1
|
Pang L, Huang Y, Li R, Guo L, Man C, Yang X, Jiang Y. Effects of postbiotics produced by Lactobacillus plantarum JM015 isolated from traditional fermented dairy products on Salmonella-induced intestinal inflammation: A preventive strategy. Food Chem 2025; 469:142549. [PMID: 39708644 DOI: 10.1016/j.foodchem.2024.142549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/03/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Affiliation(s)
- Lidong Pang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yan Huang
- Sanmenxia Polytechnic, Sanmenxia, 472000, China
| | - Runze Li
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Ling Guo
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Chaoxin Man
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xinyan Yang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Yujun Jiang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
2
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
3
|
Chen HJ, Huang TX, Jiang YX, Chen X, Wang AF. Multifunctional roles of inflammation and its causative factors in primary liver cancer: A literature review. World J Hepatol 2023; 15:1258-1271. [PMID: 38223416 PMCID: PMC10784815 DOI: 10.4254/wjh.v15.i12.1258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/06/2023] [Accepted: 11/24/2023] [Indexed: 12/25/2023] Open
Abstract
Primary liver cancer is a severe and complex disease, leading to 800000 global deaths annually. Emerging evidence suggests that inflammation is one of the critical factors in the development of hepatocellular carcinoma (HCC). Patients with viral hepatitis, alcoholic hepatitis, and steatohepatitis symptoms are at higher risk of developing HCC. However, not all inflammatory factors have a pathogenic function in HCC development. The current study describes the process and mechanism of hepatitis development and its progression to HCC, particularly focusing on viral hepatitis, alcoholic hepatitis, and steatohepatitis. Furthermore, the roles of some essential inflammatory cytokines in HCC progression are described in addition to a summary of future research directions.
Collapse
Affiliation(s)
- Hong-Jin Chen
- Department of Pharmacology, School of Basic Medical Sciences, Translational Medicine Research Center, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Ting-Xiong Huang
- School of Clinical Medical, Translational Medicine Research Center, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Yu-Xi Jiang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou 325035, Zhejiang Province, China
| | - Xiong Chen
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
- Department of Endocrinology, The People's Hospital of Yuhuan, The Yuhuan Branch of The First Affiliated Hospital of Wenzhou Medical University, Yuhuan 317600, Zhejiang Province, China
| | - Ai-Fang Wang
- Department of Endocrinology, The People's Hospital of Yuhuan, The Yuhuan Branch of The First Affiliated Hospital of Wenzhou Medical University, Yuhuan 317600, Zhejiang Province, China.
| |
Collapse
|
4
|
Zhang T, Shen HH, Qin XY, Li MQ. The metabolic characteristic of decidual immune cells and their unique properties in pregnancy loss. Immunol Rev 2022; 308:168-186. [PMID: 35582842 DOI: 10.1111/imr.13085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022]
Abstract
Maternal tolerance to semi- or fully allograft conceptus is a prerequisite for the maintenance of pregnancy. Once this homeostasis is disrupted, it may result in pregnancy loss. As a potential approach to prevent pregnancy loss, targeting decidual immune cells (DICs) at the maternal-fetal interface has been suggested. Although the phenotypic features and functions of DIC have been extensively profiled, the regulatory pathways for this unique immunological adaption have yet to be elucidated. In recent years, a pivotal mechanism has been highlighted in the area of immunometabolism, by which the changes in intracellular metabolic pathways in DIC and interaction with the adjacent metabolites in the microenvironment can alter their phenotypes and function. More inspiringly, the manipulation of metabolic profiling in DIC provides a novel avenue for the prevention and treatment of pregnancy loss. Herein, this review highlights the major metabolic programs (specifically, glycolysis, ATP-adenosine metabolism, lysophosphatidic acid metabolism, and amino acid metabolism) in multiple immune cells (including decidual NK cells, macrophages, and T cells) and their integrations with the metabolic microenvironment in normal pregnancy. Importantly, this perspective may help to provide a potential therapeutic strategy for reducing pregnancy loss via targeting this interplay.
