1
|
Kovacevic I, Schmidt PH, Kowalski A, Helms BJ, Lest CHAVD, Kluttig A, Posern G. ER stress inhibition enhances formation of triacylglcerols and protects endothelial cells from lipotoxicity. Cell Commun Signal 2024; 22:304. [PMID: 38831326 PMCID: PMC11145897 DOI: 10.1186/s12964-024-01682-y] [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: 02/26/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
Elevated concentrations of palmitate in serum of obese individuals can impair endothelial function, contributing to development of cardiovascular disease. Although several molecular mechanisms of palmitate-induced endothelial dysfunction have been proposed, there is no consensus on what signaling event is the initial trigger of detrimental palmitate effects. Here we report that inhibitors of ER stress or ceramid synthesis can rescue palmitate-induced autophagy impairment in macro- and microvascular endothelial cells. Furthermore, palmitate-induced cholesterol synthesis was reverted using these inhibitors. Similar to cell culture data, autophagy markers were increased in serum of obese individuals. Subsequent lipidomic analysis revealed that palmitate changed the composition of membrane phospholipids in endothelial cells and that these effects were not reverted upon application of above-mentioned inhibitors. However, ER stress inhibition in palmitate-treated cells enhanced the synthesis of trilglycerides and restored ceramide levels to control condition. Our results suggest that palmitate induces ER-stress presumably by shift in membrane architecture, leading to impaired synthesis of triglycerides and enhanced production of ceramides and cholesterol, which altogether enhances lipotoxicity of palmitate in endothelial cells.
Collapse
Affiliation(s)
- Igor Kovacevic
- Institute of Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany.
| | - Paula Henriette Schmidt
- Institute of Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany
| | - Annkatrin Kowalski
- Institute of Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany
| | - Bernd J Helms
- Department Biomolecular Health Sciences, Veterinary Medicine, Utrecht University, Utrecht, 3584CM, The Netherlands
| | - Chris H A van de Lest
- Department Biomolecular Health Sciences, Veterinary Medicine, Utrecht University, Utrecht, 3584CM, The Netherlands
| | - Alexander Kluttig
- Institute of Medical Epidemiology, Biostatistics, and Informatics, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Guido Posern
- Institute of Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany
| |
Collapse
|
2
|
Sadeghi A, Niknam M, Momeni-Moghaddam MA, Shabani M, Aria H, Bastin A, Teimouri M, Meshkani R, Akbari H. Crosstalk between autophagy and insulin resistance: evidence from different tissues. Eur J Med Res 2023; 28:456. [PMID: 37876013 PMCID: PMC10599071 DOI: 10.1186/s40001-023-01424-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 10/03/2023] [Indexed: 10/26/2023] Open
Abstract
Insulin is a critical hormone that promotes energy storage in various tissues, as well as anabolic functions. Insulin resistance significantly reduces these responses, resulting in pathological conditions, such as obesity and type 2 diabetes mellitus (T2DM). The management of insulin resistance requires better knowledge of its pathophysiological mechanisms to prevent secondary complications, such as cardiovascular diseases (CVDs). Recent evidence regarding the etiological mechanisms behind insulin resistance emphasizes the role of energy imbalance and neurohormonal dysregulation, both of which are closely regulated by autophagy. Autophagy is a conserved process that maintains homeostasis in cells. Accordingly, autophagy abnormalities have been linked to a variety of metabolic disorders, including insulin resistance, T2DM, obesity, and CVDs. Thus, there may be a link between autophagy and insulin resistance. Therefore, the interaction between autophagy and insulin function will be examined in this review, particularly in insulin-responsive tissues, such as adipose tissue, liver, and skeletal muscle.
Collapse
Affiliation(s)
- Asie Sadeghi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Niknam
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Shabani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Aria
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Bastin
- Clinical Research Development Center "The Persian Gulf Martyrs" Hospital, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Maryam Teimouri
- Department of Biochemistry, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Akbari
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
3
|
Walter S, Jung T, Herpich C, Norman K, Pivovarova-Ramich O, Ott C. Determination of the autophagic flux in murine and human peripheral blood mononuclear cells. Front Cell Dev Biol 2023; 11:1122998. [PMID: 36994103 PMCID: PMC10040559 DOI: 10.3389/fcell.2023.1122998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
The autophagy lysosomal system (ALS) is crucial for cellular homeostasis, contributing to maintain whole body health and alterations are associated with diseases like cancer or cardiovascular diseases. For determining the autophagic flux, inhibition of lysosomal degradation is mandatory, highly complicating autophagy measurement in vivo. To overcome this, herein blood cells were used as they are easy and routinely to isolate. Within this study we provide detailed protocols for determination of the autophagic flux in peripheral blood mononuclear cells (PBMCs) isolated from human and, to our knowledge the first time, also from murine whole blood, extensively discussing advantages and disadvantages of both methods. Isolation of PBMCs was performed using density gradient centrifugation. To minimize changes on the autophagic flux through experimental conditions, cells were directly treated with concanamycin A (ConA) for 2 h at 37°C in their serum or for murine cells in serum filled up with NaCl. ConA treatment decreased lysosomal cathepsins activity and increased Sequestosome 1 (SQSTM1) protein and LC3A/B-II:LC3A/B-I ratio in murine PBMCs, while transcription factor EB was not altered yet. Aging further enhanced ConA-associated increase in SQSTM1 protein in murine PBMCs but not in cardiomyocytes, indicating tissue-specific differences in autophagic flux. In human PBMCs, ConA treatment also decreased lysosomal activity and increased LC3A/B-II protein levels, demonstrating successful autophagic flux detection in human subjects. In summary, both protocols are suitable to determine the autophagic flux in murine and human samples and may facilitate a better mechanistic understanding of altered autophagy in aging and disease models and to further develop novel treatment strategies.
