1
|
Rivera Antonio AM, Padilla Martínez II, Torres-Ramos MA, Rosales-Hernández MC. Myeloperoxidase as a therapeutic target for oxidative damage in Alzheimer's disease. J Enzyme Inhib Med Chem 2025; 40:2456282. [PMID: 39950933 DOI: 10.1080/14756366.2025.2456282] [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: 09/26/2024] [Revised: 12/14/2024] [Accepted: 01/14/2025] [Indexed: 05/09/2025] Open
Abstract
Alzheimer's disease (AD) is a major neurodegenerative disorder more common in older adults. One of the leading AD hypotheses involves the amyloid beta (A) production, it is associated to oxidative stress, neuroinflammation, and neurovascular damage. The interaction of A with the blood vessel wall contributes to the disruption of the blood-brain barrier (BBB), allowing neutrophil infiltration containing the myeloperoxidase enzyme (MPO), which produces hypochlorous acid (HOCl) a potent oxidant. Also, MPO could be released from the microglia cells and interact with the amyloid beta plaques. This review aims to study the role of MPO in the progression of AD, in particular its contribution to oxidative stress and neuroinflammation. Furthermore, to explore the MPO-potential as AD-biomarker to evaluate the therapeutic potential of its inhibitors to mitigate the neurotoxicity. Finally, revise MPO inhibitors that could act as dual inhibitors acting on MPO and acetylcholinesterase and or another target involved in AD.
Collapse
Affiliation(s)
- Astrid Mayleth Rivera Antonio
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Salvador Díaz Mirón s/n, MéxicoCiudad de México, México
| | - Itzia Irene Padilla Martínez
- Laboratorio de Química Supramolecular y Nanociencias, Unidad Profesional Interdisciplinaria de Biotecnología, Instituto Politécnico Nacional, MéxicoCiudad de México, México
| | - Mónica A Torres-Ramos
- Dirección de investigación del Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez. Av, Ciudad de México. C.P, México
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Salvador Díaz Mirón s/n, MéxicoCiudad de México, México
| |
Collapse
|
2
|
Cai XX, Huang YH, Lin YCD, Huang HY, Chen YG, Zhang DP, Zhang T, Liu Y, Zuo HL, Huang HD. A comprehensive review of small molecules, targets, and pathways in ulcerative colitis treatment. Eur J Med Chem 2025; 291:117645. [PMID: 40279769 DOI: 10.1016/j.ejmech.2025.117645] [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: 02/11/2025] [Revised: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
Ulcerative colitis (UC), a chronic inflammatory bowel disease (IBD), poses significant clinical challenges because of its complex pathophysiology, long-term nature, and the limited efficacy of existing treatments. Small-molecule compounds, particularly those that are able to modulate inflammation-related signaling pathways and, in many cases, occur in nature, offer a promising alternative or supplement to conventional therapies. Studies on molecules for UC therapeutics reported in 1394 publications over the past 30 years were collected from the Web of Science (WOS) database. Only studies that verified therapeutic efficacy through animal experiments were included. Through an analysis of the molecular classes, structures, common targets, and pathways using network pharmacology, we identified 14 classes of compounds, 5 direct-target modules, and 3 crucial downstream pathways. Alkaloids, phenylpropanoids, flavonoids, and terpenes (and their derivatives) appeared most frequently and mainly targeted lipid metabolism, oxidative stress, immune regulation, signaling transduction, and cancer-related pathways. Notably, there has been an increasing trend of applying naturally sourced compounds in both preclinical and clinical trials, especially flavonoids, over the last five years. Although progress in UC research has been made, the majority of studies have focused on the overall therapeutic effects and biomarker alterations, with limited emphasis on the direct targets and underlying mechanisms. These findings highlight the need to explore novel small-molecule therapeutic strategies for UC, focusing on clearly defined targets and precise modes of action.
Collapse
Affiliation(s)
- Xiao-Xuan Cai
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Yi-Han Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Yang-Chi-Dung Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Hsi-Yuan Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Yi-Gang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Da-Peng Zhang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Tao Zhang
- R&D center, Better Way (Shanghai) Cosmetics Co., Ltd., Shanghai, 201103, PR China
| | - Yue Liu
- R&D center, Better Way (Shanghai) Cosmetics Co., Ltd., Shanghai, 201103, PR China
| | - Hua-Li Zuo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China.
| | - Hsien-Da Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Department of Endocrinology, Key Laboratory of Endocrinology of National Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China.
| |
Collapse
|
3
|
Song Y, Zhang K, Zhang J, Li Q, Huang N, Ma Y, Hou N, Han F, Kan C, Sun X. Epigenetic regulation of nuclear receptors: Implications for endocrine-related diseases and therapeutic strategies. Genes Dis 2025; 12:101481. [PMID: 40290121 PMCID: PMC12022648 DOI: 10.1016/j.gendis.2024.101481] [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: 05/07/2024] [Revised: 10/20/2024] [Accepted: 11/03/2024] [Indexed: 04/30/2025] Open
Abstract
The expression and function of the receptor are controlled by epigenetic changes, such as DNA methylation, histone modification, and noncoding RNAs. These modifications play a pivotal role in receptor activity and can lead to or exacerbate endocrine-related diseases. This review examines the epigenetic alterations of nuclear receptors and their significant impact on conditions such as diabetes, thyroid disorders, and endocrine-related tumors. It highlights current therapies targeting these epigenetic mechanisms, including gene editing, epigenetic drugs, and various other therapeutic approaches. This review offers fresh insight into the mechanisms of endocrine-associated disorders, highlighting the latest progress in the development of novel epigenetic therapies that can be used to address receptor-endocrine interactions.
Collapse
Affiliation(s)
- Yixin Song
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Qinying Li
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Na Huang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Yujie Ma
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Fang Han
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| |
Collapse
|
4
|
Liu J, Zhang L, Liu L, Wu T, Wang L, Han Q. The potential capacities of FTY720: Novel therapeutic functions, targets, and mechanisms against diseases. Eur J Med Chem 2025; 290:117508. [PMID: 40120496 DOI: 10.1016/j.ejmech.2025.117508] [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: 12/20/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
Fingolimod (FTY720), an antagonist of sphingosine-1-phosphate (S1P), functions by binding to S1P receptors (S1PRs), excluding S1PR2. It received approval from the Food and Drug Administration (FDA) for the treatment of multiple sclerosis (MS) in 2010. As the first non-selective oral agonist for S1PRs, FTY720's diverse and systemic receptor expression often leads to alterations in various signaling pathways and multiple systems, making it a subject of intense research. Recent studies have identified a wide range of novel or potential functions for FTY720 beyond its application in MS. These include effects on the blood-brain barrier (BBB), vascular system, organelles, and cell death, as well as potential applications in organ transplantation, immune disorders, oncological conditions, neurological and psychiatric disorders, viral infections, and hypersensitivity diseases. This paper reviews the novel roles, targets, and mechanisms of FTY720 that hold promise for clinical utility. Additionally, it summarizes FTY720's derivation and development process, the characterization and mechanism of the structure of FTY720-P bound to S1PRs, the clinical safety profile, future challenges, and potential strategies to address them. These insights aim to guide future research and applications of FTY720, maximizing its therapeutic potential.
Collapse
Affiliation(s)
- Juan Liu
- Center of Clinical Laboratory and Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, PR China
| | - Lu Zhang
- Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, PR China
| | - Le Liu
- Center of Clinical Laboratory and Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, PR China
| | - Tianfeng Wu
- Center of Clinical Laboratory and Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, PR China
| | - Lin Wang
- Center of Clinical Laboratory and Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, PR China
| | - Qingzhen Han
- Center of Clinical Laboratory and Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, PR China.
| |
Collapse
|
5
|
Márquez-Mendoza JM, Baranda-Ávila N, Lizano M, Langley E. Micro-RNAs targeting the estrogen receptor alpha involved in endocrine therapy resistance in breast cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167783. [PMID: 40057206 DOI: 10.1016/j.bbadis.2025.167783] [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: 09/13/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
Endocrine therapy resistance (ETR) in breast cancer (BC) is a multicausal phenomenon with diverse alterations in the tumor cell interactome. Within these alterations, non-coding RNAs (ncRNAs) such as micro-RNAs (miRNAs) modulate the expression of tumor suppressor genes and proto-oncogenes, such as the ESR1 gene encoding estrogen receptor alpha (ERα). This work aims to review the effects of miRNAs targeting ERα mRNA and their mechanisms related to ETR in BC. A thorough review of the literature and an in silico study were carried out to elucidate the involvement of each miRNA, thus contributing to the understanding of ETR in BC.
Collapse
Affiliation(s)
- J M Márquez-Mendoza
- Programa de Doctorado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
| | - N Baranda-Ávila
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico
| | - M Lizano
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
| | - E Langley
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico.
| |
Collapse
|
6
|
Han HL, Su JY, Zhao XH, Hou DD, Li YM. Peptide-Based Strategies in PLGA-Enhanced Tumor Therapy. J Pept Sci 2025; 31:e70020. [PMID: 40269479 DOI: 10.1002/psc.70020] [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: 02/06/2025] [Revised: 03/22/2025] [Accepted: 04/04/2025] [Indexed: 04/25/2025]
Abstract
Peptide-based therapeutics have gained attention in cancer treatment because of their good specificity, low toxicity, and ability to modulate immune responses. However, challenges such as enzymatic degradation and poor bioavailability limit their clinical application. Peptide-functionalized poly(lactic-co-glycolic acid) (PLGA) systems have emerged as a transformative platform in cancer therapy that offers unique advantages, including enhanced stability, sustained release, and precise delivery of therapeutic agents. This review highlights the synergistic integration of peptides with PLGA and addresses key challenges of peptide-based therapeutics. The application of peptide-functionalized PLGA systems encompasses a diverse range of strategies for cancer therapy. In chemotherapy, peptides disrupt critical tumor pathways, induce apoptosis, and inhibit angiogenesis, demonstrating their versatility in targeting various aspects of tumor progression. In immunotherapy, peptides act as antigens to stimulate robust immune responses or as immune checkpoint inhibitors to restore T cell activity, overcoming tumor immune evasion. These systems also harness the enhanced permeability and retention effect, facilitating preferential accumulation in tumor tissues while leveraging tumor microenvironment (TME)-responsive mechanisms, such as pH-sensitive or enzyme-triggered drug release, to achieve controlled, localized delivery. Collectively, peptide-functionalized PLGA systems represent a promising, versatile approach for precise cancer therapy that integrates innovative delivery strategies with highly specific, potent therapeutic agents.
Collapse
Affiliation(s)
- Hong-Lin Han
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, China
| | - Jing-Yun Su
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, China
| | - Xiao-Huan Zhao
- SINOPEC key Laboratory of Research and Application of Medical and Hygienic Materials, SINOPEC (Beijing) Research Institute of Chemical Industry co., ltd, Beijing, China
| | - Dan-Dan Hou
- SINOPEC key Laboratory of Research and Application of Medical and Hygienic Materials, SINOPEC (Beijing) Research Institute of Chemical Industry co., ltd, Beijing, China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
- Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
7
|
Mannan A, Mohan M, Singh TG. Revenge unraveling the fortress: Exploring anticancer drug resistance mechanisms in BC for enhanced therapeutic strategies. Crit Rev Oncol Hematol 2025; 210:104707. [PMID: 40122355 DOI: 10.1016/j.critrevonc.2025.104707] [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: 01/22/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Breast cancer (BC) is the most prevalent form of cancer in women worldwide and the main cause of cancer-related fatalities in females. BC can be classified into various types based on where cancer has begun to grow or spread, specific characteristics that influence how cancer behaves, and treatment choices. BC is multifaceted, and due to its diverse nature, the mechanisms involved are complex and have not yet been understood. Overexpression and expression of various factors involved in the functioning of mechanisms lead to abnormal changes, providing an environment supporting cancer cell growth. Understanding BC risk factors and early diagnosis through screening techniques like mammography and diagnostic techniques such as imaging and biopsies has advanced significantly. A wide range of treatment options, including surgery, radiation, chemotherapy, targeted treatments, and hormonal therapies, are now available. Daily advancements are being made in the clinical treatment of BC. Still, BC drug resistance cases remain highly prevalent and are currently one of the biggest problems faced by medical science. To increase response rates and possibly lengthen survival, there is a critical requirement for novel medicines with minimal sensitivity to overcome drug resistance. This review classifies different mechanisms that are involved in the development of BC and workable pharmacological targets and explains how they relate to the development of BC drug resistance. By concentrating on the mechanisms covered in this review, we can have a deep understanding of different mechanisms and learn innovative ways to develop novel therapeutics for the disease to combat medication resistance.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
8
|
Omofuma OO, Rusiecki JA, Petrick JL, Falk RT, Wheeler W, Pfeiffer RM, Camargo MC, Cook MB. Circulating Inflammation Biomarkers and the Risk of Esophageal Adenocarcinoma: A Nested Case-Control Study in the Department of Defense Serum Repository. Cancer Epidemiol Biomarkers Prev 2025; 34:649-657. [PMID: 40079721 PMCID: PMC12048206 DOI: 10.1158/1055-9965.epi-24-1544] [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: 10/18/2024] [Revised: 01/24/2025] [Accepted: 03/10/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND We previously identified associations of esophageal adenocarcinoma risk with four inflammation-related candidate biomarkers: TNF receptor 2 (TNFR2), IL17A, VEGFR3, and resistin. METHODS We aimed to replicate these candidates and discover novel associations with additional proteins. We conducted a nested case-control study of men with prediagnostic biospecimens stored at the US Department of Defense Serum Repository, including 203 incident esophageal adenocarcinoma cases. Controls were matched to cases in a ∼2:1 ratio by date of birth, race, service branch, and blood draw date. Multiplex immunoassays (Olink/Proseek panels) measured 254 proteins detected in ≥10% of all samples. Multivariable-adjusted conditional logistic regression models calculated associations between biomarker quantiles and esophageal adenocarcinoma. P values (<0.05) were used to indicate the statistical significance of candidates, and FDR was applied to the additional proteins. ORs from the current analysis and those from previous studies were combined for the candidate markers using fixed-effects meta-analysis. RESULTS Among the four candidates, the highest category of TNFR2 was associated with significantly increased esophageal adenocarcinoma risk (ORQ4 vs. Q1 = 1.87; 95% confidence interval: 1.02-3.42). In the meta-analysis, associations with esophageal adenocarcinoma were positive for TNFR2 (meta-analyzed ORhighest-vs.-lowest = 2.04; 1.12-2.95) and inverse for IL17A (meta-analyzed ORhighest-vs.-lowest = 0.53; 0.26-0.80). Of the additional 250 proteins, 45 were associated with esophageal adenocarcinoma risk and 6 (monocyte chemotactic protein 3, IL6, TNFR1, hepatocyte growth factor, TFF3, and FURIN) remained significant after FDR correction. CONCLUSIONS We confirmed associations of TNFR2 and IL17A with esophageal adenocarcinoma risk. Additionally, our study expands the range of proteins associated with esophageal adenocarcinoma development. IMPACT This is the largest assessment to discover novel associations of inflammation-related proteins with esophageal adenocarcinoma to date.