Collapse
Affiliation(s)
- Tao Zhang
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Hui-Hui Shen
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Xue-Yun Qin
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai, China.,Shanghai Medical School, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
5
|
de Souza Basso B, Haute GV, Ortega-Ribera M, Luft C, Antunes GL, Bastos MS, Carlessi LP, Levorse VG, Cassel E, Donadio MVF, Santarém ER, Gracia-Sancho J, Rodrigues de Oliveira J. Methoxyeugenol deactivates hepatic stellate cells and attenuates liver fibrosis and inflammation through a PPAR-ɣ and NF-kB mechanism. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114433. [PMID: 34280502 DOI: 10.1016/j.jep.2021.114433] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/02/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Studies have shown interest in nutraceuticals for the prevention of liver diseases. Methoxyeugenol, is a molecule found in foods, such as nutmeg (Myristica fragrans Houtt.) and Brazilian red propolis. These two sources of methoxyeugenol, propolis and nutmeg, are used in folk medicine for the treatment of hepatic and gastrointestinal disorders, although little is known about their effects on the prevention of liver fibrosis. Natural PPAR (Peroxisome proliferator-activated receptor) agonists would represent unique molecules for therapy, considering the lack of therapeutics to treat liver fibrosis in chronic liver disease. Thus, investigation on new alternatives are necessary, including the search for natural compounds from renewable and sustainable sources. Liver fibrosis is a pathological process characterized by an exacerbated cicatricial response in the hepatic tissue, which compromises liver function. Therefore, inhibition of HSC (hepatic stellate cell) activation and hepatocyte damage are considered major strategies for the development of new anti-fibrotic treatments. AIM OF THE STUDY This study aimed to investigate the effects of methoxyeugenol treatment on HSC phenotype modulation in human and murine cells, hepatocyte damage prevention, and protective effects in vivo, in order to evaluate its therapeutic potential for liver fibrosis prevention. METHODS We investigated the effects of methoxyeugenol in (i) in vitro models using human and murine HSC and hepatocytes, and (ii) in vivo models of CCl4 (carbon tetrachloride) -induced liver fibrosis in mice. RESULTS We herein report that methoxyeugenol decreases HSC activation through the activation of PPAR-ɣ, ultimately inducing a quiescent phenotype highlighted by an increase in lipid droplets, loss of contraction ability, and a decrease in the proliferative rate and mRNA expression of fibroblast markers. In addition, methoxyeugenol prevented hepatocytes from oxidative stress damage. Moreover, in mice submitted to chronic liver disease through CCl4 administration, methoxyeugenol decreased the inflammatory profile, liver fibrosis, mRNA expression of fibrotic genes, and the inflammatory pathway signaled by NF-kB (Nuclear factor kappa B). CONCLUSION We propose methoxyeugenol as a novel and potential therapeutic approach to treat chronic liver disease and fibrosis.
Collapse
Affiliation(s)
- Bruno de Souza Basso
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Gabriela Viegas Haute
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Martí Ortega-Ribera
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Lab, IDIBAPS Biomedical Research Institute - CIBEREHD, Barcelona, Spain
| | - Carolina Luft
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Géssica Luana Antunes
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Matheus Scherer Bastos
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Leonardo Pfeiff Carlessi
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Vitor Giancarlo Levorse
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eduardo Cassel
- School of Technology, Pontifical Catholic University of Rio Grande do Sul - PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Márcio Vinícius Fagundes Donadio
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eliane Romanato Santarém
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Lab, IDIBAPS Biomedical Research Institute - CIBEREHD, Barcelona, Spain
| | - Jarbas Rodrigues de Oliveira
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul PUCRS, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
6
|
Atypical immunometabolism and metabolic reprogramming in liver cancer: Deciphering the role of gut microbiome. Adv Cancer Res 2020; 149:171-255. [PMID: 33579424 DOI: 10.1016/bs.acr.2020.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related mortality worldwide. Much recent research has delved into understanding the underlying molecular mechanisms of HCC pathogenesis, which has revealed to be heterogenous and complex. Two major hallmarks of HCC include: (i) a hijacked immunometabolism and (ii) a reprogramming in metabolic processes. We posit that the gut microbiota is a third component in an entanglement triangle contributing to HCC progression. Besides metagenomic studies highlighting the diagnostic potential in the gut microbiota profile, recent research is pinpointing the gut microbiota as an instigator, not just a mere bystander, in HCC. In this chapter, we discuss mechanistic insights on atypical immunometabolism and metabolic reprogramming in HCC, including the examination of tumor-associated macrophages and neutrophils, tumor-infiltrating lymphocytes (e.g., T-cell exhaustion, regulatory T-cells, natural killer T-cells), the Warburg effect, rewiring of the tricarboxylic acid cycle, and glutamine addiction. We further discuss the potential involvement of the gut microbiota in these characteristics of hepatocarcinogenesis. An immediate highlight is that microbiota metabolites (e.g., short chain fatty acids, secondary bile acids) can impair anti-tumor responses, which aggravates HCC. Lastly, we describe the rising 'new era' of immunotherapies (e.g., immune checkpoint inhibitors, adoptive T-cell transfer) and discuss for the potential incorporation of gut microbiota targeted therapeutics (e.g., probiotics, fecal microbiota transplantation) to alleviate HCC. Altogether, this chapter invigorates for continuous research to decipher the role of gut microbiome in HCC from its influence on immunometabolism and metabolic reprogramming.