Collapse
Affiliation(s)
- Sophia Walter
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Tobias Jung
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Catrin Herpich
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Geriatrics and Medical Gerontology, Charité Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Berlin, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Kristina Norman
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Geriatrics and Medical Gerontology, Charité Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Berlin, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Olga Pivovarova-Ramich
- Department of Molecular Nutritional Medicine, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Christiane Ott
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- *Correspondence: Christiane Ott,
| |
Collapse
|
4
|
Malesza IJ, Malesza M, Walkowiak J, Mussin N, Walkowiak D, Aringazina R, Bartkowiak-Wieczorek J, Mądry E. High-Fat, Western-Style Diet, Systemic Inflammation, and Gut Microbiota: A Narrative Review. Cells 2021; 10:cells10113164. [PMID: 34831387 PMCID: PMC8619527 DOI: 10.3390/cells10113164] [Citation(s) in RCA: 353] [Impact Index Per Article: 88.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota is responsible for recovering energy from food, providing hosts with vitamins, and providing a barrier function against exogenous pathogens. In addition, it is involved in maintaining the integrity of the intestinal epithelial barrier, crucial for the functional maturation of the gut immune system. The Western diet (WD)—an unhealthy diet with high consumption of fats—can be broadly characterized by overeating, frequent snacking, and a prolonged postprandial state. The term WD is commonly known and intuitively understood. However, the strict digital expression of nutrient ratios is not precisely defined. Based on the US data for 1908–1989, the calory intake available from fats increased from 32% to 45%. Besides the metabolic aspects (hyperinsulinemia, insulin resistance, dyslipidemia, sympathetic nervous system and renin-angiotensin system overstimulation, and oxidative stress), the consequences of excessive fat consumption (high-fat diet—HFD) comprise dysbiosis, gut barrier dysfunction, increased intestinal permeability, and leakage of toxic bacterial metabolites into the circulation. These can strongly contribute to the development of low-grade systemic inflammation. This narrative review highlights the most important recent advances linking HFD-driven dysbiosis and HFD-related inflammation, presents the pathomechanisms for these phenomena, and examines the possible causative relationship between pro-inflammatory status and gut microbiota changes.
Collapse
Affiliation(s)
- Ida Judyta Malesza
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (I.J.M.); (J.W.)
| | - Michał Malesza
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (M.M.); (J.B.-W.)
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (I.J.M.); (J.W.)
| | - Nadiar Mussin
- Department of General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe 030012, Kazakhstan;
| | - Dariusz Walkowiak
- Department of Organization and Management in Health Care, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | - Raisa Aringazina
- Department of Internal Diseases No. 1, West Kazakhstan Marat Ospanov Medical University, Aktobe 030012, Kazakhstan;
| | | | - Edyta Mądry
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznań, Poland; (M.M.); (J.B.-W.)
- Correspondence:
| |
Collapse
|
5
|
Rong X, Xu J, Jiang Y, Li F, Chen Y, Dou QP, Li D. Citrus peel flavonoid nobiletin alleviates lipopolysaccharide-induced inflammation by activating IL-6/STAT3/FOXO3a-mediated autophagy. Food Funct 2021; 12:1305-1317. [PMID: 33439200 DOI: 10.1039/d0fo02141e] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nobiletin, a polymethoxyflavone widely present in the peel of citrus fruits, has significant anti-inflammatory activity. Autophagy plays a critical role in maintaining cell homeostasis by promoting the degradation of intracellular structures in response to various stress. Recent research suggests the involvement of autophagy in the inflammatory process and therefore some inflammation-related diseases. However, the "cross-talk" between autophagy and nobiletin's anti-inflammation response remains not well elucidated. Therefore, this study was initiated with the aim of investigating the role of autophagy in nobiletin's protective effect against inflammation in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. Results showed that nobiletin significantly (P < 0.05) inhibited the release of nitric oxide (NO) and decreased the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in a dose-dependent manner. Moreover, nobiletin significantly (P < 0.05) promoted autophagy as evidenced by the appearance of more autophagosomes, up-regulated LC3II protein, low-regulated p62 protein, and increased autophagy-related (Atg) genes' expression compared with the control treated with LPS alone. Addition of chloroquine, an autophagy inhibitor, alleviated nobiletin's anti-inflammatory effect, further supporting the requirement of an active autophagy process for the citrus peel flavonoid's biological activity. Mechanistically, we found that nobiletin treatment leads to activation of the IL-6/STAT3/FOXO3a signal pathway through the down-regulation of IL-6 and STAT3 phosphorylation and the upregulation of FOXO3a phosphorylation in the cell nucleus, which is responsible for induction of macrophage autophagy. Taken together, our study provides evidence that nobiletin suppresses inflammatory response through enhancing autophagy through activating the IL-6/STAT3/FOXO3a pathway in macrophage cells.