Collapse
Affiliation(s)
- Omonefe O. Omofuma
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| | - Jennifer A. Rusiecki
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Jessica L. Petrick
- Slone Epidemiology Center at Boston University, Boston, MA, United States
| | - Roni T. Falk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| | - William Wheeler
- Information Management Services Inc., Rockville, MD, United States
| | - Ruth M. Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| | - M. Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| | - Michael B. Cook
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
9
|
Li Y, Ouyang Q, Chen Z, Zhou D, Li Z, Yang X, Long J, Chen G, Li X, Jia S, Zi H, Qi S, Tang H, Zhang B, Niu Y, Xu A, Tong W, Jia J, Huang J. Novel role of general transcript factor IIH subunit 2 (GTF2H2) in the development and sex disparity of hepatocellular carcinoma. Oncogene 2025; 44:1323-1335. [PMID: 39972070 DOI: 10.1038/s41388-025-03301-7] [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: 07/25/2024] [Revised: 01/13/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
Sex disparity is a hepatocellular carcinoma (HCC) hallmark, demonstrating aggressiveness and mortality more frequently in men than in women. However, the components of its basis remain largely unknown. It was identified in HCC frequent loss of heterozygosity of general transcript factor IIH subunit 2 (GTF2H2), a potential estrogen pathway gene. GTF2H2 functions in nucleotide excision repair (NER) and basal transcription, but the function of GTF2H2 in cancer has not been described in depth. Here, it was identified that GTF2H2 inhibited growth and invasive mobility and induced apoptosis of HCC cells, which was up-regulated by estrogen-dependent estrogen receptor alpha (ERα) signaling. Mechanistically, in vitro estrogen-treated HCC models with GTF2H2 knockdown and over-expression showed estrogen-regulated GTF2H2 promoted NER of HCC genomic DNA and inhibited cell cycle progression, and down-regulated PAM/NF-κB pathway. Xenografted HCC mice models showed higher tumor progression of HCC with low GTF2H2 expression in ovariectomized female mice or male mice, but could be rescued by GTF2H2 over-expression, which was also observed in spontaneous tumor mice models. Clinical association analysis of HCC cases showed GTF2H2 expression was higher in female HCC, with correlation positively with ERα expression. Taken together, the estrogen-regulated GTF2H2 may be involved in the development and sex disparity of HCC by maintaining NER-related genomic stability and affecting PAM/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yanmeng Li
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qin Ouyang
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhibin Chen
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- Central Laboratory, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Donghu Zhou
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhenkun Li
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaoxi Yang
- Clinical Research Center, Beijing Children Hospital, Capital Medical University, Beijing, China
| | - Jiang Long
- Department of Oncology Minimally Invasive Interventional Radiology, Beijing You-an Hospital, Capital Medical University, Beijing, China
| | - Guangyong Chen
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaojin Li
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Siyu Jia
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Huaduan Zi
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Saiping Qi
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hengcheng Tang
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bei Zhang
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yamei Niu
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Science, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Anjian Xu
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Weimin Tong
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Science, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, 100050, China.
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Jian Huang
- Laboratory of Molecular Biology, Beijing Institute of Clinical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
10
|
Rees K, Aicheler R, Butcher L, Dodd A, Geen J, Lynch C, Massey I, Morris K, Tennant B, Webb R. Seasonal variation in the associations between self-reported long-COVID symptoms and IL-6 signalling-related factors (particularly the rs2228145 variant of the IL-6R gene): A clinical study. Cytokine 2025; 189:156884. [PMID: 39987891 DOI: 10.1016/j.cyto.2025.156884] [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: 12/19/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/25/2025]
Abstract
This observational study focused on the impact of Interleukin-6 (IL-6)-related factors (notably the IL-6 receptor (IL-6R) gene's rs2228145 polymorphism) on long-COVID risk in individuals who had previously experienced COVID-19 infection(s). The purpose of the study was to better understand such factors' contribution to long-COVID risk, and thus possibly initiate future strategies for using IL-6-related factors as biomarkers predictive of risk (while also obtaining data that may influence long-COVID management and treatment more generally). DNA and blood samples, plus questionnaire responses regarding long-COVID symptoms (including chronic fatigue and cognitive impairment), were collected from 175 participants who had previously experienced COVID-19 infection(s). Potential associations between self-reported long-COVID symptoms and participants' rs2228145 genotypes (determined using TaqMan-based genotyping assays) and/or their circulating IL-6, sIL-6R and sgp130 levels (determined using ELISA) were evaluated. Univariate-regression analyses demonstrated that odds of exhibiting long-COVID symptoms increased with severity/number of previous COVID-19 infection(s) and with hypertension as a co-morbidity, while vaccination decreased the likelihood of developing long-COVID. While long-COVID sufferers exhibited higher IL-6 signalling activity than healthy control individuals, rs2228145 genotype was not associated with long-COVID odds-ratios in- the entire-study cohort. Following identification of significant seasonal variations within our dataset, the entire-study cohort was stratified depending on when samples/questionnaire responses were obtained. In the resulting 'summer' sub-cohort (but not the 'winter' sub-cohort), the rs2228145 AA genotype was significantly over-represented amongst those exhibiting long-COVID symptoms, and long-COVID odds-ratios were significantly reduced for the CC and AC genotypes. While interpretation is complicated by seasonal variations, these findings may be of medical/biomedical value. Importantly, as IL-6 was higher in long-COVID sufferers than healthy controls, and rs2228145 AA genotype-bearing individuals within our 'summer' sub-cohort were at elevated risk of developing long-COVID, these findings point towards possible future use of IL-6 and/or rs2228145 genotype as biomarkers predictive of long-COVID risk, which may bring advantages regarding management and treatment of long-COVID.
Collapse
Affiliation(s)
- Katie Rees
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Rebecca Aicheler
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Lee Butcher
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Alan Dodd
- Clinical Biochemistry Department, Cwm Taf Morgannwg University Health Board, Merthyr Tydfil CF47 9DT, UK
| | - John Geen
- Clinical Biochemistry Department, Cwm Taf Morgannwg University Health Board, Merthyr Tydfil CF47 9DT, UK
| | - Ceri Lynch
- Intensive Care Unit, Cwm Taf Morgannwg University Health Board, Ynysmaerdy, Pontyclun, CF72 8XR, UK
| | - Isabel Massey
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Keith Morris
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK
| | - Brian Tennant
- Clinical Biochemistry Department, Cwm Taf Morgannwg University Health Board, Merthyr Tydfil CF47 9DT, UK
| | - Richard Webb
- Department of Biomedical Sciences, School of Sport/Health Sciences, Cardiff Metropolitan University, Cardiff CF5 2YB, UK.
| |
Collapse
|
11
|
Chen H, Nong S, Huang H, Liu Z, Ao W, Zhou Z, Xian M, Wang S, Liu P. Renshenjian decoction alleviates diabetes via HIF-1α/SPTLC2 pathway by targeting ceramide synthesis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119906. [PMID: 40316155 DOI: 10.1016/j.jep.2025.119906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Renshenjian decoction (RSJ) is a traditional recipe for treating diabetes in an ancient Chinese medical book, composed of Puerariae lobatae Radix and Ginseng Radix. Despite its traditional use, the mechanism of RSJ in improving diabetes has not been elucidated. AIM OF THE STUDY This research aimed to investigate the effects and underlying mechanisms of RSJ in treating diabetes. MATERIALS AND METHODS Diabetic (T2DM) mice were induced using a high-fat diet coupled with streptozotocin injections. The effect of RSJ was assessed through biochemical and histopathological analyses of blood and liver samples to evaluate liver function and tissue damage in T2DM mice. The mechanism of RSJ in treating diabetes was explored using molecular docking, metabolomics, and molecular biology techniques. Additionally, the role of RSJ was further validated in T2DM mice treated with the HIF-1α stabilizer DMOG. RESULTS RSJ could enhance insulin sensitivity and lower blood glucose and lipids in T2DM mice. These effects may be related to various components in RSJ, including puerarin, ginsenoside Rb1, and ginsenoside Re. Liver metabonomics revealed 57 differential metabolites in the model and RSJ groups, with 22 enriched pathways, predominantly related to sphingolipid metabolism. Furthermore, qPCR and Western blot results demonstrated that RSJ dramatically decreases the expression of HIF-1α and SPTLC2, as well as the ceramide content in the liver of diabetic mice. These results were partially reversed by the DMOG. CONCLUSION RSJ can inhibit the expression of ceramide through the HIF-1α/SPTLC2 pathway, which helps to improve diabetes and diabetes-related dyslipidemia and liver injury.
Collapse
Affiliation(s)
- Huien Chen
- Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Shifei Nong
- Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Hongwei Huang
- Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Zheyuan Liu
- Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Wenxing Ao
- Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Ziyou Zhou
- Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Minghua Xian
- Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Traditional Chinese Medicine Resource Germplasm Bank Management Center, Yunfu, 527300, PR China.
| | - Shumei Wang
- Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Traditional Chinese Medicine Resource Germplasm Bank Management Center, Yunfu, 527300, PR China.
| | - Peiyi Liu
- Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Huangpu People's Hospital of Zhongshan, 528429, PR China.
| |
Collapse
|
12
|
Wu CY, Vadhan A, Wei WY, Yu HS, Hsieh YC, Chen WC, Hsu CC, Liu YH, Hsu WL. LH Induces Murine Hair Loss through TRPC-Medicated Cell Aging Responses: Implications for Female Pattern Hair Loss Pathogenesis. J Invest Dermatol 2025:S0022-202X(25)00453-1. [PMID: 40311867 DOI: 10.1016/j.jid.2025.03.044] [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: 04/28/2024] [Revised: 03/08/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025]
Abstract
Menopause-related hormonal imbalances, particularly the decline in estrogen and the rise in luteinizing hormone (LH), are implicated in female pattern hair loss (FPHL). This study investigated LH's role in FPHL, as its precise function has remained unclear. Our results found that a significant association between elevated LH levels and FPHL. The binding of LH to LH receptor (LHR), activates downstream transient receptor potential canonical channels (TRPCs), which potentially mediate excess Ca2+ signals to initiate cell aging responses. We revealed that LH causes reactive oxygen species (ROS) accumulation, Ca2+ elevation and senescence in vibrissa follicles (VFs) and cell damage via DNA damage response (DDR), senescence, and senescence-associated secretory phenotype (SASP) activation in dermal papilla cells (DPCs). Hair loss in mice was due to LH-induced hair follicle (HF) damage and aging. The involvement of TRPCs in LH-induced pathogenesis was examined by treatment with TRPC inhibitors. Similarly, the balding area of FPHL showed higher levels of LHR compared to the non-balding area, while the expression of DDR-related genes, SASP-related genes and TRPCs were upregulated in scalp biopsies. Overall, we identified the impacts of LH/LHR signaling on the pathogenesis of FPHL, including TRPC-mediated cell aging responses in HFs.
Collapse
Affiliation(s)
- Ching-Ying Wu
- Department of Dermatology, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Anupama Vadhan
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin, Taiwan
| | - Wei-Yen Wei
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsin-Su Yu
- Graduate Institute of Clinical Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Chun Hsieh
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin, Taiwan
| | - Wei-Chiao Chen
- Department of Dermatology, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin-Chia Hsu
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin, Taiwan
| | - Yi-Hsuan Liu
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin, Taiwan
| | - Wen-Li Hsu
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Environmental Science and Engineering, National, Pingtung University of Science and Technology, Pingtung, Taiwan.
| |
Collapse
|
13
|
Ramezani A, Rahnama M, Mahmoudian F, Shirazi F, Ganji M, Bakhshi S, Khalesi B, Hashemi ZS, Khalili S. Current Understanding of the Exosomes and Their Associated Biomolecules in the Glioblastoma Biology, Clinical Treatment, and Diagnosis. J Neuroimmune Pharmacol 2025; 20:48. [PMID: 40299204 DOI: 10.1007/s11481-025-10204-7] [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: 07/23/2024] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
Glioblastoma is the most common and aggressive brain tumor with a low survival rate. Due to its heterogeneous composition, high invasiveness, and frequent recurrence after surgery, treatment success has been limited. In addition, due to the brain's unique immune status and the suppressor tumor microenvironment (TME), glioblastoma treatment has faced more challenges. Exosomes play a critical role in cancer metastasis by regulating cell-cell interactions that promote tumor growth, angiogenesis, metastasis, treatment resistance, and immunological regulation in the tumor microenvironment. This review explores the pivotal role of exosomes in the development of glioblastoma, with a focus on their potential as non-invasive biomarkers for prognosis, early detection and real-time monitoring of disease progression. Notably, exosome-based drug delivery methods hold promise for overcoming the blood-brain barrier (BBB) and developing targeted therapies for glioblastoma. Despite challenges in clinical translation, the potential for personalized exosome = -054321`therapies and the capacity to enhance therapeutic responses in glioblastoma, present intriguing opportunities for improving patient outcomes. It seems that getting a good and current grasp of the role of exosomes in the fight against glioblastoma would properly serve the scientific community to further their understanding of the related potentials of these biological moieties.
Collapse
Affiliation(s)
- Aghdas Ramezani
- Department of Molecular Imaging, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Rahnama
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Mahmoudian
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Shirazi
- Division of Genetics, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Mahmoud Ganji
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shohreh Bakhshi
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Education and Extension Organization, Razi Vaccine and Serum Research Institute, Agricultural Research, Karaj, 3197619751, Iran
| | - Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran.
| |
Collapse
|
14
|
Zhang X, Lin Z, Xu B, Ma C, Jiang B, Geng Y, Sheng Y, Dai Y, Xue Y, Ren Y. Unveiling gastric precancerous stages: metabolomic insights for early detection and intervention. BMC Gastroenterol 2025; 25:318. [PMID: 40301782 PMCID: PMC12042622 DOI: 10.1186/s12876-025-03898-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Gastric precancerous lesions (GPL) represent a heterogeneous, multi-stage process that involves transition from a benign to a malignant state. To optimize prevention and intervention strategies, accurate methods must clearly distinguish between precancerous stages and predict progression risks at early stages. METHODS The metabolomic profiles of 188 GPL tissues and matched normal tissues were characterized using ultra-high-performance liquid chromatography-tandem mass spectrometry. Both multivariate and univariate statistical analyses were used to identify metabolomic features differentiating normal, atrophic, and intestinal metaplasia states in the stomach, followed by preliminary functional validation. RESULTS From experiments conducted on two cohorts, we established a reliable clinical gastric tissue metabolomic map, which clearly distinguished between normal, atrophic, and intestinalized gastric tissues. We then identified metabolic biomarkers that differentiated various GPL stages. Furthermore, key metabolites were validated in in vitro studies. Relative acyl group and glycerophospholipid abundance was higher in normal gastric tissue when compared to GPL, whereas organic acids were more prevalent in precancerous tissues than in normal tissues. A combination of glycerophosphocholine, tiglylcarnitine, malate, sphingosine, and γ-glutamylglutamic acid may serve as powerful biomarkers to distinguish normal tissue from GPL. CONCLUSION We used ultra-high-performance liquid chromatography with tandem mass spectrometry to effectively characterize metabolomic profiles in clinical gastric tissue samples. Key metabolites were identified and validated using targeted metabolomics. This study identified the metabolomic profiles of gastric tissues with atrophy and intestinal metaplasia of the gastric mucosa, uncovering and preliminarily validating key metabolites that may be used to assess high-risk populations and diagnose GPL, potentially advancing targeted gastric cancer prevention and treatment efforts.
