburg, Freiburg, Germany
| |
Collapse
|
47
|
Zhang B, Cheng Y, Jian Q, Xiang S, Xu Q, Wang C, Yang C, Lin J, Zheng C. Sishen Pill and its active phytochemicals in treating inflammatory bowel disease and colon cancer: an overview. Front Pharmacol 2024; 15:1375585. [PMID: 38650627 PMCID: PMC11033398 DOI: 10.3389/fphar.2024.1375585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
Abstract
The incidence of inflammatory bowel disease (IBD) and the associated risk of colon cancer are increasing globally. Traditional Chinese medicine (TCM) treatment has unique advantages. The Sishen Pill, a common Chinese patented drug used to treat abdominal pain and diarrhea, consists mainly of Psoraleae Fructus, Myristicae Semen, Euodiae Fructus, and Schisandra Chinensis. Modern research has confirmed that Sishen Pill and its active secondary metabolites, such as psoralen, myristicin, evodiamine, and schisandrin, can improve intestinal inflammation and exert antitumor pharmacological effects. Common mechanisms in treating IBD and colon cancer mainly include regulating inflammation-related signaling pathways such as nuclear factor-kappa B, mitogen-activated protein kinase, phosphatidylinositol 3-kinase, NOD-like receptor heat protein domain-related protein 3, and wingless-type MMTV integration site family; NF-E2-related factor 2 and hypoxia-inducible factor 1α to inhibit oxidative stress; mitochondrial autophagy and endoplasmic reticulum stress; intestinal immune cell differentiation and function through the Janus kinase/signal transducer and activator of transcription pathway; and improving the gut microbiota and intestinal barrier. Overall, existing evidence suggests the potential of the Sishen pill to improve IBD and suppress inflammation-to-cancer transformation. However, large-scale randomized controlled clinical studies and research on the safety of these clinical applications are urgently required.
Collapse
Affiliation(s)
- Boxun Zhang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yingying Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qin Jian
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sirui Xiang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qi Xu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuchu Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Yang
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junzhi Lin
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Zheng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Sichuan Provincial Engineering Research Center of Innovative Re-development of Famous Classical Formulas, Tianfu TCM Innovation Harbour, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
48
|
Al Assaad M, Michaud O, Semaan A, Sigouros M, Tranquille M, Phan A, Levine MF, Gundem G, Medina-Martínez JS, Papaemmanuil E, Manohar J, Wilkes D, Sboner A, Hoda SAF, Elemento O, Mosquera JM. Whole-Genome Sequencing Analysis of Male Breast Cancer Unveils Novel Structural Events and Potential Therapeutic Targets. Mod Pathol 2024; 37:100452. [PMID: 38369186 DOI: 10.1016/j.modpat.2024.100452] [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: 11/30/2023] [Revised: 01/17/2024] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
The molecular characterization of male breast cancer (MaBC) has received limited attention in research, mostly because of its low incidence rate, accounting for only 0.5% to 1% of all reported cases of breast cancer each year. Managing MaBC presents significant challenges, with most treatment protocols being adapted from those developed for female breast cancer. Utilizing whole-genome sequencing (WGS) and state-of-the-art analyses, the genomic features of 10 MaBC cases (n = 10) were delineated and correlated with clinical and histopathologic characteristics. Using fluorescence in situ hybridization, an additional cohort of 18 patients was interrogated to supplement WGS findings. The genomic landscape of MaBC uncovered significant genetic alterations that could influence diagnosis and treatment. We found common somatic mutations in key driver genes, such as FAT1, GATA3, SMARCA4, and ARID2. Our study also mapped out structural variants that impact cancer-associated genes, such as ARID1A, ESR1, GATA3, NTRK1, and NF1. Using a WGS-based classifier, homologous recombination deficiency (HRD) was identified in 2 cases, both presenting with deleterious variants in BRCA2. Noteworthy was the observation of FGFR1 amplification in 21% of cases. Altogether, we identified at least 1 potential therapeutic target in 8 of the 10 cases, including high tumor mutational burden, FGFR1 amplification, and HRD. Our study is the first WGS characterization of MaBC, which uncovered potentially relevant variants, including structural events in cancer genes, HRD signatures, and germline pathogenic mutations. Our results demonstrate unique genetic markers and potential treatment targets in MaBC, thereby underlining the necessity of tailoring treatment strategies for this understudied patient population. These WGS-based findings add to the growing knowledge of MaBC genomics and highlight the need to expand research on this type of cancer.