Collapse
Affiliation(s)
- Xue Rong
- Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, College of Food Science and Engineering, Shandong Agricultural University, Taian 271018, China.
| | - Jie Xu
- Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, College of Food Science and Engineering, Shandong Agricultural University, Taian 271018, China.
| | - Yang Jiang
- Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, College of Food Science and Engineering, Shandong Agricultural University, Taian 271018, China.
| | - Feng Li
- Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, College of Food Science and Engineering, Shandong Agricultural University, Taian 271018, China.
| | - Yilun Chen
- Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, College of Food Science and Engineering, Shandong Agricultural University, Taian 271018, China.
| | - Q Ping Dou
- Barbara Ann Karmanos Cancer Institute and Departments of Oncology, Pharmacology and Pathology, Wayne State University School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Dapeng Li
- Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, College of Food Science and Engineering, Shandong Agricultural University, Taian 271018, China.
| |
Collapse
|
6
|
Amorim FT, Nava RC, Escobar KA, Li Z, Welch AM, Fennel ZJ, McKenna ZJ, Gibson AL. Autophagy in peripheral blood mononuclear cells is associated with body fat percentage. Arch Physiol Biochem 2021:1-7. [PMID: 33724888 DOI: 10.1080/13813455.2021.1887267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
CONTEXT Numerous chronic conditions including obesity exhibit autophagic dysfunction. Association of immune cell autophagic marker regulation by body fat percentage (%BF) is unknown. OBJECTIVE Investigate autophagy activity in peripheral blood mononuclear cells (PBMCs) of adults with distinct %BFs and obesity-related circulating inflammatory markers. MATERIALS AND METHODS Sixteen individuals (eight males) with %BF above (n = 8, 36.9 ± 3.6 years, 27.1 ± 8.1%BF) and below (n = 8, 37.1 ± 3.7 years, 13.3 ± 3.7%BF) their age- and sex-specific 50th percentile value based on the American College Sports Medicine guidelines participated. Body fat percentage was calculated from hydrostatic weighing. PBMCs were isolated from venous blood, and PBMC autophagic flux markers (LC3-I, LC3-II, and p62) were measured via Western blot. CRP, resistin, leptin, and adiponectin were measured via ELISA. RESULTS LC3-II/LC3-I ratio correlated with %BF (r=-0.56, p=.023). Insulin (p=.05) and CRP (p=.018) were higher in high %BF participants. DISCUSSION AND CONCLUSIONS Autophagic activity markers in PBMCs correlate with %BF, but are not different between %BF groups.
Collapse
Affiliation(s)
- Fabiano T Amorim
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Roberto C Nava
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Kurt A Escobar
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, NM, USA
- Department of Kinesiology, Physiology of Exercise and Sport Lab, California State University Long Beach, Long Beach, CA, USA
| | - Zidong Li
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Anna M Welch
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Zachary J Fennel
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Zachary J McKenna
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Ann L Gibson
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
7
|
Bastin AR, Nazari-Robati M, Sadeghi H, Doustimotlagh AH, Sadeghi A. Trehalose and N-Acetyl Cysteine Alleviate Inflammatory Cytokine Production and Oxidative Stress in LPS-Stimulated Human Peripheral Blood Mononuclear Cells. Immunol Invest 2021; 51:963-979. [PMID: 33632046 DOI: 10.1080/08820139.2021.1891095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Background: Evidence has shown that inflammation and oxidative stress are implicated in the development of a great number of human diseases. Trehalose possesses various biological effects including antioxidant and anti-inflammatory activities. However, there is little data on the effects of trehalose on human cells including peripheral blood mononuclear cells (PBMCs). Here, we aimed to investigate whether trehalose could attenuate oxidative stress and inflammation induced by lipopolysaccharides (LPS) in PBMCs.Methods: The enzyme-linked immunosorbent assay (ELISA) and RT-PCR were used to assess the levels of inflammatory cytokines. To investigate the phosphorylation of c-Jun N-terminal kinase (JNK) and NF-κB, western blot analysis was utilized. Oxidant-antioxidant markers were assessed using ELISA and colorimetric procedures.Results: The results revealed that trehalose significantly mitigated the effect of LPS on the phosphorylation of JNK and NF-κB-P65 (p < .00). This mitigation was associated with significantly reduced levels of inflammatory cytokines IL-6, TNF-α, and IL-1β and increased levels of anti-inflammatory cytokine IL-10 (P < .05). The antioxidant N-acetyl cysteine (NAC) also showed similar effects on JNK and NF-κB-P65 phosphorylation and inflammatory cytokines (p < .00). Furthermore, trehalose alleviated oxidative stress in LPS-stimulated PBMCs as it reversed the altered levels of malondialdehyde and total thiols (p ≤ .05) and restored the activity of antioxidant enzymes glutathione peroxidase and manganese superoxide dismutase (p < .001).Conclusion: The results of this study indicated that trehalose prevented inflammation and oxidative stress in the LPS-stimulated PBMCs, providing evidence for the benefits of trehalose as a potential therapeutic agent in inflammatory conditions.Abbreviations: LPS: Lipopolysaccharide; NAC: N-Acetyl cysteine; ROS: Reactive oxygen species; IL-6: Interleukin-6; TNF-α: Tumor necrosis factor-alpha; SOD: Superoxide dismutase; GPx: Glutathione peroxidase; MDA: Malondialdehyde; MAPK: Mitogen-activated protein kinases; JNK: c-Jun N-terminal kinase; NF-κB: Nuclear factor kappa-light-chain-enhancer of activated B cells.