Collapse
Affiliation(s)
- Xiaoyue Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Ziming Lin
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Boyan Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Chenyu Ma
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Bowen Jiang
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yan Geng
- School of Life Science and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Yingyue Sheng
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Yuanyuan Dai
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Yuzheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China.
| | - Yilin Ren
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China.
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
15
|
Dawalibi A, Bakir M, Mohammad KS. The genetic architecture of bone metastases: unveiling the role of epigenetic and genetic modifications in drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:19. [PMID: 40342734 PMCID: PMC12059479 DOI: 10.20517/cdr.2025.28] [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: 02/05/2025] [Revised: 03/26/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025]
Abstract
Bone metastases represent frequent and severe complications in various cancers, notably impacting prognosis and quality of life. This review article delves into the genetic and epigenetic mechanisms underpinning drug resistance in bone metastases, a key challenge in effective cancer treatment. The development of drug resistance in cancer can manifest as either intrinsic or acquired, with genetic heterogeneity playing a pivotal role. Intrinsic resistance is often due to pre-existing mutations, while acquired resistance evolves through genetic and epigenetic alterations during treatment. These alterations include mutations in driver genes like TP53 and RB1, epigenetic modifications such as DNA methylation and histone changes, and pathway alterations, notably involving RANK-RANKL signaling and the PI3K/AKT/mTOR cascade. Recent studies underline the significance of the tumor microenvironment in fostering drug resistance, with components such as cancer-associated fibroblasts and hypoxia playing crucial roles. The interactions between metastatic cancer cells and the bone microenvironment facilitate survival and the proliferation of drug-resistant clones. This review highlights the necessity of understanding these complex interactions to develop targeted therapies that can overcome resistance and improve treatment outcomes. Current therapeutic strategies and future directions are discussed, emphasizing the integration of genomic profiling and targeted interventions in managing bone metastases. The evolving landscape of genetic research, including the application of next-generation sequencing and CRISPR technology, offers promising avenues for novel and more effective therapeutic strategies. This comprehensive exploration aims to provide insights into the molecular intricacies of drug resistance in bone metastases, paving the way for improved clinical management and patient care.
Collapse
Affiliation(s)
- Ahmad Dawalibi
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Mohamad Bakir
- Department of Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
16
|
Yang X, Liu Y, Cao J, Wu C, Tang L, Bian W, Chen Y, Yu L, Wu Y, Li S, Shen Y, Xia J, Du J. Targeting epigenetic and post-translational modifications of NRF2: key regulatory factors in disease treatment. Cell Death Discov 2025; 11:189. [PMID: 40258841 PMCID: PMC12012105 DOI: 10.1038/s41420-025-02491-z] [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: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/23/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a key transcription factor involved in regulating cellular antioxidant defense and detoxification mechanisms. It mitigates oxidative stress and xenobiotic-induced damage by inducing the expression of cytoprotective enzymes, including HO-1 and NQO1. NRF2 also modulates inflammatory responses by inhibiting pro-inflammatory genes and mediates cell death pathways, including apoptosis and ferroptosis. Targeting NRF2 offers potential therapeutic avenues for treating various diseases. NRF2 is regulated through two principal mechanisms: post-translational modifications (PTMs) and epigenetic alterations. PTMs, including phosphorylation, ubiquitination, and acetylation, play a pivotal role in modulating NRF2's stability, activity, and subcellular localization, thereby precisely controlling its function in the antioxidant response. For instance, ubiquitination can lead to NRF2 degradation and reduced antioxidant activity, while deubiquitination enhances its stability and function. Epigenetic modifications, such as DNA methylation, histone modifications, and interactions with non-coding RNAs (e.g., MALAT1, PVT1, MIR4435-2HG, and TUG1), are essential for regulating NRF2 expression by modulating chromatin architecture and gene accessibility. This paper systematically summarizes the molecular mechanisms by which PTMs and epigenetic alterations regulate NRF2, and elucidates its critical role in cellular defense and disease. By analyzing the impact of PTMs, such as phosphorylation, ubiquitination, and acetylation, as well as DNA methylation, histone modifications, and non-coding RNA interactions on NRF2 stability, activity, and expression, the study reveals the complex cellular protection network mediated by NRF2. Furthermore, the paper explores how these regulatory mechanisms affect NRF2's roles in oxidative stress, inflammation, and cell death, identifying novel therapeutic targets and strategies. This provides new insights into the treatment of NRF2-related diseases, such as cancer, neurodegenerative disorders, and metabolic syndrome. This research deepens our understanding of NRF2's role in cellular homeostasis and lays the foundation for the development of NRF2-targeted therapies.
Collapse
Affiliation(s)
- Xinyi Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yingchao Liu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jinghao Cao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Cuiyun Wu
- Cancer Center, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lusheng Tang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Wenxia Bian
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuhan Chen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Lingyan Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yunyi Wu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Sainan Li
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuhuan Shen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Jun Xia
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
17
|
Sheng C, Yue R. Investigating metabolic characteristics of type 2 diabetes mellitus-related cognitive dysfunction and correlating therapeutic effects of Di Dang Tang in animal models. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119338. [PMID: 39826792 DOI: 10.1016/j.jep.2025.119338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/24/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Di Dang Tang is a classic formula from Shang Han Lun, originally used to treat conditions such as blood stasis and heat accumulation. It is widely applied in the treatment of diabetes and its complications, but its effects on Type 2 Diabetes Mellitus-related Cognitive Dysfunction (T2DM-CD) remain unclear. AIM OF THE STUDY The study aimed to investigate the metabolic characteristics of patients with T2DM-CD. Additionally, it sought to evaluate the effects of Di Dang Tang on cognitive function in T2DM-CD model rats by targeting the metabolic pathways identified in the clinical analysis, exploring the underlying mechanisms through animal experiments. METHODS Fasting venous serum was collected from patients with Type 2 Diabetes Mellitus (T2DM) to detect metabolism-related products, and KEGG annotation analysis was performed. Separately, thirty rats were randomly divided using a random number table method, with six rats selected as the blank control group. Twenty-four successfully modeled rats were then randomly divided into the model group and three Di Dang Tang groups (low, medium, and high doses). After administering the medication, the relevant indicators in the rats were assessed. RESULTS Clinical metabolomics detected 32 key differential metabolites between the T2DM-CD and the blank control groups. Between the T2DM-CD and T2DM groups, 29 key differential metabolites were identified. In animal experiments, blood glucose levels in the model group were significantly higher compared to the blank control group at the same time points, whereas the high dose groups of Di Dang Tang exhibited reduced blood glucose levels at weeks 6 and 8 relative to the model group. In the Morris water maze test, the model group had longer escape latencies than the blank control group. The medium and high dose groups of Di Dang Tang showed shorter latencies. Additionally, compared to the model group, the Di Dang Tang groups spent more time and covered more distance in the target quadrant but had reduced average proximity and fewer platform entries. HE staining observation of the hippocampal CA1 area showed no apparent pathological changes in the blank group, obvious pathological damage in the model group, and no significant pathological changes in the medium and high dose groups of Di Dang Tang. Compared to the blank control group, the model group showed significant increases in the levels of Arachidonic Acid (AA), Ceramide (Cer), Glutamate (Glu), TNF- α, IL-1β, TG, and LDL-C, and a significant decrease in HDL-C levels. Compared to the model group, the groups of Di Dang Tang significantly modulated the levels of the above indicators. In Western Blot (WB) assays, compared to the blank control group, the model group rats exhibited significantly higher levels of cPLA2, PKC, ERK, and JNK , and significantly lower levels of claudin-5, NMDA, CaMKII, CREB, and BDNF. The Di Dang Tang groups significantly altered the levels of the above indicators compared to the model group. CONCLUSION Amino acid metabolism, sphingolipid signaling pathways, glycerophospholipid metabolism, and various signaling pathways play significant roles in the pathogenesis of T2DM-CD. Di Dang Tang can improve learning and memory abilities in T2DM model rats and ameliorate cognitive impairments, potentially by regulating metabolic levels and inflammatory responses.
Collapse
Affiliation(s)
- Changting Sheng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Rensong Yue
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
18
|
Mielcarska S, Kot A, Kula A, Dawidowicz M, Sobków P, Kłaczka D, Waniczek D, Świętochowska E. B7H3 in Gastrointestinal Tumors: Role in Immune Modulation and Cancer Progression: A Review of the Literature. Cells 2025; 14:530. [PMID: 40214484 PMCID: PMC11988818 DOI: 10.3390/cells14070530] [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: 03/11/2025] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
B7-H3 (CD276), a member of the B7 immune checkpoint family, plays a critical role in modulating immune responses and has emerged as a promising target in cancer therapy. It is highly expressed in various malignancies, where it promotes tumor evasion from T cell surveillance and contributes to cancer progression, metastasis, and therapeutic resistance, showing a correlation with the poor prognosis of patients. Although its receptors were not fully identified, B7-H3 signaling involves key intracellular pathways, including JAK/STAT, NF-κB, PI3K/Akt, and MAPK, driving processes crucial for supporting tumor growth such as cell proliferation, invasion, and apoptosis inhibition. Beyond immune modulation, B7-H3 influences cancer cell metabolism, angiogenesis, and epithelial-to-mesenchymal transition, further exacerbating tumor aggressiveness. The development of B7-H3-targeting therapies, including monoclonal antibodies, antibody-drug conjugates, and CAR-T cells, offers promising avenues for treatment. This review provides an up-to-date summary of the B7H3 mechanisms of action, putative receptors, and ongoing clinical trials evaluating therapies targeting B7H3, focusing on the molecule's role in gastrointestinal tumors.
Collapse
Affiliation(s)
- Sylwia Mielcarska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Anna Kot
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Agnieszka Kula
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Miriam Dawidowicz
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Piotr Sobków
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Daria Kłaczka
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-514 Katowice, Poland; (A.K.); (M.D.); (D.W.)
| | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland; (A.K.); (P.S.); (D.K.)
| |
Collapse
|
19
|
Abbas AH, Haji MR, Shimal AA, Kurmasha YH, Al-Janabi AAH, Azeez ZT, Al-Ali ARS, Al-Najati HMH, Al-Waeli ARA, Abdulhadi NASA, Al-Tuaama AZH, Al-Ashtary MM, Hussin OA. A multidisciplinary review of long COVID to address the challenges in diagnosis and updated management guidelines. Ann Med Surg (Lond) 2025; 87:2105-2117. [PMID: 40212158 PMCID: PMC11981394 DOI: 10.1097/ms9.0000000000003066] [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: 11/22/2024] [Accepted: 02/04/2025] [Indexed: 04/13/2025] Open
Abstract
Long COVID has emerged as a significant challenge since the COVID-19 pandemic, which was declared as an outbreak in March 2020, marked by diverse symptoms and prolonged duration of disease. Defined by the WHO as symptoms persisting or emerging for at least two months post-SARS-CoV-2 infection without an alternative cause, its prevalence varies globally, with estimates of 10-20% in Europe, 7.3% in the USA, and 3.0% in the UK. The condition's etiology remains unclear, involving factors, such as renin-angiotensin system overactivation, persistent viral reservoirs, immune dysregulation, and autoantibodies. Reactivated viruses, like EBV and HSV-6, alongside epigenetic alterations, exacerbate mitochondrial dysfunction and energy imbalance. Emerging evidence links SARS-CoV-2 to chromatin and gut microbiome changes, further influencing long-term health impacts. Diagnosis of long COVID requires detailed systemic evaluation through medical history and physical examination. Management is highly individualized, focusing mainly on the patient's symptoms and affected systems. A multidisciplinary approach is essential, integrating diverse perspectives to address systemic manifestations, underlying mechanisms, and therapeutic strategies. Enhanced understanding of long COVID's pathophysiology and clinical features is critical to improving patient outcomes and quality of life. With a growing number of cases expected globally, advancing research and disseminating knowledge on long COVID remain vital for developing effective diagnostic and management frameworks, ultimately supporting better care for affected individuals.
Collapse
Affiliation(s)
- Abbas Hamza Abbas
- Department of Internal Medicine, Collage of Medicine, University of Basra, Basra, Iraq
| | - Maryam Razzaq Haji
- Department of Internal Medicine, Collage of Medicine, University of Kufa, Najaf, Iraq
| | - Aya Ahmed Shimal
- Department of Internal Medicine, College of Medicine, University of Baghdad, Baghdad, Iraq
| | | | | | - Zainab Tawfeeq Azeez
- Department of Internal Medicine, Al-Zahraa College of Medicine, University of Basra, Basra, Iraq
| | | | | | | | | | | | - Mustafa M. Al-Ashtary
- Department of Internal Medicine, College of Medicine, University of Baghdad, Baghdad, Iraq
| | - Ominat Amir Hussin
- Department of Internal Medicine, Almanhal Academy for Science, Khartoum, Sudan
| |
Collapse
|
20
|
Sulaiman X, Han Y, Liu S, Li K, Shang M, Yang L, White K, Zang Y, Shen J, Wan J. Enrichment of G-to-U Substitution in SARS-CoV-2 Functional Regions and Its Compensation via Concurrent Mutations. J Med Virol 2025; 97:e70353. [PMID: 40249051 PMCID: PMC12007394 DOI: 10.1002/jmv.70353] [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: 02/03/2025] [Revised: 03/17/2025] [Accepted: 04/01/2025] [Indexed: 04/19/2025]
Abstract
We surveyed single nucleotide variant (SNV) patterns from 5 903 647 complete SARS-CoV-2 genomes. Among 10 012 SNVs, APOBEC-mediated C-to-U (C > U) deamination was the most prevalent, followed by G > U and other RNA editing-related substitutions including (A > G, U > C, G > A). However, C > U mutations were less frequent in functional regions, for example, S protein, intrinsic disordered regions, and nonsynonymous mutations, where G > U were over-represented. Notably, G-loss substitutions rarely appeared together. Instead, G-gain mutations tended to more frequently co-occur with others, with a marked preference in the S protein, suggesting a compensatory mechanism for G loss in G > U mutations. The temporal patterns revealed C > U frequency declined until late 2021 then resurged in early 2022. Conversely, G > U steadily decreased, with a pronounced drop in January 2022, coinciding with reduced COVID-19 severity. Vaccinated individuals exhibited a slightly but significantly higher C > U frequency and a notably lower G > U frequency compared to the unvaccinated group. Additionally, cancer patients had higher G > U frequency than general patients during the same period. Interestingly, none of the C > U SNVs were uniquely identified in 2724 environmental samples. These findings suggest novel functional roles of G > U in COVID-19 symptoms, potentially linked to oxidative stress and reactive oxygen species, while C > U remains the dominant substitution, likely driven by host immune-mediated RNA editing.