Collapse
|
7
|
Costa BP, Nassr MT, Diz FM, Carlessi LP, Fernandes KHA, Nunes FB, Branchini G, de Oliveira JR. Fructose-1,6-bisphosphate induces generation of reactive oxygen species and activation of p53-dependent cell death in human endometrial cancer cells. J Appl Toxicol 2020; 41:1050-1062. [PMID: 33078453 DOI: 10.1002/jat.4091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/10/2020] [Accepted: 10/01/2020] [Indexed: 11/11/2022]
Abstract
Fructose-1,6-bisphosphate (F1,6BP), an intermediate of the glycolytic pathway, has been found to play a promising anticancer effect; nevertheless, the mechanisms involved remain poorly understood. The present study aimed to evaluate the effect and mechanisms of F1,6BP in a human endometrial cancer cell line (Ishikawa). F1,6BP showed an antiproliferative and non-cytotoxic effect on endometrial cancer cells. These effects are related to the increase in reactive oxygen species (ROS) levels and mitochondrial membrane potential (ΔΨm). These harmful stimuli trigger the upregulation of the expression of pro-apoptotic genes (p53 and Bax), leading to the reduction of cell proliferation through inducing programmed cell death by apoptosis. Furthermore, F1,6BP-treated cells had the formation of autophagosomes induced, as well as a decrease in their proliferative capacity after withdrawing the treatment. Our results demonstrate that F1,6BP acts as an anticancer agent through the generation of mitochondrial instability, loss of cell function, and p53-dependent cell death. Thus, F1,6BP proves to be a potential molecule for use in the treatment against endometrial cancer.
Collapse
Affiliation(s)
- Bruna Pasqualotto Costa
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Marcella Tornquist Nassr
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Fernando Mendonça Diz
- Program in Materials Engineering and Technology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Leonardo Pfeiff Carlessi
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Krist Helen Antunes Fernandes
- Laboratory of Clinical and Experimental Immunology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Fernanda Bordignon Nunes
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Laboratory of Cellular, Molecular and Computational Biophysics, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Gisele Branchini
- Laboratory of Cellular, Molecular and Computational Biophysics, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
8
|
Smecellato FB, Marsilli LRB, Nakamura JE, Jordani MC, Évora PRB, Castro-e-Silva O. Comparative study between fructose 1-6 bisphosphate and histidine-tryptophan-ketoglutarate in liver preservation in rats submitted to total cold ischemia. Acta Cir Bras 2020; 35:e202000603. [PMID: 32667586 PMCID: PMC7357840 DOI: 10.1590/s0102-865020200060000003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/18/2020] [Accepted: 05/22/2020] [Indexed: 11/22/2022] Open
Abstract
Purpose To compare Fructose-1,6-Bisphosphate (FBP) to Histidine-Tryptophan-Ketoglutarate (HTK) in liver preservation at cold ischemia. Methods Male rats (Sprague-Dawley: 280-340g) divided into three groups (n=7): Control; Fructose-1,6-bisphosphate (FBP); Histidine-Tryptophan-Ketoglutarate (HTK). Animals underwent laparotomy-thoracotomy for perfusion of livers with saline. Livers were removed and deposited into solutions. Mitochondria were isolated to determine State 3 (S3), State 4 (S4), Respiratory Control Ratio (RCR) and Swelling (S). Liver enzymes (AST, ALT, LDH) were determined in solution. At tissue, Malondialdehyde (MDA) and Nitrate (NOx) were determined. All parameters were analyzed at 0.6 and 24 hours of hypothermic preservation. Statistics analysis were made by Mann-Whitney test (p<0.05). Results Regarding ALT, there was a difference between FBP-6h/HTK-6h, lower in HTK. Regarding AST, there was a significant difference between FBP-24h/HTK-24h, lower in FBP. Regarding NOx, there was a difference between 0h and 6h, as well as 0h and 24h for both solutions. Regarding S3, there was a significant difference in 24h compared to Control-0h for both solutions, and a significant difference between FBP-6h/FBP-24h. Regarding S4, there was a difference between Control-0h/HTK-24h and FBP-24h/HTK-24h, higher in HTK. There was a difference between Control-0h/FBP-24h for Swelling, higher in FBP. Conclusion Fructose-1,6-Bisphosphate showed better performance at nitrate and aspartate aminotransferase compared to histidine-tryptophan-ketoglutarate.