Collapse
Affiliation(s)
- Ali Reza Bastin
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Clinical Research Development Center "The Persian Gulf Martyrs" Hospital, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mahdieh Nazari-Robati
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Sadeghi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Amir Hossein Doustimotlagh
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Asie Sadeghi
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Student Research Committee, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
8
|
Nagarajan SR, Butler LM, Hoy AJ. The diversity and breadth of cancer cell fatty acid metabolism. Cancer Metab 2021; 9:2. [PMID: 33413672 PMCID: PMC7791669 DOI: 10.1186/s40170-020-00237-2] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor cellular metabolism exhibits distinguishing features that collectively enhance biomass synthesis while maintaining redox balance and cellular homeostasis. These attributes reflect the complex interactions between cell-intrinsic factors such as genomic-transcriptomic regulation and cell-extrinsic influences, including growth factor and nutrient availability. Alongside glucose and amino acid metabolism, fatty acid metabolism supports tumorigenesis and disease progression through a range of processes including membrane biosynthesis, energy storage and production, and generation of signaling intermediates. Here, we highlight the complexity of cellular fatty acid metabolism in cancer, the various inputs and outputs of the intracellular free fatty acid pool, and the numerous ways that these pathways influence disease behavior.
Collapse
Affiliation(s)
- Shilpa R Nagarajan
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
| | - Lisa M Butler
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Andrew J Hoy
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
9
|
Liu XC, Lu JJ, Chen YM, Qiu Y, Zheng MD, Wang ZL, Li XW. [Roles of autophagy onself-renewal and differentiation of mesenchymal stem cells]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2020; 38:704-707. [PMID: 33377351 DOI: 10.7518/hxkq.2020.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mesenchymal stem cells (MSCs), which have the potential of self-replication and differentiation, are a very valuable cell source for stem cell-based medical therapy. Their application has opened up a new way for disease research. Although MSCs can maintain cell stemness through self-renewal, with the prolongation of cell passage and culture time, the stemness of MSCs gradually decays, and the cell aging and differentiation potential decreases gradually. Autophagy is a highly conserved cytological process that degrades the modified, excess, and deleterious cytoplasmic components in autophagosomes, which are then degraded by fusion with lysosomes. As the main intracellular degradation and recycling pathway, autophagy plays an active role in maintaining cell homeostasis, self-renewal and pluripotency. In this paper, the role of autophagy in self-renewal and maintenance of multidirectional differentiation potential of MSCs was reviewed, which laid a theoretical foundation and practical basis for the research and application of MSCs.
Collapse
Affiliation(s)
- Xin-Chen Liu
- Dept. of Endodontics, Jilin University Hospital of Stomatology, Changchun 130021, China;Jilin Province Key Laboratory of Dental Development and Jaw Remodeling and Regeneration, Changchun 130021, China
| | - Jin-Jin Lu
- Jilin Province Key Laboratory of Dental Development and Jaw Remodeling and Regeneration, Changchun 130021, China;Dept. of Pediatric Dentistry, Jilin University Hospital of Stomatology, Changchun 130021, China
| | - Yu-Meng Chen
- Jilin Province Key Laboratory of Dental Development and Jaw Remodeling and Regeneration, Changchun 130021, China;Dept. of Pathology, Jilin University Hospital of Stomatology, Changchun 130021, China
| | - Ying Qiu
- Dept. of Endodontics, Jilin University Hospital of Stomatology, Changchun 130021, China;Jilin Province Key Laboratory of Dental Development and Jaw Remodeling and Regeneration, Changchun 130021, China
| | - Meng-Dan Zheng
- Jilin Province Key Laboratory of Dental Development and Jaw Remodeling and Regeneration, Changchun 130021, China;Dept. of Pathology, Jilin University Hospital of Stomatology, Changchun 130021, China
| | - Zi-Lin Wang
- Jilin Province Key Laboratory of Dental Development and Jaw Remodeling and Regeneration, Changchun 130021, China;Dept. of Pathology, Jilin University Hospital of Stomatology, Changchun 130021, China
| | - Xiang-Wei Li
- Dept. of Endodontics, Jilin University Hospital of Stomatology, Changchun 130021, China
| |
Collapse
|
10
|
Effect of high-fat diet on peripheral blood mononuclear cells and adipose tissue in early stages of diet-induced weight gain. Br J Nutr 2020; 122:1359-1367. [PMID: 31554524 DOI: 10.1017/s0007114519002472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Subcutaneous adipose tissue (scAT) and peripheral blood mononuclear cells (PBMC) play a significant role in obesity-associated systemic low-grade inflammation. High-fat diet (HFD) is known to induce inflammatory changes in both scAT and PBMC. However, the time course of the effect of HFD on these systems is still unknown. The aim of the present study was to determine the time course of the effect of HFD on PBMC and scAT. New Zealand white rabbits were fed HFD for 5 or 10 weeks (i.e. HFD-5 and HFD-10) or regular chow (i.e. control (CNT)-5 and CNT-10). Thereafter, metabolic and inflammatory parameters of PBMC and scAT were quantified. HFD induced hyperfattyacidaemia in HFD-5 and HFD-10 groups, with the development of insulin resistance in HFD-10, while no changes were observed in scAT lipid metabolism and inflammatory status. HFD activated the inflammatory pathways in PBMC of HFD-5 group and induced modified autophagy in that of HFD-10. The rate of fat oxidation in PBMC was directly associated with the expression of inflammatory markers and tended to inversely associate with autophagosome formation markers in PBMC. HFD affected systemic substrate metabolism, and the metabolic, inflammatory and autophagy pathways in PBMC in the absence of metabolic and inflammatory changes in scAT. Dietary approaches or interventions to avert HFD-induced changes in PBMC could be essential to prevent metabolic and inflammatory complications of obesity and promote healthier living.