Collapse
Affiliation(s)
- Xierzhatijiang Sulaiman
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Yan Han
- Department of Biostatistics & Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Sheng Liu
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kailing Li
- Department of BioHealth InformaticsIndiana University School of Informatics and Computing at IUPUIIndianapolisIndianaUSA
| | | | - Lei Yang
- Center for Computational Biology and BioinformaticsIndiana University School of MedicineIndianapolisIndianaUSA
- Herman B Wells Center for Pediatric Research, Department of PediatricsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kenneth White
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Yong Zang
- Department of Biostatistics & Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jikui Shen
- The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jun Wan
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of BioHealth InformaticsIndiana University School of Informatics and Computing at IUPUIIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
21
|
Maines LW, Keller SN, Smith RA, Smith CD. Opaganib Promotes Weight Loss and Suppresses High-Fat Diet-Induced Obesity and Glucose Intolerance. Diabetes Metab Syndr Obes 2025; 18:969-983. [PMID: 40191829 PMCID: PMC11971975 DOI: 10.2147/dmso.s514548] [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: 12/28/2024] [Accepted: 03/22/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Sphingolipid metabolism has been implicated in many diseases including cancer, pathologic inflammation, viral infection, neurologic pathologies and metabolic pathologies, including obesity and diabetes. We have previously shown that opaganib (aka ABC294640) inhibits three key enzymes in the sphingolipid metabolism pathway: sphingosine kinase-2, dihydroceramide desaturase and glucosylceramide synthase. We and others have demonstrated anticancer, anti-inflammatory and antiviral activities of opaganib in multiple experimental models. Furthermore, opaganib has been studied in clinical trials with patients having cancer or severe Covid-19. In the present studies, the effects of opaganib in the well-established model of High-Fat Diet (HFD)-induced obesity have been studied. Methods Male or female C57BL/6 mice were fed Control Diet (CD) or HFD and treated with vehicle or opaganib by oral gavage once daily, 5 days per week. Body weights were monitored and glucose tolerance was measured periodically for up to 16 weeks. In some experiments, obese HFD-fed mice were treated with vehicle, opaganib alone, semaglutide alone or opaganib plus semaglutide. Results Treatment with opaganib markedly suppressed weight gain in male mice fed the HFD but not in mice given the CD. Compared with mice given CD, mice on the HFD demonstrated poor glucose tolerance at 8, 12 and 16 weeks, consistent with the progression of obesity. Importantly, opaganib treatment of the HFD-fed mice abolished this developing glucose intolerance at all times of measurement. Opaganib treatment also reduced the elevation of hemoglobin A1c and the deposition of inguinal fat in HFD-fed mice. Similar results were obtained with female mice, indicating equivalent efficacy of opaganib in both sexes. Additionally, opaganib and semaglutide were equally effective in promoting body weight loss and improving glucose tolerance in obese mice. Opaganib administered either concurrently with semaglutide or as a single drug following cessation of semaglutide treatment eliminated weight rebound. Conclusion Overall, the data indicate that opaganib effectively suppresses the loss of metabolic control in mice on HFD, suggesting that opaganib may be useful alone or in combination with existing therapies for weight management and improve conditions associated with obesity and diabetes.
Collapse
Affiliation(s)
- Lynn W Maines
- Apogee Biotechnology Corporation, Hummelstown, PA, USA
| | | | - Ryan A Smith
- Apogee Biotechnology Corporation, Hummelstown, PA, USA
| | | |
Collapse
|
22
|
Ren W, Zhang H, Li Y, Sun W, Peng H, Guo H, Hou T, Wang M, Hu Z, Wu T, Liu B. Efficacy and safety of PD-1/PD-L1 inhibitors as first-line treatment for esophageal squamous cell carcinoma: a systematic review and meta-analysis. Front Immunol 2025; 16:1563300. [PMID: 40207226 PMCID: PMC11979238 DOI: 10.3389/fimmu.2025.1563300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Purpose This study aims to investigate the efficacy and safety of PD-1/PD-L1 inhibitors in the first-line treatment of esophageal squamous cell carcinoma (ESCC) and identify factors influencing efficacy through a meta-analysis of multiple phase 3 randomized controlled trials (RCTs). Methods A systematic literature search was conducted in Cochrane, PubMed, and Embase databases. Two researchers independently extracted trial data, including efficacy-related outcomes such as overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and duration of response (DoR), along with their subgroup data and safety-related indicators. The overall hazard ratio (HR) and 95% confidence interval (CI) were calculated for OS and PFS, while the overall odds ratio (OR) and 95% CI were computed for ORR to compare the classification and predictive abilities of combined positive score (CPS) and tumor proportion score (TPS) for PD-L1 status. Additionally, survival outcomes across different subgroups were evaluated to explore the potential influencing factors for the efficacy of PD-1/PD-L1 inhibitors in ESCC. Results This meta-analysis included eight phase 3 RCTs encompassing 4,479 participants. PD-1/PD-L1 inhibitors combined with chemotherapy significantly improved OS (HR: 0.68, 95% CI: 0.63-0.74) and PFS (HR: 0.62, 95% CI: 0.58-0.67) in ESCC patients compared to non-combination therapy. Patients with higher PD-L1 expression (CPS>1 or TPS>1) demonstrated superior responses to PD-1/PD-L1 inhibitions, with CPS identified as a stronger predictor of therapeutic benefit, particularly at a threshold of CPS =10. Subgroup analysis revealed that male, Asian, smoking, and liver metastasis patients exhibited a greater trend toward improved disease control with PD-1/PD-L1 inhibitors. However, there was no significant difference in treatment efficacy between immune therapy combined with TP (taxol [paclitaxel] + cisplatin) and FP (5-fluorouracil [5-FU] + cisplatin) regimens (POS =0.51, PPFS =0.11). Finally, PD-1/PD-L1 inhibition was associated with a higher incidence of grade ≥3 adverse events compared to chemotherapy alone (HR: 1.21, 95% CI: 1.07-1.37). Conclusions This study confirms that the combination of PD-1/PD-L1 inhibitors and chemotherapy provides significant clinical benefits in ESCC. CPS =10 serves as a key threshold for predicting treatment response. There is a trend suggesting that male, Asian, smoking, and liver metastasis patients may experience better survival benefits, while no significant difference was observed between TP- and FP-based regimens. Systematic Review Registration https://www.crd.york.ac.uk/prospero, identifier CRD42024536221.
Collapse
Affiliation(s)
- Wei Ren
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Hanyu Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Yixin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Wu Sun
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Hexiang Peng
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Huangda Guo
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Tianjiao Hou
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Mengying Wang
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
| | - Zhendong Hu
- Department of Esophageal Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Tao Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| |
Collapse
|
23
|
Jin S, He Y, Feng C, Yuan J, Guo Y, Guo Z, Wang X. Cellular Discrepancy of Platinum Complexes in Interfering with Mitochondrial DNA. ACS CENTRAL SCIENCE 2025; 11:393-403. [PMID: 40161961 PMCID: PMC11950849 DOI: 10.1021/acscentsci.4c01941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 04/02/2025]
Abstract
Mitochondria are associated with cellular energy metabolism, proliferation, and mode of death. Damage to mitochondrial DNA (mtDNA) greatly affects mitochondrial function by interfering with energy production and the signaling pathway. Monofunctional trinuclear platinum complex MTPC demonstrates different actions on the mtDNA of cancerous and normal cells. It severely impairs the integrity and function of mitochondria in the human lung cancer A549 cells, such as dissipating mitochondrial membrane potential, decreasing the copy number of mtDNA, interfering in nucleoid proteins and polymerase gamma gene, reducing adenosine triphosphate (ATP), and inducing mitophagy, whereas it barely affects the mtDNA of the human kidney 2 (HK-2) cells. Moreover, MTPC promotes the release of mtDNA into the cytosol and stimulates the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, thus showing the potential to trigger antitumor immunity. MTPC displays significant cytotoxicity against A549 cells, while it exhibits weak toxicity toward HK-2 cells, therefore displaying great advantage to overcome the lingering nephrotoxicity of platinum anticancer drugs. Discrepant effects of a metal complex on mitochondria of different cells mean that targeting mitochondria has special significance in cancer therapy.
Collapse
Affiliation(s)
- Suxing Jin
- School
of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, P. R. China
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Yafeng He
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Chenyao Feng
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Jian Yuan
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yan Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Zijian Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xiaoyong Wang
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
24
|
Tian R, Kong J, Zang H, Li S, Liu X, Cheng Y, Ni G, Gong L. Overexpression of KIF2C amplifies tamoxifen resistance and lung metastasis of breast cancer through PLK1/C-Myc pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04040-y. [PMID: 40100379 DOI: 10.1007/s00210-025-04040-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/07/2025] [Indexed: 03/20/2025]
Abstract
We highlighted the importance of KIF2C in the development of resistance to tamoxifen and its role in promoting lung metastasis in breast cancer, as well as the mechanisms that underpin these processes. KIF2C overexpression and knockdown lentiviruses were transfected into MCF-7 and MCF-7/TAM cells. A nude mouse model of MCF-7/TAM tumors and lung metastasis was established. The PLK1 inhibitor BI2536 was used to explore the underlying mechanism. KIF2C is elevated in tamoxifen-resistant breast cancer. KIF2C knockdown MCF7/TAM cells show increased sensitivity to tamoxifen, indicated by fewer cell clones, invasive cells, migration area, and lumen count, as well as a higher rate of cell apoptosis. KIF2C is linked to the PLK1/c-Myc signaling pathway, and BI2536 inhibits its enhancement of tamoxifen resistance. Results from an in situ experiment on breast cancer in mice are consistent with in vitro findings. KIF2C upregulation is linked to greater tamoxifen resistance in breast cancer, facilitating progression and lung metastasis in resistant cases. KIF2C's potential mechanism of action is linked to the PLK1/c-Myc signaling pathway.
Collapse
Affiliation(s)
- Rui Tian
- Department of Breast Surgery, Yantaishan Hospital, No. 10087 Keji Road, Laishan District, Yantai, 264000, Shandong, China
| | - Jilin Kong
- Department of Breast Surgery, Yantaishan Hospital, No. 10087 Keji Road, Laishan District, Yantai, 264000, Shandong, China
| | - Hongyan Zang
- Department of Breast Surgery, Yantaishan Hospital, No. 10087 Keji Road, Laishan District, Yantai, 264000, Shandong, China
| | - Shuyan Li
- Department of Breast Surgery, Yantaishan Hospital, No. 10087 Keji Road, Laishan District, Yantai, 264000, Shandong, China
| | - Xiangjuan Liu
- Department of Breast Surgery, Yantaishan Hospital, No. 10087 Keji Road, Laishan District, Yantai, 264000, Shandong, China
| | - Yan Cheng
- Department of Breast Surgery, Yantaishan Hospital, No. 10087 Keji Road, Laishan District, Yantai, 264000, Shandong, China
| | - Gaofeng Ni
- Department of Breast Surgery, Yantaishan Hospital, No. 10087 Keji Road, Laishan District, Yantai, 264000, Shandong, China
| | - Liguo Gong
- Department of Breast Surgery, Yantaishan Hospital, No. 10087 Keji Road, Laishan District, Yantai, 264000, Shandong, China.
| |
Collapse
|
25
|
Weigel B, Inderyas M, Eaton-Fitch N, Thapaliya K, Marshall-Gradisnik S. Health-related quality of life in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Post COVID-19 Condition: a systematic review. J Transl Med 2025; 23:318. [PMID: 40075382 PMCID: PMC11905571 DOI: 10.1186/s12967-025-06131-z] [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: 10/08/2024] [Accepted: 01/15/2025] [Indexed: 03/14/2025] Open
Abstract
PURPOSE Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) and Post COVID-19 Condition (PCC) are debilitating, chronic multi-systemic illnesses that require multidisciplinary care. However, people with ME/CFS (pwME/CFS) and people with PCC (pwPCC) are often precluded from accessing necessary disability and social support services. These unmet care needs exacerbate the existing illness burdens experienced by pwME/CFS and pwPCC. To deliver appropriate care and optimise health outcomes for pwME/CFS and pwPCC, the development of evidence-based healthcare policies that recognise the disabling impacts of these illnesses must be prioritised. This systematic review summarises the health-related quality of life (HRQoL) of pwME/CFS and pwPCC when compared with healthy controls (HCs) to elucidate the impacts of these illnesses and guide healthcare policy reform. METHODS CINAHL, Embase, MEDLINE, PubMed, PsycINFO and the Web of Science Core Collection were systematically searched from 1st January 2003 to 23rd July 2024. Eligible publications included observational studies capturing quantitative HRQoL data among pwME/CFS or pwPCC when compared with HCs. The use of validated patient-reported outcome measures (PROMs) was mandatory. Eligible studies were also required to employ the most stringent diagnostic criteria currently available, including the Canadian Consensus Criteria or International Consensus Criteria for ME/CFS and the World Health Organization case definition for PCC (PROSPERO ID: CRD42024501309). RESULTS This review captured 16 studies, including eight studies among pwME/CFS, seven studies among pwPCC and one study among both illness cohorts. Most participants were female and middle-aged. All pwPCC had experienced prolonged COVID-19 symptoms for at least three months. When compared with HCs, all HRQoL domains were significantly impaired among pwME/CFS and pwPCC. Both illnesses had a salient impact on physical health, including pain and ability to perform daily and work activities. While direct comparisons between pwME/CFS and pwPCC were limited by inconsistencies in the PROMs employed, comparable impact trends across HRQoL domain scores were observed. CONCLUSION ME/CFS and PCC have similar, profound impacts on HRQoL that warrant access to multidisciplinary disability and social support services. Future research must harmonise HRQoL data collection and prioritise longitudinal investigations among pwME/CFS and pwPCC to characterise PCC subgroups (including those fulfilling ME/CFS criteria) and predictors of prognosis.
Collapse
Affiliation(s)
- Breanna Weigel
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, 1 Parklands Drive, Southport, Gold Coast, QLD, 4222, Australia.
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast, QLD, 4222, Australia.