Collapse
Affiliation(s)
- Fernanda Bombonato Smecellato
- Graduate student, Faculdade de Medicina de Marília (FAMEMA), Brazil. Technical procedures; acquisition, analysis and interpretation of data; manuscript preparation
| | - Lucas Ricardo Benfatti Marsilli
- Graduate student, Faculdade de Medicina de Marília (FAMEMA), Brazil. Technical procedures; acquisition, analysis and interpretation of data; manuscript preparation
| | - Julia Eico Nakamura
- Graduate student, Faculdade de Medicina de Marília (FAMEMA), Brazil. Technical procedures; acquisition, analysis and interpretation of data; manuscript preparation
| | - Maria Cecília Jordani
- Master, Biochemistry, Division of Digestive Surgery, Department of Surgery and Anatomy, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (FMRP-USP), Ribeirao Preto-SP, Brazil. Acquisition and interpretation of data, statistical analysis
| | - Paulo Roberto Barbosa Évora
- PhD, Full Professor, Division of Thoracic and Cardiovascular Surgery, Department of Surgery and Anatomy, FMRP-USP, Ribeirao Preto-SP, Brazil. Conception and design of the study, manuscript writing, critical revision
| | - Orlando Castro-e-Silva
- PhD, Full Professor, Department of Surgery and Anatomy, FMRP-USP, Ribeirao Preto-SP, Brazil. Conception and design of the study, analysis and interpretation of data, manuscript writing, critical revision
| |
Collapse
|
9
|
Dias HB, de Oliveira JR, Donadio MVF, Kimura S. Fructose-1,6-bisphosphate prevents pulmonary fibrosis by regulating extracellular matrix deposition and inducing phenotype reversal of lung myofibroblasts. PLoS One 2019; 14:e0222202. [PMID: 31509566 PMCID: PMC6738633 DOI: 10.1371/journal.pone.0222202] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023] Open
Abstract
Pulmonary fibrosis (PF) is the result of chronic injury where fibroblasts become activated and secrete large amounts of extracellular matrix (ECM), leading to impaired fibroblasts degradation followed by stiffness and loss of lung function. Fructose-1,6-bisphosphate (FBP), an intermediate of glycolytic pathway, decreases PF development, but the underlying mechanism is unknown. To address this issue, PF was induced in vivo using a mouse model, and pulmonary fibroblasts were isolated from healthy and fibrotic animals. In PF model mice, lung function was improved by FBP as revealed by reduced collagen deposition and downregulation of ECM gene expression such as collagens and fibronectin. Fibrotic lung fibroblasts (FLF) treated with FBP for 3 days in vitro showed decreased proliferation, contraction, and migration, which are characteristic of myofibroblast to fibroblast phenotype reversal. ECM-related genes and proteins such as collagens, fibronectin and α-smooth muscle actin, were also downregulated in FBP-treated FLF. Moreover, matrix metalloproteinase (MMP) 1, responsible for ECM degradation, was produced only in fibroblasts obtained from healthy lungs (HLF) and FBP did not alter its expression. On the other hand, tissue inhibitor of metalloproteinase (TIMP)-1, a MMP1 inhibitor, and MMP2, related to fibroblast tissue-invasion, were predominantly produced by FLF and FBP was able to downregulate its expression. These results demonstrate that FBP may prevent bleomycin-induced PF development through reduced expression of collagen and other ECM components mediated by a reduced TIMP-1 and MMP2 expression.