Collapse
|
11
|
Librán-Pérez M, Pereiro P, Figueras A, Novoa B. Antiviral activity of palmitic acid via autophagic flux inhibition in zebrafish (Danio rerio). FISH & SHELLFISH IMMUNOLOGY 2019; 95:595-605. [PMID: 31676430 DOI: 10.1016/j.fsi.2019.10.055] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/11/2019] [Accepted: 10/28/2019] [Indexed: 06/10/2023]
Abstract
Fatty acids (FAs) are key elements that affect not only growth but also different immune functions, and therefore, nutrition is important for growing healthy fish. Zebrafish (Danio rerio) is a good model for assessing the beneficial effects of immunostimulants, including FAs, before applying them in aquaculture. Accordingly, this study evaluated the effects of palmitic acid (PA) treatment on different immune parameters of zebrafish and on the mortality caused by the spring viremia of carp virus (SVCV). The results suggest that PA modulates the infection outcome in vivo, which benefits zebrafish and results in reduced mortality and viral titres. The antiviral protection elicited by this FA seems to be associated with the inhibition of autophagy and is independent of other immune processes, such as neutrophil proliferation or type I interferon (IFN) activity. The use of PA as an immunostimulant at low concentrations showed great potential in the prevention of SVCV infections; therefore, this FA could help to prevent the mortality and morbidity caused by viral agents in aquacultured fish. Nevertheless, the potentially detrimental effects of suppressing autophagy in the organism should be taken into account.
Collapse
Affiliation(s)
- Marta Librán-Pérez
- Instituto de Investigaciones Marinas (IIM), CSIC, Eduardo Cabello 6, 36208, Vigo, Spain
| | - Patricia Pereiro
- Instituto de Investigaciones Marinas (IIM), CSIC, Eduardo Cabello 6, 36208, Vigo, Spain
| | - Antonio Figueras
- Instituto de Investigaciones Marinas (IIM), CSIC, Eduardo Cabello 6, 36208, Vigo, Spain
| | - Beatriz Novoa
- Instituto de Investigaciones Marinas (IIM), CSIC, Eduardo Cabello 6, 36208, Vigo, Spain.
| |
Collapse
|
12
|
Chatterjee T, Pattanayak R, Ukil A, Chowdhury S, Bhattacharyya M. Autophagy protects peripheral blood mononuclear cells against inflammation, oxidative and nitrosative stress in diabetic dyslipidemia. Free Radic Biol Med 2019; 143:309-323. [PMID: 31369843 DOI: 10.1016/j.freeradbiomed.2019.07.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/28/2019] [Accepted: 07/28/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) results in severe oxidative and nitrosative stress and inflammation when associated with hyperlipidemia. In this study, we have attempted to explore the role of autophagy in T2DM subjects with or without dyslipidemia. METHODS Experiments were carried out in isolated Peripheral blood mononuclear cells (PBMC) from study subjects and insulin resistant HepG2 cells utilizing flow cytometry, confocal microscopy and molecular biology techniques like western blotting, immunofluorescence and real time PCR. RESULTS In case of T2DM with dyslipidemia, higher population of autophagy positive cell was detected compared to T2DM which may have been originated due to higher stress. Flow cytometric data indicated autophagy to be triggered by both oxidative and nitrosative stress in PBMC of diabetic dyslipidemic patients, which is a novel finding of our work. Expression of LC3 puncta, a hallmark of autophagy was observed at periphery of PBMC and Hep G2 cells in case of diabetic dyslipidemic condition. Increased expression of ATG5, LC3B and Beclin1 supports the autophagic pathway in both PBMC and Hep G2 cells. Upon blocking autophagy by 3-methyl adenine (3MA), the apoptotic cell population increased significantly. Autophagy was also been evidenced to control oxidative stress mediated up-regulation of inflammatory markers like IL-6, TNF-α. CONCLUSION Induction of autophagy emerged to be a protective mechanism for the diabetic cells coupled with dyslipidemia. Not only Reactive oxygen species, but also reactive nitrogen species was involved in autophagy induction process. Moreover inhibition study documented autophagy to have a protective role in pro-inflammatory responses. Thus, enhancing autophagic activity may be an efficient mechanism leading to new therapeutic strategy to restore the glycemic regulation.
Collapse
Affiliation(s)
- Tanima Chatterjee
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India
| | - Rudradip Pattanayak
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India; Jagadis Bose National Science Talent Search, 1300, Rajdanga Main Road, Kolkata, 700109, India
| | - Anindita Ukil
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India
| | - Subhankar Chowdhury
- Institute of Postgraduate Medical Education and Research, Government of West Bengal, 224, Acharya Jagadish Chandra Bose Road, Kolkata, 700020, India
| | - Maitree Bhattacharyya
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India; Jagadis Bose National Science Talent Search, 1300, Rajdanga Main Road, Kolkata, 700109, India.