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, 4222, Australia.
| | - Maira Inderyas
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, 1 Parklands Drive, Southport, Gold Coast, QLD, 4222, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Natalie Eaton-Fitch
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, 1 Parklands Drive, Southport, Gold Coast, QLD, 4222, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Kiran Thapaliya
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, 1 Parklands Drive, Southport, Gold Coast, QLD, 4222, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, 1 Parklands Drive, Southport, Gold Coast, QLD, 4222, Australia
- Consortium Health International for Myalgic Encephalomyelitis, Griffith University, Gold Coast, QLD, 4222, Australia
| |
Collapse
|
26
|
Grothey B, Lyu SI, Quaas A, Simon AG, Jung JO, Schröder W, Bruns CJ, Schiffmann LM, Popp FC, Schmidt T, Knipper K. Proteomic characterization of MET-amplified esophageal adenocarcinomas reveals enrichment of alternative splicing- and androgen signaling-related proteins. Cell Mol Life Sci 2025; 82:112. [PMID: 40074836 PMCID: PMC11904063 DOI: 10.1007/s00018-025-05635-7] [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: 09/10/2024] [Revised: 02/11/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Esophageal adenocarcinomas (EACs) represent an evolving tumor entity with high mortality rates. MET amplification is a recurrent driver in EACs and is associated with decreased patient survival. However, the response to MET inhibitors is limited. Recent studies have identified several mechanisms that lead to resistance against MET inhibitors in different tumor entities. Nonetheless, a characterization of additional vulnerable targets beyond MET has not been conducted in MET-amplified EACs. METHODS In this study, we determined the MET amplification status in a cohort of more than 900 EACs using fluorescence in situ hybridization (FISH) and compared the proteomes of MET-amplified (n = 20) versus non-amplified tumors (n = 39) by mass spectrometry. RESULTS We identified a phenotype, present in almost all MET-amplified tumors, which shows an enrichment of alternative RNA splicing, and androgen receptor signaling proteins, as well as decreased patient survival. Additionally, our analyses revealed a negative correlation between MET expression and patient survival in MET-amplified EACs, indicating biological heterogeneity with clinical relevance despite the presence of MET amplification as the predominant oncogenic driver. Furthermore, quantitative immunohistochemical analysis of the inflammatory tumor microenvironment showed that an increased percentage of M2 macrophages is associated with lower overall survival in MET-amplified EACs. CONCLUSIONS Our results provide valuable insights into possible new therapeutic approaches for MET-amplified EACs for further research.
Collapse
Affiliation(s)
- Bastian Grothey
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Su Ir Lyu
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Adrian Georg Simon
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Jin-On Jung
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Wolfgang Schröder
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Christiane J Bruns
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Lars M Schiffmann
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Felix C Popp
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Thomas Schmidt
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Karl Knipper
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
27
|
Cai M, Lai W, Chen H, Cao D, Zhang B, Wang F, Xian M, Wang S. Puerarin Targets HIF-1α to Modulate Hypoxia-Related Sphingolipid Metabolism in Diabetic Hepatopathy via the SPTLC2/Ceramide Pathway. Pharmaceuticals (Basel) 2025; 18:398. [PMID: 40143173 PMCID: PMC11945571 DOI: 10.3390/ph18030398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Background and Objectives: Diabetic hepatopathy, characterized by hepatic hypoxia and metabolic dysregulation, has a rising global incidence and prevalence, with limited effective treatments. Hepatic hypoxia activates hypoxia-inducible factor-1 alpha (HIF-1α), regulating sphingolipid metabolism and elevating ceramide, a key factor in insulin resistance. Puerarin (Pue), a flavonoid derived from Pueraria lobata, exhibits therapeutic effects in diabetes, but its effects on hypoxia-related hepatic metabolism are unclear. This study investigates Pue's mechanisms in modulating hepatic metabolism, focusing on HIF-1α and sphingolipid metabolism. Methods: Using bioinformatics and molecular docking, HIF-1α was identified as a key target in diabetic liver disease, confirmed via drug affinity responsive target stability. In vitro experiments utilized insulin-resistant HepG2 cells to assess glucose intake and HIF-1α expression. In vivo, type 2 diabetes mellitus (T2DM) was induced in mice using a high-fat diet and streptozotocin injections. Pue administration was evaluated for its effects on fasting blood glucose, oral glucose tolerance, and hepatoprotective effects. Liver metabolomics and qPCR/Western blot analyses were conducted to assess metabolic pathways. Results: Pue increased glucose uptake in HepG2 cells and bound HIF-1α. Pue reduced HIF-1α expression in HepG2 cells, an effect attenuated by the HIF-1α stabilizer DMOG. Pue improved fasting blood glucose, oral glucose tolerance, and hepatoprotective effects in T2DM mice, which DMOG reversed. Metabolomics revealed that Pue modulates sphingolipid metabolism, decreasing ceramide content. qPCR and Western blot results confirmed that Pue dramatically decreases HIF-1α and SPTLC2 expression. Conclusions: Pue improves diabetic hepatopathy by reducing ceramide expression through the HIF-1α/SPTLC2 pathway, offering a novel therapeutic strategy for diabetes management.
Collapse
Affiliation(s)
- Mangui Cai
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wenxi Lai
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huien Chen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dongmin Cao
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Boyan Zhang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Feng Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Minghua Xian
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Traditional Chinese Medicine Resource Germplasm Bank Management Center, Yunfu 527300, China
| | - Shumei Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Traditional Chinese Medicine Resource Germplasm Bank Management Center, Yunfu 527300, China
| |
Collapse
|
28
|
Ji W, Xie X, Bai G, Fan Y, He Y, Zhang L, Zhou H, Li L, Li H. Proteomics Reveals That Vitamin D Deficiency Leads to Immunoglobulin Abnormalities and Immune Dysregulation in Patients with Post-COVID-19 Condition. J Proteome Res 2025; 24:1449-1461. [PMID: 39979118 DOI: 10.1021/acs.jproteome.4c01120] [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: 02/22/2025]
Abstract
Vitamin D (VD) levels are closely related to the occurrence of post-COVID-19 conditions (PCCs), but the specific mechanism is still unclear. Here, a total of 50 PCC patient serum samples were divided into the VD sufficient group (VD ≥ 30 ng/mL), the VD insufficient group (20 ng/mL ≤ VD < 30 ng/mL), and the VD deficiency group (VD < 20 ng/mL) and then subjected to proteomic analysis. We identified 15 differential abundance proteins (DAPs) between the VD sufficient and VD insufficient groups, including 5 immunoglobulin proteins (JCHAJN, IGHV4-28, GHV4-34, IGHM, and IGLV2-11), which were significantly negatively correlated with VD levels in PCC patients. These DAPs were primarily enriched in immune-related pathways such as the TNF signaling pathway and the B cell receptor signaling pathway. Compared with the VD insufficient group, VD deficiency resulted in 4 proteins being upregulated and 8 proteins being downregulated. The declined abundance of IGLV1-47 negatively correlated with serum VD levels. Albumin (ALB) was in the center of the protein-protein interaction (PPI) network map of all DAPs and was negatively correlated with serum VD levels. In conclusion, VD insufficiency/deficiency leads to abnormalities in immunoglobulin heavy, light, and J chains, resulting in PCC syndrome. VD supplementation may be a potential therapeutic strategy to alleviate the symptoms of PCC.
Collapse
Affiliation(s)
- Wenrui Ji
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan 750001, China
- The Second Clinical Medical College of Ningxia Medical University, Yinchuan 75001, China
| | - Xiaomin Xie
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan 750001, China
- The Second Clinical Medical College of Ningxia Medical University, Yinchuan 75001, China
| | - Guirong Bai
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan 750001, China
- The Second Clinical Medical College of Ningxia Medical University, Yinchuan 75001, China
| | - Yalei Fan
- The Second Clinical Medical College of Ningxia Medical University, Yinchuan 75001, China
| | - Yanting He
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan 750001, China
- The Second Clinical Medical College of Ningxia Medical University, Yinchuan 75001, China
| | - Li Zhang
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan 750001, China
- The Second Clinical Medical College of Ningxia Medical University, Yinchuan 75001, China
| | - Haiyan Zhou
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan 750001, China
- The Second Clinical Medical College of Ningxia Medical University, Yinchuan 75001, China
| | - Ling Li
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan 750001, China
- The Second Clinical Medical College of Ningxia Medical University, Yinchuan 75001, China
| | - Huan Li
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan 750001, China
- The Second Clinical Medical College of Ningxia Medical University, Yinchuan 75001, China
| |
Collapse
|
29
|
Yao X, Yuen T, Qingchuan C, Jianjun Z, Yefu L, Shulan S. Melanophilin inhibit the growth and lymph node metastasis of triple negative breast cancer via the NONO-SPHK1-S1P axis. J Transl Med 2025; 23:284. [PMID: 40050909 PMCID: PMC11887221 DOI: 10.1186/s12967-025-06240-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is the most aggressive breast cancer subtype with the worst prognosis, and there are no targeted treatments available. TNBC patients are more likely to develop metastases and relapse than patients with other breast cancer subtypes. Lymph node metastasis is the first sign of metastatic spread. We aimed to characterize the mechanism of lymph node metastasis in TNBC to provide a new strategy for the treatment of TNBC. METHODS Gene Expression Omnibus (GEO) TNBC database was utilized to screen for genes related to N staging. Screening the downstream target of Melanophilin (MLPH) in TNBC through RNA sequencing (RNA seq) analysis. Protein mass spectrometry was utilized to analyze the protein which interacts with MLPH, and RNA binding protein immunoprecipitation and quantitative real-time PCR (RIP qPCR) were utilized to verify the regulation of sphingosine kinase 1 (SPHK1) expression by MLPH through Non-POU domain-containing octamer-binding protein (NONO). Cell functional assays and in vivo models experiments further confirmed the effects of MLPH on proliferation and lymph node metastasis of TNBC through the SPHK1-S1P axis. RESULTS MLPH is downregulated in TNBC and inhibits tumor growth and lymph node metastasis though the MLPH-NONO-SPHK1-S1P pathway. NONO was identified as an essential factor involved in SPHK1 mRNA splicing. MLPH interacts with NONO to inhibit SPHK1 mRNA splicing of SPHK1, which reduces the content of S1P, thereby inhibiting growth and lymph node metastasis in TNBC. CONCLUSIONS This study preliminarily elucidated a mechanism underlying lymph node metastasis in TNBC and identified the role of the MLPH-NONO-SPHK1-S1P axis in regulating proliferation and lymph node metastasis in TNBC. These findings may help design strategies for predicting and treating metastasis in TNBC.
Collapse
Affiliation(s)
- Xing Yao
- Central Laboratory, Cancer Hospital of Dalian University of Technology (Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, 110042, P. R. China
| | - Tan Yuen
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology (Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, 110042, P. R. China
| | - Chen Qingchuan
- Department of Gastric Surgery, School of Medicine, Sichuan Cancer Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhang Jianjun
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology (Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, 110042, P. R. China.
| | - Liu Yefu
- Department of Hepatopancreatobiliary Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110042, P. R. China.
| | - Sun Shulan
- Central Laboratory, Cancer Hospital of Dalian University of Technology (Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, 110042, P. R. China.
| |
Collapse
|
30
|
Taylor OB, DeGroff N, El-Hodiri HM, Gao C, Fischer AJ. Sphingosine-1-phosphate signaling regulates the ability of Müller glia to become neurogenic, proliferating progenitor-like cells. eLife 2025; 13:RP102151. [PMID: 40047533 PMCID: PMC11884796 DOI: 10.7554/elife.102151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025] Open
Abstract
The purpose of these studies is to investigate how Sphingosine-1-phosphate (S1P) signaling regulates glial phenotype, dedifferentiation of Müller glia (MG), reprogramming into proliferating MG-derived progenitor cells (MGPCs), and neuronal differentiation of the progeny of MGPCs in the chick retina. We found that S1P-related genes are highly expressed by retinal neurons and glia, and levels of expression were dynamically regulated following retinal damage. Drug treatments that activate S1P receptor 1 (S1PR1) or increase levels of S1P suppressed the formation of MGPCs. Conversely, treatments that inhibit S1PR1 or decrease levels of S1P stimulated the formation of MGPCs. Inhibition of S1P receptors or S1P synthesis significantly enhanced the neuronal differentiation of the progeny of MGPCs. We report that S1P-related gene expression in MG is modulated by microglia and inhibition of S1P receptors or S1P synthesis partially rescues the loss of MGPC formation in damaged retinas missing microglia. Finally, we show that TGFβ/Smad3 signaling in the resting retina maintains S1PR1 expression in MG. We conclude that the S1P signaling is dynamically regulated in MG and MGPCs in the chick retina, and activation of S1P signaling depends, in part, on signals produced by reactive microglia.
Collapse
Affiliation(s)
- Olivia B Taylor
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
- Neuroscience Graduate Program, The Ohio State UniversityColumbusUnited States
| | - Nicholas DeGroff
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
| | - Heithem M El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
| | - Chengyu Gao
- Campus Chemical Instrument Center, Mass Spectrometry and Proteomics Facility, The Ohio State UniversityColumbusUnited States
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
| |
Collapse
|
31
|
Adilović M. COVID-19 related complications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:259-314. [PMID: 40246346 DOI: 10.1016/bs.pmbts.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The COVID-19 pandemic has significantly impacted global healthcare systems, revealed vulnerabilities and prompted a re-evaluation of medical practices. Acute complications from the virus, including cardiovascular and neurological issues, have underscored the necessity for timely medical interventions. Advances in diagnostic methods and personalized therapies have been pivotal in mitigating severe outcomes. Additionally, Long COVID has emerged as a complex challenge, affecting various body systems and leading to respiratory, cardiovascular, neurological, psychological, and musculoskeletal problems. This broad spectrum of complications highlights the importance of multidisciplinary management approaches that prioritize therapy, rehabilitation, and patient-centered care. Vulnerable populations such as paediatric patients, pregnant women, and immunocompromised individuals face unique risks and complications, necessitating continuous monitoring and tailored management strategies to reduce morbidity and mortality associated with COVID-19.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnička cesta, Sarajevo, Bosnia and Herzegovina.
| |
Collapse
|
32
|
Xie J, Yang Z, Li Z, Zhang T, Chen H, Chen X, Dai Z, Chen T, Hou J. Triple-positive breast cancer: navigating heterogeneity and advancing multimodal therapies for improving patient outcomes. Cancer Cell Int 2025; 25:77. [PMID: 40045297 PMCID: PMC11881339 DOI: 10.1186/s12935-025-03680-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/07/2025] [Indexed: 03/09/2025] Open
Abstract
Triple-positive breast cancer (TPBC), a unique subtype of luminal breast cancer, is characterized by concurrent positivity for estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Owing to the crosstalk between the ER and HER2 signaling pathways, the standard of care and drug resistance of this particular subtype are difficult challenges. Recent research and clinical trials have indicated a shift in the treatment paradigm for TPBC from single-target therapies to multi-pathway, multitarget strategies aiming to comprehensively modulate intricate signaling networks, thereby overcoming resistance and enhancing therapeutic outcomes. Among multiple strategies, triple-drug therapy has emerged as a promising treatment modality, demonstrating potential efficacy in patients with TPBC. Moving forward, there is a critical need to perform in-depth analyses of specific mechanisms of cancer pathogenesis and metastasis, decipher the complex interactions between different genes or proteins, and identify concrete molecular targets, thus paving the way for the development of tailored therapeutic strategies to combat TPBC effectively.