Collapse
Affiliation(s)
- Henrique Bregolin Dias
- Laboratory of Cellular Biophysics and Inflammation, PUCRS, Porto Alegre, RS, Brazil
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | | | | | - Shioko Kimura
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
10
|
Ning Y, Feng W, Cao X, Ren K, Quan M, Chen A, Xu C, Qiu Y, Cao J, Li X, Luo X. Genistein inhibits stemness of SKOV3 cells induced by macrophages co-cultured with ovarian cancer stem-like cells through IL-8/STAT3 axis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:19. [PMID: 30646963 PMCID: PMC6334437 DOI: 10.1186/s13046-018-1010-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/17/2018] [Indexed: 01/27/2023]
Abstract
BACKGROUND Recent studies showed that macrophages co-cultured with ovarian cancer stem-like cells (OCSLCs) induced SKOV3 cell stemness via IL-8/STAT3 signaling. Genistein (GEN) demonstrates chemopreventive activity in inflammation-associated cancers. The present study aimed to examine whether and if GEN inhibits the stemness of SKOV3 and OVCA-3R cells induced by co-culture of THP-1 macrophages and SKOV3-derived OCSLCs. METHODS The co-culture was treated with or without different concentrations (10, 20, and 40 μmol/L) of GEN for 24 h. Depletion or addition of IL-8 in Co-CM and knockdown or overexpression of STAT3 in THP-1 macrophages was performed to demonstrate the possible associated mechanisms. The combined effects of GEN and STAT3 knockdown were examined with the nude mouse modle by co-injection of SKOV3-derived OCSLCs with THP-1 macrophages. RESULTS Our results showed that GEN down-regulated CD163 and p-STAT3 expression of THP-1 macrophage, decreased the levels of IL-10, increased the levels of IL-12 and nitric oxide (NO) in the conditioned medium, and reduced the clonogenic and sphere-forming capacities and the expression of CD133 and CD44 in SKOV3 cells induced by co-culture of THP-1 macrophages and OCSLCs in a dose-dependent manner. Moreover, depletion or addition of IL-8 enhanced or attenuated the effect of GEN. Additionally, knockdown or overepression of STAT3 in THP-1 macrophages potentiated or attenuated the inhibitory effects of GEN. Importantly, STAT3 overexpression retrieved the effects of IL-8 combined with GEN depletion on M2 polarization of THP-1 macrophages and stemness of SKOV3 cells induced by co-culture. The combination of GEN and STAT3 knockdown cooperatively inhibited the growth of tumors co-inoculated with OCSLCs/THP-1 macrophages in nude mice in vivo through blocking IL-8/STAT3 signaling. CONCLUSIONS In summary, our findings suggested that GEN can inhibit the increased M2 polarization of macrophages and stemness of ovarian cancer cells by co-culture of macrophages with OCSLCs through disrupting IL-8/STAT3 signaling axis. This assisted GEN to be as a potential chemotherapeutic agent in human ovarian cancer.
Collapse
Affiliation(s)
- Yingxia Ning
- grid.470124.4Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120 China ,0000 0004 1760 3828grid.412601.0The First Affiliated Hospital of Jinan University, Guangzhou, 510632 China
| | - Weifeng Feng
- 0000 0004 1760 3828grid.412601.0The First Affiliated Hospital of Jinan University, Guangzhou, 510632 China
| | - Xiaocheng Cao
- 0000 0001 0089 3695grid.411427.5Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013 China ,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013 China
| | - Kaiqun Ren
- 0000 0001 0089 3695grid.411427.5Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013 China ,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013 China
| | - Meifang Quan
- 0000 0001 0089 3695grid.411427.5Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013 China ,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013 China
| | - A. Chen
- 0000 0001 0089 3695grid.411427.5Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013 China ,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013 China
| | - Chang Xu
- 0000 0001 0089 3695grid.411427.5Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013 China ,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013 China
| | - Yebei Qiu
- 0000 0001 0089 3695grid.411427.5Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013 China ,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013 China
| | - Jianguo Cao
- 0000 0001 0089 3695grid.411427.5Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013 China ,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013 China
| | - Xiang Li
- 0000 0001 0089 3695grid.411427.5Department of preclinical medicine, Medical College, Hunan Normal University, Changsha, 410013 China ,0000 0001 0089 3695grid.411427.5Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013 China ,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013 China
| | - Xin Luo
- 0000 0004 1760 3828grid.412601.0The First Affiliated Hospital of Jinan University, Guangzhou, 510632 China
| |
Collapse
|
11
|
Friend or foe? Mitochondria as a pharmacological target in cancer treatment. Future Med Chem 2017; 9:2197-2210. [PMID: 29182013 DOI: 10.4155/fmc-2017-0110] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mitochondria have acquired numerous functions over the course of evolution, such as those involved in controlling energy production, cellular metabolism, cell survival, apoptosis and autophagy within host cells. Tumor cells can develop defects in mitochondrial function, presenting a potential strategy for designing selective anticancer therapies. Therefore, cancer has been the main focus of recent research to uncover possible mitochondrial targets for therapeutic benefit. This comprehensive review covers not only the recent discoveries of the roles of mitochondria in cancer development, progression and therapeutic implications but also the findings regarding emerging mitochondrial therapeutic targets and mitochondria-targeted agents. Current challenges and future directions for developments and applications of mitochondrial-targeted therapeutics are also discussed.
Collapse
|