| |
Collapse
|
13
|
Sadeghi A, Shabani M, Alizadeh S, Meshkani R. Interplay between oxidative stress and autophagy function and its role in inflammatory cytokine expression induced by palmitate in skeletal muscle cells. Cytokine 2019; 125:154835. [PMID: 31479873 DOI: 10.1016/j.cyto.2019.154835] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/05/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023]
Abstract
Autophagy is a cellular process activated in response to various stresses such as starvation, hypoxia, and oxidative stress. Autophagy was reported to modulate the inflammatory pathways. However, whether autophagy is involved in regulation of palmitate-induced inflammation of skeletal muscle C2C12 cells is still unknown. The present study aimed to investigate the autophagic pathway in C2C12 cells treated with 0.5 mM palmitate. The results showed that the protein levels of LC3BII and P62 were increased in C2C12 cells after 12 h palmitate treatment. Besides, inhibition of autophagy by chloroquine or 3-methyladenin and its activation by rapamycin were associated with elevated mRNA and protein levels of IL-6 and TNF-α inflammatory cytokines in C2C12 cells. To study the mechanism by which autophagy impairment leads to activation of inflammatory responses, reactive oxygen species (ROS) levels in palmitate-treated cells were measured. The results showed that while palmitate stimulates ROS production, pretreatment of the cells with N-acetyl cysteine (NAC), a ROS scavenger, reduced inflammatory responses and also improved LC3-BII and P62 protein in the C2C12 cells exposed to palmitate. These findings suggest that palmitate-induced defect of autophagic flux leads to elevated inflammatory cytokine expression in the skeletal muscle cells by regulating the oxidative stress process.
Collapse
Affiliation(s)
- Asie Sadeghi
- Student Research Committee, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Maryam Shabani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Alizadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
The Mitochondrial Antioxidant SS-31 Modulates Oxidative Stress, Endoplasmic Reticulum Stress, and Autophagy in Type 2 Diabetes. J Clin Med 2019; 8:jcm8091322. [PMID: 31466264 PMCID: PMC6780723 DOI: 10.3390/jcm8091322] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/09/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial dysfunction has been shown to play a central role in the pathophysiology of type 2 diabetes (T2D), and mitochondria-targeted agents such as SS-31 are emerging as a promising strategy for its treatment. We aimed to study the effects of SS-31 on leukocytes from T2D patients by evaluating oxidative stress, endoplasmic reticulum (ER) stress and autophagy. Sixty-one T2D patients and 53 controls were included. Anthropometric and analytical measurements were performed. We also assessed reactive oxygen species (ROS) production, calcium content, the expression of ER stress markers GRP78, CHOP, P-eIF2α, and autophagy-related proteins Beclin1, LC3 II/I, and p62 in leukocytes from T2D and control subjects treated or not with SS-31. Furthermore, we have evaluated the action of SS-31 on leukocyte-endothelium interactions. T2D patients exhibited elevated ROS concentration, calcium levels and presence of ER markers (GRP78 and CHOP gene expression, and GRP78 and P-eIF2α protein expression), all of which were reduced by SS-31 treatment. SS-31 also led to a drop in BECN1 gene expression, and Beclin1 and LC3 II/I protein expression in T2D patients. In contrast, the T2D group displayed reduced p62 protein levels that were restored by SS-31. SS-20 (with non-antioxidant activity) did not change any analyzed parameter. In addition, SS-31 decreased rolling flux and leukocyte adhesion, and increased rolling velocity in T2D patients. Our findings suggest that SS-31 exerts potentially beneficial effects on leukocytes of T2D patients modulating oxidative stress and autophagy, and ameliorating ER stress.
Collapse
|
15
|
Di-n-butyl phthalate, butylbenzyl phthalate, and their metabolites exhibit different apoptotic potential in human peripheral blood mononuclear cells. Food Chem Toxicol 2019; 133:110750. [PMID: 31390533 DOI: 10.1016/j.fct.2019.110750] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 07/23/2019] [Accepted: 08/02/2019] [Indexed: 11/20/2022]
Abstract
Human peripheral blood mononuclear cells (PBMCs) are one of the main cell models used in studies concerning the exposure of humans (in vitro) to various chemical substances. Changes in PBMCs may reflect the general reaction of the organism regarding the effect of xenobiotics. The aim of this work was to evaluate the effect of di-n-butyl phthalate (DBP), butylbenzyl phthalate (BBP) and their metabolites: mono-n-butylphthalate (MBP), mono-benzylphthalate (MBzP) upon the induction of apoptosis in human peripheral blood mononuclear cells in vitro. PBMCs were incubated with the studied compounds at concentrations from 1 to 100 μg/mL for 12 h and/or 24 h. In order to clarify the mechanism of phthalates-induced programmed cell death, the changes in the calcium ions (Ca2+) level, alterations in the transmembrane mitochondrial potential (ΔѰm) and caspase-8, -9, -3 activity as well as externalization of phosphatidylserine have been determined. An increased Ca2+ level and a reduction of the ΔѰm were observed in PBMCs incubated with all of the studied compounds, and particularly with DBP and BBP. Phthalates caused an increase of caspases activity. The most pronounced increase was observed for caspase -9. The most pronounced pro-apoptotic changes were caused by DBP followed by BBP and then by their metabolites.