Collapse
Affiliation(s)
- Jie Xie
- GuiZhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Zhihui Yang
- Zunyi Medical University, No.6 Xuefu West Road, Zunyi, 563006, Guizhou Province, China
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Zhuolin Li
- GuiZhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Tianyu Zhang
- Urology Department, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Huan Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Xueru Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Zehua Dai
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Tao Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China
| | - Jing Hou
- Department of Breast Surgery, Guizhou Provincial People's Hospital, NO.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, China.
| |
Collapse
|
33
|
Ghazizadeh Y, Sharifi-Ardani SE, Tajik N, Mirzaei R, Pourahmad J. Exploring the Potential of Mitochondria-Targeted Drug Delivery for Enhanced Breast Cancer Therapy. Int J Breast Cancer 2025; 2025:3013009. [PMID: 40224721 PMCID: PMC11991819 DOI: 10.1155/ijbc/3013009] [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: 10/15/2024] [Accepted: 02/11/2025] [Indexed: 04/15/2025] Open
Abstract
Breast cancer stands as the utmost prevalent malignancy in women, impacting the epithelial tissue of the breast and often displaying resistance to effective treatment due to its diverse molecular and histological features. Current treatment modalities may exhibit decreasing efficacy over time and can lead to disease progression. The mitochondria, a crucial organelle responsible for cellular metabolism and energy supply, stand highly sensitive to both heat and reactive oxygen species, presenting an assuring target for photodynamic and photothermal therapies (PTTs) in cancer cure. The employment of nanodrug carriers for combination deliveries holds promise in addressing challenges related to drug degradation and off-target toxicity. By circumventing the reticuloendothelial system, nanocarriers bolster the drug's bioavailability at the intended site and ensure controlled codelivery of multiple drugs, thereby maintaining the normal pharmacokinetic features and the regular pharmacodynamic characteristics of different therapeutic mechanisms. The precision and efficacy of this innovative technology have revolutionized drug delivery, substantially enhancing treatment effectiveness. In the pursuit of targeting mitochondrial modifications in cancer cells, various combination therapies such as photodynamic therapy (PDT), PTT, and chemodynamic therapy (CDT) have been explored. These therapies have improved the efficiency of mitochondria-targeted cancer treatment due to their advantageous properties of minimal toxicity, noninvasiveness, reduced drug resistance, and a safer profile. Our review article provides an exhaustive overview of alterations in the mitochondrial environment in BC, their impact on BC development, potential mitochondrial targets for BC treatment, nanotherapeutic approaches for targeting mitochondria, and the limitations of these approaches.
Collapse
Affiliation(s)
- Yalda Ghazizadeh
- Student Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Negin Tajik
- Student Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Mirzaei
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalal Pourahmad
- Department of Toxicology and Pharmacology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Shakerian N, Tafazoli A, Razavinia A, Sadrzadeh Aghajani Z, Bana N, Mard-Soltani M, Khalesi B, Hashemi ZS, Khalili S. Current Understanding of Therapeutic and Diagnostic Applications of Exosomes in Pancreatic Cancer. Pancreas 2025; 54:e255-e267. [PMID: 39661050 DOI: 10.1097/mpa.0000000000002414] [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] [Indexed: 12/12/2024]
Abstract
ABSTRACT Unusual symptoms, rapid progression, lack of reliable early diagnostic biomarkers, and lack of efficient treatment choices are the ongoing challenges of pancreatic cancer. Numerous research studies have demonstrated the correlation between exosomes and various aspects of pancreatic cancer. In light of these facts, exosomes possess the potential to play functional roles in the treatment, prognosis, and diagnosis of the pancreatic cancer. In the present study, we reviewed the most recent developments in approaches for exosome separation, modification, monitoring, and communication. Moreover, we discussed the clinical uses of exosomes as less invasive liquid biopsies and drug carriers and their contribution to the control of angiogenic activity of pancreatic cancer. Better investigation of exosome biology would help to effectively engineer therapeutic exosomes with certain nucleic acids, proteins, and even exogenous drugs as their cargo. Circulating exosomes have shown promise as reliable candidates for pancreatic cancer early diagnosis and monitoring in high-risk people without clinical cancer manifestation. Although we have tried to reflect the status of exosome applications in the treatment and detection of pancreatic cancer, it is evident that further studies and clinical trials are required before exosomes may be employed as a routine therapeutic and diagnostic tools for pancreatic cancer.
Collapse
Affiliation(s)
- Neda Shakerian
- From the Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful
| | - Aida Tafazoli
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz
| | - Amir Razavinia
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, IR
| | | | - Nikoo Bana
- Kish International Campus, University of Teheran
| | - Maysam Mard-Soltani
- From the Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj
| | - Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| |
Collapse
|
35
|
Seelemann ER, Panchakshari S, Labana PK, Wolverton MM, Deng Y, Abdelwahab H, Consmueller C, Stewart DJ, Chaudhary KR. Sexual dimorphism in right ventricular adaptation to pressure overload involves differential angiogenic response. Am J Physiol Heart Circ Physiol 2025; 328:H496-H508. [PMID: 39873638 DOI: 10.1152/ajpheart.00549.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 08/29/2024] [Accepted: 01/09/2025] [Indexed: 01/30/2025]
Abstract
This study investigated the sexual dimorphism in right ventricle (RV) remodeling in right heart failure susceptible Fischer CDF rats using the pulmonary artery banding (PAB) model. Echocardiography and hemodynamic measurements were performed in adult male and female Fischer CDF rats at 1- or 2-wk post-PAB. RV systolic pressure and RV hypertrophy were significantly elevated in PAB rats compared with sham control at 1- and 2-wk post-PAB; however, no differences were observed between male and female rats. Increase in cardiomyocyte cross-sectional area and RV end-diastolic diameter was observed in male rats compared with female rats at 2-wk post-PAB. Conversely, higher fractional area change and cardiac index were observed in female rats compared with male rats at 2-wk post-PAB. To explore the mechanisms, a focused PCR array was performed and higher expression of angiogenic genes, including sphingosine kinase-1 (Sphk1), was observed in the RV of female rats compared with male rats. Consistent with the higher angiogenic gene expression, female rats had a higher RV vascular density at 2-wk post-PAB compared with male rats. Female RV endothelial cells (RVECs) had better angiogenic ability compared with male cells that was potentiated by estradiol. Furthermore, effect of estradiol on RVECs was inhibited by Sphk1 inhibitor (PF-543). Together, female Fischer CDF rats develop adaptive RV remodeling post-PAB compared with maladaptive remodeling in male rats. Moreover, the adaptive remodeling in female rats is associated with better RV angiogenic response that may result from better angiogenic ability of female RVECs and proangiogenic effects of estradiol through Sphk1.NEW & NOTEWORTHY Female patients with pulmonary hypertension have better right ventricular adaptation compared with male. These sex differences were modeled in right heart failure susceptible Fischer CDF rat using pulmonary artery banding model. Preservation of right ventricular function in female rats is linked to better right ventricular angiogenic response that involves higher intrinsic angiogenic ability of female right ventricular endothelial cells together with the proangiogenic effects of female sex hormone estradiol through sphingosine kinase-1.
Collapse
MESH Headings
- Animals
- Male
- Female
- Ventricular Remodeling
- Rats, Inbred F344
- Neovascularization, Physiologic/drug effects
- Ventricular Function, Right
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/genetics
- Disease Models, Animal
- Rats
- Sex Factors
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/genetics
- Endothelial Cells/metabolism
- Endothelial Cells/drug effects
- Heart Failure/physiopathology
- Heart Failure/metabolism
- Heart Failure/genetics
- Sex Characteristics
- Estradiol/pharmacology
- Pulmonary Artery/surgery
- Pulmonary Artery/physiopathology
- Adaptation, Physiological
- Heart Ventricles/physiopathology
- Ventricular Pressure
Collapse
Affiliation(s)
- Erica R Seelemann
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Sheethal Panchakshari
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Parabhjot Kaur Labana
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Maxwell M Wolverton
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Yupu Deng
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Haya Abdelwahab
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Chris Consmueller
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Duncan J Stewart
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ketul R Chaudhary
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
36
|
Mclaughlin M, Sanal-Hayes NEM, Hayes LD, Berry EC, Sculthorpe NF. People with Long COVID and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Exhibit Similarly Impaired Vascular Function. Am J Med 2025; 138:560-566. [PMID: 37832757 DOI: 10.1016/j.amjmed.2023.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND This study aimed to compare flow-mediated dilation values between individuals with long COVID, individuals with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), and healthy age-matched controls to assess the potential implications for clinical management and long-term health outcomes. METHODS A case-case-control approach was employed, and flow-mediated dilation measurements were obtained from 51 participants (17 long COVID patients, 17 ME/CFS patients, and 17 healthy age-matched controls). Flow-mediated dilation values were analyzed using 1-way analysis of variance for between-group comparisons. RESULTS Results revealed significantly impaired endothelial function in both long COVID and ME/CFS groups compared with healthy age-matched controls as determined by maximum % brachial artery diameter post-occlusion compared with pre-occlusion resting diameter (6.99 ± 4.33% and 6.60 ± 3.48% vs 11.30 ± 4.44%, respectively, both P < .05). Notably, there was no difference in flow-mediated dilation between long COVID and ME/CFS groups (P = .949), despite significantly longer illness duration in the ME/CFS group (ME/CFS: 16 ± 11.15 years vs long COVID: 1.36 ± 0.51 years, P < .0001). CONCLUSION The study demonstrates that both long COVID and ME/CFS patients exhibit similarly impaired endothelial function, indicating potential vascular involvement in the pathogenesis of these post-viral illnesses. The significant reduction in flow-mediated dilation values suggests an increased cardiovascular risk in these populations, warranting careful monitoring and the development of targeted interventions to improve endothelial function and mitigate long-term health implications.
Collapse
Affiliation(s)
- Marie Mclaughlin
- Sport and Physical Activity Research Institute, School of Health and Life Sciences, University of the West of Scotland, Glasgow, United Kingdom; School of Sport, Exercise & Rehabilitation Sciences, Faculty of Health Sciences, University of Hull, United Kingdom.
| | - Nilihan E M Sanal-Hayes
- Sport and Physical Activity Research Institute, School of Health and Life Sciences, University of the West of Scotland, Glasgow, United Kingdom; School of Health and Society, University of Salford, United Kingdom
| | - Lawrence D Hayes
- Sport and Physical Activity Research Institute, School of Health and Life Sciences, University of the West of Scotland, Glasgow, United Kingdom
| | - Ethan C Berry
- Sport and Physical Activity Research Institute, School of Health and Life Sciences, University of the West of Scotland, Glasgow, United Kingdom
| | - Nicholas F Sculthorpe
- Sport and Physical Activity Research Institute, School of Health and Life Sciences, University of the West of Scotland, Glasgow, United Kingdom
| |
Collapse
|
37
|
Miwa K. Oral minocycline therapy as first-line treatment in patients with Myalgic encephalomyelitis and long COVID: A pilot study. eNeurologicalSci 2025; 38:100537. [PMID: 39720104 PMCID: PMC11665601 DOI: 10.1016/j.ensci.2024.100537] [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: 06/06/2024] [Revised: 09/11/2024] [Accepted: 11/13/2024] [Indexed: 12/26/2024] Open
Abstract
Background Myalgic encephalomyelitis (ME) is associated with long COVID and also untoward sequelae after anti-coronavirus vaccination. Recently, oral minocycline therapy has been reported to ameliorate symptoms in patients with ME, particularly at the initial stage of the disease. Methods Oral minocycline (100 mg × 2 on the first day, followed by 100 mg/day for 41 days) was administered to 55 patients with ME that emerged during the "Corona era," including 19 patients with long COVID and 5 patients diagnosed with untoward sequalae following coronavirus vaccination. Results Acute adverse effects including nausea and/or dizziness caused four (7 %) patients to discontinue treatment in the first few days. Among the other 51 patients who completed therapy, favorable effects were observed, including a decrease in performance status score or index for restricted activities of daily living ≥2 points in 41 (80 %) patients. Disease duration was inversely associated with the favorable therapeutic effects (p = 0.02) and the disease duration within 6 months was significantly associated with the favorable therapeutic effects (27/30, 90 %, p = 0.02, hazard ratio: 3.6, 95 % confidence interval, 1.2-10.6). The favorable effects were observed in 16 (89 %) of 18 patients with long COVID. Significant amelioration of subjective symptoms of fatigue, post-exertional malaise, unrefreshing sleep, brain fog, disequilibrium, orthostatic intolerance, and neuropathic pain were observed. Conclusions Oral minocycline was effective at ameliorating symptoms in patients with ME including long COVID and post-coronavirus vaccination sequalae. It represents an effective first-line therapeutic option for these patients in the initial stage.
Collapse
Affiliation(s)
- Kunihisa Miwa
- Department of Internal Medicine, Miwa Naika Clinic, Toyama, Japan
| |
Collapse
|
38
|
Hippee CE, Durnell LA, Kaufman JW, Murray E, Singh BK, Sinn PL. Epithelial-to-mesenchymal transition and live cell extrusion contribute to measles virus release from human airway epithelia. J Virol 2025; 99:e0122024. [PMID: 39791903 PMCID: PMC11852777 DOI: 10.1128/jvi.01220-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025] Open
Abstract
Measles virus (MeV) is a highly contagious respiratory virus transmitted via aerosols. To understand how MeV exits the airways of an infected host, we use unpassaged primary cultures of human airway epithelial cells (HAE). MeV typically remains cell-associated in HAE and forms foci of infection, termed infectious centers, by directly spreading cell-to-cell. We previously described the phenomenon in which infectious centers detach en masse from HAE and remain viable. Here, we investigate the mechanism of this cellular detachment. Via immunostaining, we observed loss of tight junction and cell adhesion proteins within infectious centers. These morphological changes indicate activation of epithelial-to-mesenchymal transition (EMT). EMT can contribute to wound healing in respiratory epithelia by mobilizing nearby cells. Inhibiting TGF-β, and thus EMT, reduced infectious center detachment. Compared with uninfected cells, MeV-infected cells also expressed increased levels of sphingosine kinase 1 (SK1), a regulator of live cell extrusion. Live cell extrusion encourages cells to detach from respiratory epithelia by contracting the actomyosin of neighboring cells. Inhibition or induction of live cell extrusion impacted infectious center detachment rates. Thus, these two related pathways contributed to infectious center detachment in HAE. Detached infectious centers contained high titers of virus that may be protected from the environment, allowing the virus to live on surfaces longer and infect more hosts.IMPORTANCEMeasles virus (MeV) is an extremely contagious respiratory pathogen that continues to cause large, disruptive outbreaks each year. Here, we examine mechanisms of detachment of MeV-infected cells. MeV spreads cell-to-cell in human airway epithelial cells (HAE) to form groups of infected cells, termed "infectious centers". We reported that infectious centers ultimately detach from HAE as a unit, carrying high titers of virus. Viral particles within cells may be more protected from environmental conditions, such as ultraviolet radiation and desiccation. We identified two host pathways, epithelial-to-mesenchymal transition and live cell extrusion, that contribute to infectious center detachment. Perturbing these pathways altered the kinetics of infectious center detachment. These pathways influence one another and contribute to epithelial wound healing, suggesting that infectious center detachment may be a usurped consequence of the host's response to infection that benefits MeV by increasing its transmissibility between hosts.