Collapse
Affiliation(s)
- Majd Al Assaad
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York
| | - Olivier Michaud
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York; Département de Pathologie, Université Laval, Quebec City, Quebec, Canada
| | - Alissa Semaan
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York
| | - Michael Sigouros
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York
| | - Marvel Tranquille
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York
| | - Andy Phan
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | | | | | | | | | - Jyothi Manohar
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York
| | - David Wilkes
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York
| | - Andrea Sboner
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York
| | - Syed A F Hoda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York; New York Genome Center, New York, New York.
| |
Collapse
|
49
|
Saeinasab M, Atlasi Y, M Matin M. Functional role of lncRNAs in gastrointestinal malignancies: the peculiar case of small nucleolar RNA host gene family. FEBS J 2024; 291:1353-1385. [PMID: 36282516 DOI: 10.1111/febs.16668] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/18/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Long noncoding RNAs (lncRNAs) play crucial roles in normal physiology and are often de-regulated in disease states such as cancer. Recently, a class of lncRNAs referred to as the small nucleolar RNA host gene (SNHG) family have emerged as important players in tumourigenesis. Here, we discuss new findings describing the role of SNHGs in gastrointestinal tumours and summarize the three main functions by which these lncRNAs promote carcinogenesis, namely: competing with endogenous RNAs, modulating protein function, and regulating epigenetic marking. Furthermore, we discuss how SNHGs participate in different hallmarks of cancer, and how this class of lncRNAs may serve as potential biomarkers in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Morvarid Saeinasab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
| | - Yaser Atlasi
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, UK
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Iran
| |
Collapse
|
50
|
Wani AK, Prakash A, Sena S, Akhtar N, Singh R, Chopra C, Ariyanti EE, Mudiana D, Yulia ND, Rahayu F. Unraveling molecular signatures in rare bone tumors and navigating the cancer pathway landscapes for targeted therapeutics. Crit Rev Oncol Hematol 2024; 196:104291. [PMID: 38346462 DOI: 10.1016/j.critrevonc.2024.104291] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/23/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Rare cancers (RCs), which account for over 20% of cancer cases, face significant research and treatment challenges due to their limited prevalence. This results in suboptimal outcomes compared to more common malignancies. Rare bone tumors (RBTs) constitute 5-10% of rare cancer cases and pose unique diagnostic complexities. The therapeutic potential of anti-cancer drugs for RBTs remains largely unexplored. Identifying molecular alterations in cancer-related genes and their associated pathways is essential for precision medicine in RBTs. Small molecule inhibitors and monoclonal antibodies targeting specific RBT-associated proteins show promise. Ongoing clinical trials aim to define RBT biomarkers, subtypes, and optimal treatment contexts, including combination therapies and immunotherapeutic agents. This review addresses the challenges in diagnosing, treating, and studying RBTs, shedding light on the current state of RBT biomarkers, potential therapeutic targets, and promising inhibitors. Rare cancers demand attention and innovative solutions to improve clinical outcomes.
Collapse
Affiliation(s)
- Atif Khurshid Wani
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar 144411, India.
| | - Ajit Prakash
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Saikat Sena
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar 144411, India
| | - Nahid Akhtar
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar 144411, India
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar 144411, India
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar 144411, India
| | - Esti Endah Ariyanti
- Research Center for Applied Botany, National Research and Innovation Agency, Bogor 16911, Indonesia
| | - Deden Mudiana
- Research Center for Ecology and Ethnobiology, National Research and Innovation Agency, Bogor 16911, Indonesia
| | - Nina Dwi Yulia
- Research Center for Applied Botany, National Research and Innovation Agency, Bogor 16911, Indonesia
| | - Farida Rahayu
- Research Center for Genetic Engineering, National Research and Innovation Agency, Bogor 16911, Indonesia
| |
Collapse
|