Collapse
|
16
|
Balaban S, Nassar ZD, Zhang AY, Hosseini-Beheshti E, Centenera MM, Schreuder M, Lin HM, Aishah A, Varney B, Liu-Fu F, Lee LS, Nagarajan SR, Shearer RF, Hardie RA, Raftopulos NL, Kakani MS, Saunders DN, Holst J, Horvath LG, Butler LM, Hoy AJ. Extracellular Fatty Acids Are the Major Contributor to Lipid Synthesis in Prostate Cancer. Mol Cancer Res 2019; 17:949-962. [PMID: 30647103 DOI: 10.1158/1541-7786.mcr-18-0347] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 09/22/2018] [Accepted: 01/07/2019] [Indexed: 02/06/2023]
Abstract
Prostate cancer cells exhibit altered cellular metabolism but, notably, not the hallmarks of Warburg metabolism. Prostate cancer cells exhibit increased de novo synthesis of fatty acids (FA); however, little is known about how extracellular FAs, such as those in the circulation, may support prostate cancer progression. Here, we show that increasing FA availability increased intracellular triacylglycerol content in cultured patient-derived tumor explants, LNCaP and C4-2B spheroids, a range of prostate cancer cells (LNCaP, C4-2B, 22Rv1, PC-3), and prostate epithelial cells (PNT1). Extracellular FAs are the major source (∼83%) of carbons to the total lipid pool in all cell lines, compared with glucose (∼13%) and glutamine (∼4%), and FA oxidation rates are greater in prostate cancer cells compared with PNT1 cells, which preferentially partitioned extracellular FAs into triacylglycerols. Because of the higher rates of FA oxidation in C4-2B cells, cells remained viable when challenged by the addition of palmitate to culture media and inhibition of mitochondrial FA oxidation sensitized C4-2B cells to palmitate-induced apoptosis. Whereas in PC-3 cells, palmitate induced apoptosis, which was prevented by pretreatment of PC-3 cells with FAs, and this protective effect required DGAT-1-mediated triacylglycerol synthesis. These outcomes highlight for the first-time heterogeneity of lipid metabolism in prostate cancer cells and the potential influence that obesity-associated dyslipidemia or host circulating has on prostate cancer progression. IMPLICATIONS: Extracellular-derived FAs are primary building blocks for complex lipids and heterogeneity in FA metabolism exists in prostate cancer that can influence tumor cell behavior.
Collapse
Affiliation(s)
- Seher Balaban
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Zeyad D Nassar
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia.,South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Alison Y Zhang
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute for Medical Research, Darlinghurst, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | - Elham Hosseini-Beheshti
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Margaret M Centenera
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia.,South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Mark Schreuder
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.,Faculty of Medicine, University of Utrecht, Utrecht, the Netherlands
| | - Hui-Ming Lin
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute for Medical Research, Darlinghurst, New South Wales, Australia
| | - Atqiya Aishah
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Bianca Varney
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Frank Liu-Fu
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Lisa S Lee
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Shilpa R Nagarajan
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Robert F Shearer
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute for Medical Research, Darlinghurst, New South Wales, Australia
| | - Rae-Anne Hardie
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Origins of Cancer Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Nikki L Raftopulos
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Meghna S Kakani
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Darren N Saunders
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jeff Holst
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Origins of Cancer Program, Centenary Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Lisa G Horvath
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute for Medical Research, Darlinghurst, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia.,School of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia.,Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Lisa M Butler
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia.,South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Andrew J Hoy
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.
| |
Collapse
|
17
|
Balaban S, Lee LS, Varney B, Aishah A, Gao Q, Shearer RF, Saunders DN, Grewal T, Hoy AJ. Heterogeneity of fatty acid metabolism in breast cancer cells underlies differential sensitivity to palmitate-induced apoptosis. Mol Oncol 2018; 12:1623-1638. [PMID: 30099850 PMCID: PMC6120225 DOI: 10.1002/1878-0261.12368] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 07/18/2018] [Accepted: 08/03/2018] [Indexed: 01/02/2023] Open
Abstract
Breast cancer (BrCa) metabolism is geared toward biomass synthesis and maintenance of reductive capacity. Changes in glucose and glutamine metabolism in BrCa have been widely reported, yet the contribution of fatty acids (FAs) in BrCa biology remains to be determined. We recently reported that adipocyte coculture alters MCF-7 and MDA-MB-231 cell metabolism and promotes proliferation and migration. Since adipocytes are FA-rich, and these FAs are transferred to BrCa cells, we sought to elucidate the FA metabolism of BrCa cells and their response to FA-rich environments. MCF-7 and MDA-MB-231 cells incubated in serum-containing media supplemented with FAs accumulate extracellular FAs as intracellular triacylglycerols (TAG) in a dose-dependent manner, with MDA-MB-231 cells accumulating more TAG. The differences in TAG levels were a consequence of distinct differences in intracellular partitioning of FAs, and not due to differences in the rate of FA uptake. Specifically, MCF-7 cells preferentially partition FAs into mitochondrial oxidation, whereas MDA-MB-231 cells partition FAs into TAG synthesis. These differences in intracellular FA handling underpin differences in the sensitivity to palmitate-induced lipotoxicity, with MDA-MB-231 cells being highly sensitive, whereas MCF-7 cells are partially protected. The attenuation of palmitate-induced lipotoxicity in MCF-7 cells was reversed by inhibition of FA oxidation. Pretreatment of MDA-MB-231 cells with FAs increased TAG synthesis and reduced palmitate-induced apoptosis. Our results provide novel insight into the potential influences of obesity on BrCa biology, highlighting distinct differences in FA metabolism in MCF-7 and MDA-MB-231 cells and how lipid-rich environments modulate these effects.