Collapse
Affiliation(s)
- Camilla E. Hippee
- Microbiology and Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Lorellin A. Durnell
- Microbiology and Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Justin W. Kaufman
- Stead Family Department of Pediatrics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Eileen Murray
- Stead Family Department of Pediatrics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Brajesh K. Singh
- Stead Family Department of Pediatrics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Patrick L. Sinn
- Microbiology and Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
- Stead Family Department of Pediatrics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
39
|
Hill M, Kancheva R, Velíková M, Kančeva L, Včelák J, Ampapa R, Židó M, Štětkářová I, Libertínová J, Vosátková M, Vítků J, Kolátorová L, Škodová T, Kubala Havrdová E. Effect of Treatment on Steroidome in Women with Multiple Sclerosis. Int J Mol Sci 2025; 26:1835. [PMID: 40076462 PMCID: PMC11899614 DOI: 10.3390/ijms26051835] [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: 01/10/2025] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory neurodegenerative disease of the central nervous system. The manifestation of MS is related to steroid changes during the menstrual cycle and pregnancy. As data focusing on the effect of anti-MS drug treatment on steroidome are scarce, we evaluated steroidomic changes (79 steroids) in 61 female MS patients of reproductive age 39 (29, 47) years (median with quartiles) after treatment with anti-MS drugs on the GC-MS/MS platform and immunoassays (cortisol and estradiol). The changes were assessed using steroid levels and steroid molar ratios (SMRs) that may reflect the activities of steroidogenic enzymes (SMRs). A repeated measures ANOVA, followed by multiple comparisons and OPLS models, were used for statistical analyses. The anti-MS treatment decreased steroid levels in the follicular phase. Anti-CD20 monoclonal antibodies (mAb), such as ofatumumab and ocrelizumab; inhibitors of the sphingosine-1-phosphate receptor (S1PRI); and IFNβ-1a decreased circulating 17-hydroxy-pregnanes and shifted the CYP17A1 functioning from the hydroxylase- toward the lyase step. Decreased conjugated/unconjugated steroid ratios were found after treatment with anti-MS drugs, especially for glatiramer acetate and anti-CD20 mAb. In the luteal phase, IFN-β1a treatment increased steroidogenesis; both IFN-β1a and ocrelizumab increased AKR1D1, and S1PRI increased SRD5A functioning. Anti-CD20 mAb reduced the functioning of enzymes catalyzing the synthesis of immunomodulatory 7α/β and 16α-hydroxy-androgens, which may affect the severity of MS. The above findings may be important concerning the alterations in bioactive steroids, such as cortisol; active androgens and estrogens; and neuroactive, neuroprotective, and immunomodulatory steroids in terms of optimization of anti-MS treatment.
Collapse
Affiliation(s)
- Martin Hill
- Institute of Endocrinology, 110 00 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.); (J.V.); (L.K.); (T.Š.)
| | - Radmila Kancheva
- Institute of Endocrinology, 110 00 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.); (J.V.); (L.K.); (T.Š.)
| | - Marta Velíková
- Institute of Endocrinology, 110 00 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.); (J.V.); (L.K.); (T.Š.)
| | - Ludmila Kančeva
- Institute of Endocrinology, 110 00 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.); (J.V.); (L.K.); (T.Š.)
| | - Josef Včelák
- Institute of Endocrinology, 110 00 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.); (J.V.); (L.K.); (T.Š.)
| | - Radek Ampapa
- MS Center, Jihlava Hospital, 586 01 Jihlava, Czech Republic;
| | - Michal Židó
- Department of Neurology 3FM CU and UHKV, Third Faculty of Medicine, Charles University, 100 34 Prague, Czech Republic; (M.Ž.); (I.Š.)
| | - Ivana Štětkářová
- Department of Neurology 3FM CU and UHKV, Third Faculty of Medicine, Charles University, 100 34 Prague, Czech Republic; (M.Ž.); (I.Š.)
| | - Jana Libertínová
- MS Center, Second Faculty of Medicine, Charles University, 150 06 Prague, Czech Republic;
| | - Michala Vosátková
- Institute of Endocrinology, 110 00 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.); (J.V.); (L.K.); (T.Š.)
| | - Jana Vítků
- Institute of Endocrinology, 110 00 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.); (J.V.); (L.K.); (T.Š.)
| | - Lucie Kolátorová
- Institute of Endocrinology, 110 00 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.); (J.V.); (L.K.); (T.Š.)
| | - Tereza Škodová
- Institute of Endocrinology, 110 00 Prague, Czech Republic; (M.V.); (L.K.); (J.V.); (M.V.); (J.V.); (L.K.); (T.Š.)
| | - Eva Kubala Havrdová
- Department of Neurology, First Faculty of Medicine, Charles University, 128 21 Prague, Czech Republic;
| |
Collapse
|
40
|
Huang X, Liu B, Shen S. Lipid Metabolism in Breast Cancer: From Basic Research to Clinical Application. Cancers (Basel) 2025; 17:650. [PMID: 40002245 PMCID: PMC11852908 DOI: 10.3390/cancers17040650] [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: 01/23/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Breast cancer remains the most prevalent cancer among women globally, with significant links to obesity and lipid metabolism abnormalities. This review examines the role of lipid metabolism in breast cancer progression, highlighting its multifaceted contributions to tumor biology. We discuss key metabolic processes, including fatty acid metabolism, triglyceride metabolism, phospholipid metabolism, and cholesterol metabolism, detailing the reprogramming that occurs in these pathways within breast cancer cells. Alterations in lipid metabolism are emphasized for their roles in supporting energy production, membrane biogenesis, and tumor aggressiveness. Furthermore, we examine how lipid metabolism influences immune responses in the tumor microenvironment, affecting immune cell function and therapeutic efficacy. The potential of lipid metabolism as a target for novel therapeutic strategies is also addressed, with a focus on inhibitors of key metabolic enzymes. By integrating lipid metabolism with breast cancer research, this review underscores the importance of lipid metabolism in understanding breast cancer biology and developing treatment approaches aimed at improving patient outcomes.
Collapse
Affiliation(s)
- Xiangyu Huang
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China; (X.H.); (B.L.)
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bowen Liu
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China; (X.H.); (B.L.)
| | - Songjie Shen
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China; (X.H.); (B.L.)
- Ambulatory Medical Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China
| |
Collapse
|
41
|
Vona R, Cittadini C, Ortona E, Matarrese P. Sex Disparity in Cancer: Role of Autophagy and Estrogen Receptors. Cells 2025; 14:273. [PMID: 39996745 PMCID: PMC11854201 DOI: 10.3390/cells14040273] [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: 12/12/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Autophagy, a cellular process essential for maintaining homeostasis, plays a fundamental role in recycling damaged components and in adapting to stress. The dysregulation of autophagy is implicated in numerous human diseases, including cancer, where it exhibits a dual role as both a suppressor and a promoter, depending on the context and the stage of tumor development. The significant sex differences observed in autophagic processes are determined by biological factors, such as genetic makeup and sex hormones. Estrogens, through their interaction with specific receptors, modulate autophagy and influence tumor progression, therapy resistance, and the immune response to tumors. In females, the escape from X inactivation and estrogen signaling may be responsible for the advantages, in terms of lower incidence and longer survival, observed in oncology. Women often show better responses to traditional chemotherapy, while men respond better to immunotherapy. The action of sex hormones on the immune system could contribute to these differences. However, women experience more severe adverse reactions to anticancer drugs. The estrogen/autophagy crosstalk-involved in multiple aspects of the tumor, i.e., development, progression and the response to therapy-deserves an in-depth study, as it could highlight sex-specific mechanisms useful for designing innovative and gender-tailored treatments from the perspective of precision medicine.
Collapse
Affiliation(s)
- Rosa Vona
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy; (C.C.); (E.O.)
| | | | | | - Paola Matarrese
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy; (C.C.); (E.O.)
| |
Collapse
|
42
|
Dye JA, Stewart EJ, Schladweiler MC, Nguyen HH, Grindstaff RD, Padgett WT, Fisher AA, Miller CN. Maternal Exposure to Ozone During Implantation Promotes a Feminized Transcriptomic Profile in the Male Adolescent Liver. Endocrinology 2025; 166:bqaf018. [PMID: 39865885 DOI: 10.1210/endocr/bqaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Maternal exposure to ozone during implantation results in reduced fetal weight gain in rats. Offspring from ozone-exposed dams demonstrate sexually dimorphic risks to high-fat diet feeding in adolescence. To better understand the adolescent hepatic metabolic landscape following fetal growth restriction, RNA sequencing was performed to characterize the effects of ozone-induced fetal growth restriction on male and female offspring. Pregnant Long-Evans rats were exposed to filtered air or 0.8 ppm ozone for 4 hours on both gestation days 5 and 6 (n = 6/group). At approximately postnatal day 48, liver tissue was obtained for RNA sequencing from offspring. Peri-implantation exposure to ozone in the dam had greater effects on hepatic gene expression in male offspring than in the females. Interestingly, heatmaps of these differentially expressed genes suggested that male offspring from ozone-exposed dams had a transcriptomic pattern like that of female offspring. Using a filtered set of highly female-predominant genes (n = 390), 57% were upregulated in the male offspring from ozone-exposed dams. Upregulated canonical pathways included sirtuin and orexin signaling, estrogen receptor signaling, and integration of energy metabolism. Relatively few genes altered in the male offspring from ozone-exposed dams were associated with endpoints of sexual maturity, signifying the likely source of the observed feminization was not attributed to sex hormones. This study provides initial evidence that growth restriction in utero may increase the risk of hepatic feminization in male offspring. Additional work is needed to further understand the relationship between developmental undernutrition and feminization in the male liver.
Collapse
Affiliation(s)
- Janice A Dye
- Cardiopulmonary Immunotoxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Erica J Stewart
- Oak Ridge Institute for Science and Education, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Mette C Schladweiler
- Cardiopulmonary Immunotoxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Helen H Nguyen
- Oak Ridge Institute for Science and Education, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Rachel D Grindstaff
- Neuroendocrine Toxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - William T Padgett
- Neuroendocrine Toxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Anna A Fisher
- Cardiopulmonary Immunotoxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Colette N Miller
- Cardiopulmonary Immunotoxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| |
Collapse
|
43
|
Alalhareth IS, Alyami SM, Alshareef AH, Ajeibi AO, Al Munjem MF, Elfifi AA, Alsharif MM, Alzahrani SA, Alqaad MA, Bakir MB, Abdel-Wahab BA. Cellular Epigenetic Targets and Epidrugs in Breast Cancer Therapy: Mechanisms, Challenges, and Future Perspectives. Pharmaceuticals (Basel) 2025; 18:207. [PMID: 40006021 PMCID: PMC11858621 DOI: 10.3390/ph18020207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/31/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Breast cancer is the most common malignancy affecting women, manifesting as a heterogeneous disease with diverse molecular characteristics and clinical presentations. Recent studies have elucidated the role of epigenetic modifications in the pathogenesis of breast cancer, including drug resistance and efflux characteristics, offering potential new diagnostic and prognostic markers, treatment efficacy predictors, and therapeutic agents. Key modifications include DNA cytosine methylation and the covalent modification of histone proteins. Unlike genetic mutations, reprogramming the epigenetic landscape of the cancer epigenome is a promising targeted therapy for the treatment and reversal of drug resistance. Epidrugs, which target DNA methylation and histone modifications, can provide novel options for the treatment of breast cancer by reversing the acquired resistance to treatment. Currently, the most promising approach involves combination therapies consisting of epidrugs with immune checkpoint inhibitors. This review examines the aberrant epigenetic regulation of breast cancer initiation and progression, focusing on modifications related to estrogen signaling, drug resistance, cancer progression, and the epithelial-mesenchymal transition (EMT). It examines existing epigenetic drugs for treating breast cancer, including agents that modify DNA, inhibitors of histone acetyltransferases, histone deacetylases, histone methyltransferases, and histone demethyltransferases. It also delves into ongoing studies on combining epidrugs with other therapies and addresses the upcoming obstacles in this field.
Collapse
Affiliation(s)
- Ibrahim S. Alalhareth
- College of Pharmacy, Najran University, Najran 66256, Saudi Arabia; (I.S.A.); (S.M.A.)
| | - Saleh M. Alyami
- College of Pharmacy, Najran University, Najran 66256, Saudi Arabia; (I.S.A.); (S.M.A.)
| | - Ali H. Alshareef
- Department of Pharmaceuticals Care, Ministry of Defense, Najran 66281, Saudi Arabia; (A.H.A.); (A.O.A.); (A.A.E.); (M.M.A.)
| | - Ahmed O. Ajeibi
- Department of Pharmaceuticals Care, Ministry of Defense, Najran 66281, Saudi Arabia; (A.H.A.); (A.O.A.); (A.A.E.); (M.M.A.)
| | - Manea F. Al Munjem
- King Khaled Hospital -Najran Health Cluster, Najran 66261, Saudi Arabia;
| | - Ahmad A. Elfifi
- Department of Pharmaceuticals Care, Ministry of Defense, Najran 66281, Saudi Arabia; (A.H.A.); (A.O.A.); (A.A.E.); (M.M.A.)
| | - Meshal M. Alsharif
- Department of Pharmaceuticals Care, Ministry of Defense, Najran 66281, Saudi Arabia; (A.H.A.); (A.O.A.); (A.A.E.); (M.M.A.)
| | - Seham A. Alzahrani
- Pharmacy Department, Khamis Mushait General Hospital, King Khalid Rd, Al Shifa, Khamis Mushait 62433, Saudi Arabia;
| | - Mohammed A. Alqaad
- Department of Pharmaceutical Care Services, Al Noor Specialized Hospital, Makkah Health, Cluster, Makkah 24241, Saudi Arabia;
| | - Marwa B. Bakir
- Department of Medical Education, College of Medicine, Najran University, Najran 1988, Saudi Arabia;
| | - Basel A. Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran 1988, Saudi Arabia
| |
Collapse
|
44
|
Livieratos A, Lockley SW, Tsiodras S. Post infectious fatigue and circadian rhythm disruption in long-COVID and other infections: a need for further research. EClinicalMedicine 2025; 80:103073. [PMID: 39896874 PMCID: PMC11787434 DOI: 10.1016/j.eclinm.2025.103073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
Chronic fatigue syndrome (CFS) remains a subject of scientific research specifically with regards to its association with infections, including the more recently described Long COVID condition. Chronic fatigue and sleep disturbances in Long COVID are intricately linked to disruptions in circadian rhythms, driven by distinct molecular and cellular mechanisms triggered by SARS-CoV-2 infection. This can be driven by various mechanisms including dysregulation of key clock genes (CLOCK, BMAL1, PER2), mitochondrial dysfunction impairing oxidative phosphorylation, and cytokine-induced neuroinflammation (e.g., interleukin-6, tumor necrosis factor-alpha). Epigenetic changes, including DNA methylation at clock-related loci, particularly in peripheral tissues, further contribute to systemic circadian dysregulation. This work underscores the multifaceted molecular and systemic disruptions to circadian regulation in relation to fatigue and sleep disturbances identified as post-infectious sequelae, focusing on the Long COVID condition.