Collapse
Affiliation(s)
- Seher Balaban
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, The University of Sydney, Australia
| | - Lisa S Lee
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, The University of Sydney, Australia
| | - Bianca Varney
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, The University of Sydney, Australia
| | - Atqiya Aishah
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, The University of Sydney, Australia
| | - Quanqing Gao
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Australia
| | - Robert F Shearer
- Kinghorn Cancer Center, Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Australia
| | - Andrew J Hoy
- Discipline of Physiology, Faculty of Medicine and Health, School of Medical Sciences & Bosch Institute, Charles Perkins Centre, The University of Sydney, Australia
| |
Collapse
|
18
|
Pontrelli P, Oranger A, Barozzino M, Divella C, Conserva F, Fiore MG, Rossi R, Papale M, Castellano G, Simone S, Laviola L, Giorgino F, Piscitelli D, Gallone A, Gesualdo L. Deregulation of autophagy under hyperglycemic conditions is dependent on increased lysine 63 ubiquitination: a candidate mechanism in the progression of diabetic nephropathy. J Mol Med (Berl) 2018; 96:645-659. [PMID: 29806072 DOI: 10.1007/s00109-018-1656-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 11/30/2022]
Abstract
Diabetic nephropathy patients (DN) are characterized by increased lysine63 ubiquitination (Lys63-Ub) at the tubular level. Autophagy is deregulated under diabetic conditions, even though the molecular mechanisms and the consequences of this alteration need to be elucidated. The aim of this study was to investigate the link between Lys63-Ub and autophagy in DN and the involvement of these two processes in tubular cell fate. Immunohistochemistry of beclin-1, LC3, and p62 on kidney biopsies highlighted increased protein expression of all these autophagic factors at the tubular level in DN compared to other nephritis. Transmission electron microscopy confirmed the presence of diffuse vacuolization and autophago(lyso)somal structures in proximal tubular cells in DN. Accumulation of Lys63-Ub proteins in DN increased in accordance with the tubular damage and was associated to increased LC3 expression both in vivo and in vitro. Hyperglycemia (HG) induced LC3 and p62 protein expression in HK2 cells together with Lys63-ubiquitinated proteins, and the inhibition of HG-induced Lys63-Ub by NSC697923 inhibitor, significantly reduced both LC3 and p62 expression. Moreover, in DN, those tubules expressing LC3 showed increased caspase-3 expression, supporting the hypothesis that deregulated autophagy induces apoptosis of tubular cells. In vitro, we confirmed a tight association between impaired autophagy, Lys63-Ub, and apoptosis since Lys63-Ub inhibition by NSC697923 abrogated HG-induced cell death and LC3 silencing also blocked hyperglycemia-induced caspase-3 activation. Our data suggested that prolonged hyperglycemia in diabetic patients can impair autophagy as a consequence of Lys63-Ub protein accumulation, thus promoting intracellular autophagic vesicles increase, finally leading to tubular cell death in DN. KEY MESSAGES In vivo autophagy is deregulated in diabetic patients with renal disease (DN). Accumulation of Lys63 ubiquitinated proteins is associated to autophagy deregulation. Accumulation of Lys63 ubiquitinated proteins correlated with apoptosis activation. Lys63 ubiquitination inhibition abrogated hyperglycemia-induced autophagy and apoptosis.
Collapse
Affiliation(s)
- Paola Pontrelli
- Department of Emergency and Organ Transplantation - Division of Nephrology, University of Bari Aldo Moro, Bari, Italy.
| | - Annarita Oranger
- Department of Emergency and Organ Transplantation - Division of Nephrology, University of Bari Aldo Moro, Bari, Italy
| | - Mariagrazia Barozzino
- Department of Emergency and Organ Transplantation - Division of Nephrology, University of Bari Aldo Moro, Bari, Italy
| | - Chiara Divella
- Department of Emergency and Organ Transplantation - Division of Nephrology, University of Bari Aldo Moro, Bari, Italy
| | - Francesca Conserva
- Department of Emergency and Organ Transplantation - Division of Nephrology, University of Bari Aldo Moro, Bari, Italy
| | - Maria Grazia Fiore
- Department of Emergency and Organ Transplantation - Division of Pathological Anatomy, University of Bari Aldo Moro, Bari, Italy
| | - Roberta Rossi
- Department of Emergency and Organ Transplantation - Division of Pathological Anatomy, University of Bari Aldo Moro, Bari, Italy
| | - Massimo Papale
- Department of Emergency and Organ Transplantation - Division of Nephrology, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Castellano
- Department of Emergency and Organ Transplantation - Division of Nephrology, University of Bari Aldo Moro, Bari, Italy
| | - Simona Simone
- Department of Emergency and Organ Transplantation - Division of Nephrology, University of Bari Aldo Moro, Bari, Italy
| | - Luigi Laviola
- Department of Emergency and Organ Transplantation - Division of Endocrinology, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Giorgino
- Department of Emergency and Organ Transplantation - Division of Endocrinology, University of Bari Aldo Moro, Bari, Italy
| | - Domenico Piscitelli
- Department of Emergency and Organ Transplantation - Division of Pathological Anatomy, University of Bari Aldo Moro, Bari, Italy
| | - Anna Gallone
- Department of Basic Medical Sciences, Neurosciences and Sense Organs - Division of Applied Biology, University of Bari Aldo Moro, Bari, Italy
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation - Division of Nephrology, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|