Collapse
Affiliation(s)
| | - Steven W Lockley
- Surrey Sleep Research Centre, School of Biosciences, University of Surrey, Surrey, GU2 7YW, UK
| | - Sotirios Tsiodras
- 4th Department of Internal Medicine, Attikon University Hospital, Athens 124 62, Greece
| |
Collapse
|
45
|
Singh D, Prasad S. A Pioneer Review on Lactoferrin-Conjugated Extracellular Nanovesicles for Targeting Cellular Melanoma: Recent Advancements and Future Prospects. Assay Drug Dev Technol 2025; 23:55-69. [PMID: 39654517 DOI: 10.1089/adt.2024.045] [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: 03/21/2025] Open
Abstract
Melanoma, a highly aggressive form of skin cancer, presents a formidable challenge in terms of treatment due to its propensity for metastasis and resistance to conventional therapies. The development of innovative nanocarriers for targeted drug delivery has opened new avenues in cancer therapy. Lactoferrin-conjugated extracellular nanovesicles (LF-EVs) have emerged as a promising vehicle in the targeted treatment of cellular melanoma, owing to their natural biocompatibility, enhanced bioavailability, and ability to traverse biological barriers effectively. This review synthesizes recent advancements in the use of LF-EVs as a novel drug delivery system for melanoma, emphasizing their unique capacity to enhance cellular uptake through LF's receptor-mediated endocytosis pathways. Key studies demonstrate that LF conjugation significantly increases the specificity of extracellular nanovesicles for melanoma cells, minimizes off-target effects, and promotes efficient intracellular drug release. Furthermore, we explore how LF-EVs interact with the tumor microenvironment, potentially inhibiting melanoma progression and metastasis while supporting antitumor immune responses. Future prospects in this field include optimizing LF conjugation techniques, improving the scalability of LF-EV production, and integrating multifunctional payloads to target drug resistance mechanisms. This review highlights the potential of LF-EVs to transform melanoma treatment strategies, bridging current gaps in therapeutic delivery and paving the way for personalized and less invasive melanoma therapies.
Collapse
Affiliation(s)
- Dilpreet Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India
| | - Sonima Prasad
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| |
Collapse
|
46
|
Zarei P, Sedeh PA, Vaez A, Keshteli AH. Using metabolomics to investigate the relationship between the metabolomic profile of the intestinal microbiota derivatives and mental disorders in inflammatory bowel diseases: a narrative review. Res Pharm Sci 2025; 20:1-24. [PMID: 40190827 PMCID: PMC11972020 DOI: 10.4103/rps.rps_273_23] [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: 12/20/2023] [Revised: 04/30/2024] [Accepted: 05/28/2024] [Indexed: 04/09/2025] Open
Abstract
Individuals with inflammatory bowel disease (IBD) are at a higher risk of developing mental disorders, such as anxiety and depression. The imbalance between the intestinal microbiota and its host, known as dysbiosis, is one of the factors, disrupting the balance of metabolite production and their signaling pathways, leading to disease progression. A metabolomics approach can help identify the role of gut microbiota in mental disorders associated with IBD by evaluating metabolites and their signaling comprehensively. This narrative review focuses on metabolomics studies that have comprehensively elucidated the altered gut microbial metabolites and their signaling pathways underlying mental disorders in IBD patients. The information was compiled by searching PubMed, Web of Science, Scopus, and Google Scholar from 2005 to 2023. The findings indicated that intestinal microbial dysbiosis in IBD patients leads to mental disorders such as anxiety and depression through disturbances in the metabolism of carbohydrates, sphingolipids, bile acids, neurotransmitters, neuroprotective, inflammatory factors, and amino acids. Furthermore, the reduction in the production of neuroprotective factors and the increase in inflammation observed in these patients can also contribute to the worsening of psychological symptoms. Analyzing the metabolite profile of the patients and comparing it with that of healthy individuals using advanced technologies like metabolomics, aids in the early diagnosis and prevention of mental disorders. This approach allows for the more precise identification of the microbes responsible for metabolite production, enabling the development of tailored dietary and pharmaceutical interventions or targeted manipulation of microbiota.
Collapse
Affiliation(s)
- Parvin Zarei
- Department of Bioinformatics, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Peyman Adibi Sedeh
- Isfahan Gastroenterology and Hepatology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Vaez
- Department of Bioinformatics, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Epidemiology, University of Groningen, University Medical Centre Groningen, 9713 GZ Groningen, The Netherlands
| | | |
Collapse
|
47
|
Bulduk BK, Tortajada J, Torres‐Egurrola L, Valiente‐Pallejà A, Martínez‐Leal R, Vilella E, Torrell H, Muntané G, Martorell L. High frequency of mitochondrial DNA rearrangements in the peripheral blood of adults with intellectual disability. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2025; 69:137-152. [PMID: 39506491 PMCID: PMC11735882 DOI: 10.1111/jir.13197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Mitochondrial DNA (mtDNA) rearrangements are recognised factors in mitochondrial disorders and ageing, but their involvement in neurodevelopmental disorders, particularly intellectual disability (ID) and autism spectrum disorder (ASD), remains poorly understood. Previous studies have reported mitochondrial dysfunction in individuals with both ID and ASD. The aim of this study was to investigate the prevalence of large-scale mtDNA rearrangements in ID and ID with comorbid ASD (ID-ASD). METHOD We used mtDNA-targeted next-generation sequencing and the MitoSAlt high-throughput computational pipeline in peripheral blood samples from 76 patients with ID (mean age 52.5 years, 37% female), 59 patients with ID-ASD (mean age 41.3 years, 46% female) and 32 healthy controls (mean age 42.4 years, 47% female) from Catalonia. RESULTS The study revealed a high frequency of mtDNA rearrangements in patients with ID, with 10/76 (13.2%) affected individuals. However, the prevalence was significantly lower in patients with ID-ASD 1/59 (1.7%) and in HC 1/32 (3.1%). Among the mtDNA rearrangements, six were identified as deletions (median size 6937 bp and median heteroplasmy level 2.3%) and six as duplications (median size 10 455 bp and median heteroplasmy level 1.9%). One of the duplications, MT-ATP6 m.8765-8793dup (29 bp), was present in four individuals with ID with a median heteroplasmy level of 3.9%. CONCLUSIONS Our results show that mtDNA rearrangements are frequent in patients with ID, but not in those with ID-ASD, when compared to HC. Additionally, MitoSAlt has demonstrated high sensitivity and accuracy in detecting mtDNA rearrangements, even at very low heteroplasmy levels in blood samples. While the high frequency of mtDNA rearrangements in ID is noteworthy, the role of these rearrangements is currently unclear and needs to be confirmed with further data, particularly in post-mitotic tissues and through age-matched control studies.
Collapse
Affiliation(s)
- B. K. Bulduk
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
| | - J. Tortajada
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
| | - L. Torres‐Egurrola
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
| | - A. Valiente‐Pallejà
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
- CIBER de Salud Mental (CIBERSAM)Instituto de Salud Carlos IIIMadridSpain
| | - R. Martínez‐Leal
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
- CIBER de Salud Mental (CIBERSAM)Instituto de Salud Carlos IIIMadridSpain
- Genètica i Ambient en PsiquiatriaIntellectual Disability and Developmental Disorders Research Unit (UNIVIDD), Fundació VillablancaReusCataloniaSpain
| | - E. Vilella
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
- CIBER de Salud Mental (CIBERSAM)Instituto de Salud Carlos IIIMadridSpain
| | - H. Torrell
- Centre for Omic Sciences (COS)Joint Unit Universitat Rovira i Virgili‐EURECAT Technology Centre of Catalonia, Unique Scientific and Technical InfrastructuresReusCataloniaSpain
| | - G. Muntané
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
- CIBER de Salud Mental (CIBERSAM)Instituto de Salud Carlos IIIMadridSpain
- Institut de Biologia Evolutiva (UPF‐CSIC), Department of Medicine and Life SciencesUniversitat Pompeu Fabra, Parc de Recerca Biomèdica de BarcelonaBarcelonaCataloniaSpain
| | - L. Martorell
- Àrea de RecercaHospital Universitari Institut Pere Mata (HUIPM)ReusCataloniaSpain
- Institut d'Investigació Sanitària Pere Virgili (IISPV‐CERCA)Universitat Rovira i Virgili (URV)ReusCataloniaSpain
- CIBER de Salud Mental (CIBERSAM)Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
48
|
Mustafa M, Abbas K, Habib S. A Comprehensive Report on Long COVID, Emerging Challenges, and Research Priorities in the Pandemic. CORONAVIRUSES 2025; 6. [DOI: 10.2174/0126667975293898240227062513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/30/2024] [Accepted: 02/14/2024] [Indexed: 05/04/2025]
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, India
| | - Kashif Abbas
- Department of
Zoology, Faculty of Life Sciences Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
49
|
Wang L, Kong Q, Leng X, Leung H, Li Y. The sphingosine-1-phosphate signaling pathway (sphingosine-1-phosphate and its receptor, sphingosine kinase) and epilepsy. Epilepsia Open 2025; 10:55-73. [PMID: 39727628 PMCID: PMC11803289 DOI: 10.1002/epi4.13112] [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: 04/15/2024] [Revised: 11/08/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Epilepsy is one of the common chronic neurological diseases, affecting more than 70 million people worldwide. The brains of people with epilepsy exhibit a pathological and persistent propensity for recurrent seizures. Epilepsy often coexists with cardiovascular disease, cognitive dysfunction, depression, etc., which seriously affects the patient's quality of life. Although our understanding of epilepsy has advanced, the pathophysiological mechanisms leading to epileptogenesis, drug resistance, and associated comorbidities remain largely unknown. The use of newer antiepileptic drugs has increased, but this has not improved overall outcomes. We need to deeply study the pathogenesis of epilepsy and find drugs that can not only prevent the epileptogenesis and interfere with the process of epileptogenesis but also treat epilepsy comorbidities. Sphingosine-1-phosphate (S1P) is an important lipid molecule. It not only forms the basis of cell membranes but is also an important bioactive mediator. It can not only act as a second messenger in cells to activate downstream signaling pathways but can also exert biological effects by being secreted outside cells and binding to S1P receptors on the cell membrane. Fingolimod (FTY720) is the first S1P receptor modulator developed and approved for the treatment of multiple sclerosis. More and more studies have proven that the S1P signaling pathway is closely related to epilepsy, drug-resistant epilepsy, epilepsy comorbidities, or other epilepsy-causing diseases. However, there is much controversy over the role of certain natural molecules in the pathway and receptor modulators (such as FTY720) in epilepsy. Here, we summarize and analyze the role of the S1P signaling pathway in epilepsy, provide a basis for finding potential therapeutic targets and/or epileptogenic biomarkers, analyze the reasons for these controversies, and put forward our opinions. PLAIN LANGUAGE SUMMARY: This article combines the latest research literature at home and abroad to review the sphingosine 1-phosphate signaling pathway and epileptogenesis, drug-resistant epilepsy, epilepsy comorbidities, other diseases that can cause epilepsy, as well as the sphingosine-1-phosphate signaling pathway regulators and epilepsy, with the expectation of providing a certain theoretical basis for finding potential epilepsy treatment targets and/or epileptogenic biomarkers in the sphingosine-1-phosphate signaling pathway.
Collapse
Affiliation(s)
- Lin Wang
- Department of NeurologyAffiliated Hospital of Jining Medical UniversityJining CityChina
- Epilepsy CenterAffiliated Hospital of Jining Medical UniversityJining CityChina
- The Chinese University of Hong Kong, Department of Medicine and TherapeuticsThe Chinese University of Hong Kong, Central AveHong KongHong Kong
| | - Qingxia Kong
- Department of NeurologyAffiliated Hospital of Jining Medical UniversityJining CityChina
- Epilepsy CenterAffiliated Hospital of Jining Medical UniversityJining CityChina
| | - Xinyi Leng
- The Chinese University of Hong Kong, Department of Medicine and TherapeuticsThe Chinese University of Hong Kong, Central AveHong KongHong Kong
| | - Howan Leung
- Division of Neurology, Department of Medicine and Therapeutics, Prince of Wales Hospital7/F Clinical Science Building, Prince of Wales HospitalHong KongHong Kong
| | - Yang Li
- Department of OncologyAffiliated Hospital of Jining Medical UniversityJining CityChina
| |
Collapse
|
50
|
Soudy M, Bars SL, Glaab E. Sex-dependent molecular landscape of Alzheimer's disease revealed by large-scale single-cell transcriptomics. Alzheimers Dement 2025; 21:e14476. [PMID: 39737748 PMCID: PMC11848167 DOI: 10.1002/alz.14476] [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: 10/07/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 01/01/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) shows significant sex differences in prevalence and clinical manifestations, but the underlying molecular mechanisms remain unclear. METHODS This study used a large-scale, single-cell transcriptomic atlas of the human prefrontal cortex to investigate sex-dependent molecular changes in AD. Our approach combined cell type-specific and sex-specific differential gene expression analysis, pathway enrichment, gene regulatory network construction, and cell-cell communication analysis to identify sex-dependent changes. RESULTS We found significant sex-specific gene expression patterns and pathway alterations in AD. Male astrocytes showed changes in cell death pathways, with RPTOR as a key regulator, while female astrocytes had alterations in Wnt signaling and cell cycle regulation. Cell-cell communication analysis uncovered sex-specific intercellular signaling patterns, with male-specific impacts on apoptosis-related signaling and female-specific alterations in Wnt and calcium signaling. DISCUSSION This study reveals sex-dependent gene expression patterns, pathway alterations, and intercellular communication changes, suggesting the need for sex-specific approaches in AD research. HIGHLIGHTS Single-cell transcriptomics reveals significant sex-specific molecular signatures in Alzheimer's disease (AD). Male astrocytes show enhanced modulation of apoptotic and cell death pathways in AD; female astrocytes exhibit distinct alterations in Wnt signaling and cell-cycle regulation. Sex-dimorphic changes in mitochondrial gene expression are observed in excitatory neurons, suggesting divergent energy metabolism profiles may contribute to AD sex differences. RPTOR is identified as a key regulator of male-specific changes in astrocytes, implicating the mechanistic target of rapamycin pathway in sex-dependent AD pathology. New cell-cell communication analyses reveal sex-specific patterns of intercellular signaling, providing insights into how cellular interactions may differentially contribute to AD pathology in males and females.
Collapse
Affiliation(s)
- Mohamed Soudy
- Biomedical Data Science GroupLuxembourg Centre for Systems Biomedicine (LCSB)University of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Sophie Le Bars
- Biomedical Data Science GroupLuxembourg Centre for Systems Biomedicine (LCSB)University of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Enrico Glaab
- Biomedical Data Science GroupLuxembourg Centre for Systems Biomedicine (LCSB)University of LuxembourgEsch‐sur‐AlzetteLuxembourg
| |
Collapse
|