1
|
Brzozowa-Zasada M, Piecuch A, Bajdak-Rusinek K, Michalski M, Klymenko O, Matysiak N, Janelt K, Czuba Z. Glutathione Reductase Expression and Its Prognostic Significance in Colon Cancer. Int J Mol Sci 2024; 25:1097. [PMID: 38256170 PMCID: PMC10816751 DOI: 10.3390/ijms25021097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Maintaining a balanced redox state within cells is crucial for the sustenance of life. The process involves continuous cytosolic disulfide reduction reactions to restore oxidized proteins to their reduced thiol forms. There are two main cellular antioxidant pathways-the thioredoxin (Trx) and glutathione (GSH)/glutaredoxin (Grx) systems. In the GSH/Grx system, glutathione reductase (GR; GSR) catalyses the reduction of GSH disulfide (GSSG) to its sulfhydryl form (GSH), which can then further reduce oxidized Grxs. GR is an essential enzyme that helps in maintaining the supply of reduced glutathione-GSH, which is a significant reducing thiol found in most cells and known for its antioxidant properties. Therefore, it can have a significant impact on cancer development. To investigate this further, we performed an immunohistochemical analysis of GR protein expression in colon adenocarcinoma samples collected from patients with primary colon adenocarcinoma (stage I and II) and patients with metastasis to regional lymph nodes (stage III). The results of our study revealed a significant relationship between the immunohistochemical expression of GR and tumour histological grade, depth of invasion, regional lymph node involvement, staging, and PCNA immunohistochemical expression. It was found that 95% of patients with stage I had low levels of GR expression, whereas 89% of patients with stage III had high levels of immunohistochemical expression. A high level of expression was also detected in the patients with stage II of the disease, where almost 63% were characterized by a high expression of GR. The Western blot method revealed that the highest level of expression was found in the LS 174T cell line, which corresponds to stage II. The results of our study indicate that the immunohistochemical expression of GR may act as an independent prognostic factor associated with colon adenocarcinoma patients' prognosis.
Collapse
Affiliation(s)
- Marlena Brzozowa-Zasada
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Adam Piecuch
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Karolina Bajdak-Rusinek
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Marek Michalski
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
- Zabrze Silesian Nanomicroscopy Centre in Zabrze, Silesia LabMed—Research and Implementation Centre, Medical University of Silesia, 40-055 Katowice, Poland
| | - Olesya Klymenko
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Natalia Matysiak
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Kamil Janelt
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Zenon Czuba
- Department of Microbiology and Immunology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Jordana 19, 41-808 Zabrze, Poland
| |
Collapse
|
2
|
Salyha N, Oliynyk I. Hypoxia modeling techniques: A review. Heliyon 2023; 9:e13238. [PMID: 36718422 PMCID: PMC9877323 DOI: 10.1016/j.heliyon.2023.e13238] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/08/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Hypoxia is the main cause and effect of a large number of diseases, including the most recent one facing the world, the coronavirus disease (COVID-19). Hypoxia is divided into short-term, long-term, and periodic, it can be the result of diseases, climate change, or living and traveling in the high mountain regions of the world. Since each type of hypoxia can be a cause and a consequence of various physiological changes, the methods for modeling these hypoxias are also different. There are many techniques for modeling hypoxia under experimental conditions. The most common animal for modeling hypoxia is a rat. Hypoxia models (hypoxia simulations) in rats are a tool to study the effect of various conditions on the oxygen supply of the body. These models can provide a necessary information to understand hypoxia and also provide effective treatment, highlighting the importance of various reactions of the body to hypoxia. The main parameters when choosing a model should be reproducibility and the goal that the scientist wants to achieve. Hypoxia in rats can be reproduced both ways exogenously and endogenously. The reason for writing this review was the aim to systematize the models of rats available in the literature in order to facilitate their selection by scientists. The relative strengths and limitations of each model need to be identified and understood in order to evaluate the information obtained from these models and extrapolate these results to humans to develop the necessary generalizations. Despite these problems, animal models have been and remain vital to understanding the mechanisms involved in the development and progression of hypoxia. The eligibility criteria for the selected studies was a comprehensive review of the methods and results obtained from the studies. This made it possible to make generalizations and give recommendations on the application of these methods. The review will assist scientists in choosing an appropriate hypoxia simulation method, as well as assist in interpreting the results obtained with these methods.
Collapse
Affiliation(s)
- Nataliya Salyha
- Institute of Animal Biology NAAS, Lviv, Ukraine,Corresponding author
| | | |
Collapse
|
3
|
Qiu ZK, Zhang MZ, Zhang WC, Li ZJ, Si LB, Long X, Yu NZ, Wang XJ. Role of HIF-1α in pathogenic mechanisms of keloids. J Cosmet Dermatol 2023; 22:1436-1448. [PMID: 36718786 DOI: 10.1111/jocd.15601] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/08/2022] [Accepted: 12/12/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUDS AND OBJECTIVE Keloids are defined as overrepairing products that develop after skin lesions. Keloids are characterized by the proliferation of fibroblasts and the overaccumulation of extracellular matrix components (mainly collagen), leading to a locally hypoxic microenvironment. Hence, this article was aimed to review hypoxia in pathogenesis of keloids. METHODS We reviewed and summarized the relevant published studies. RESULTS Hypoxia results in the accumulation of hypoxia-inducible factor 1α (HIF-1α) in keloids, contributing to overactivation of the fibrotic signaling pathway, epithelial-mesenchymal transition, and changes in metabolism, eventually leading to aggravated fibrosis, infiltrative growth, and radiotherapy resistance. CONCLUSION It is, therefore, essential to understand the role of HIF-1α in the pathogenic mechanisms of keloids in order to develop new therapeutic approaches.
Collapse
Affiliation(s)
- Zi-Kai Qiu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Zi Zhang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen-Chao Zhang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Jin Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lou-Bin Si
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan-Ze Yu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Jun Wang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Norda S, Papadantonaki R. Regulation of cells of the arterial wall by hypoxia and its role in the development of atherosclerosis. VASA 2023; 52:6-21. [PMID: 36484144 DOI: 10.1024/0301-1526/a001044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The cell's response to hypoxia depends on stabilization of the hypoxia-inducible factor 1 complex and transactivation of nuclear factor kappa-B (NF-κB). HIF target gene transcription in cells resident to atherosclerotic lesions adjoins a complex interplay of cytokines and mediators of inflammation affecting cholesterol uptake, migration, and inflammation. Maladaptive activation of the HIF-pathway and transactivation of nuclear factor kappa-B causes monocytes to invade early atherosclerotic lesions, maintaining inflammation and aggravating a low-oxygen environment. Meanwhile HIF-dependent upregulation of the ATP-binding cassette transporter ABCA1 causes attenuation of cholesterol efflux and ultimately macrophages becoming foam cells. Hypoxia facilitates neovascularization by upregulation of vascular endothelial growth factor (VEGF) secreted by endothelial cells and vascular smooth muscle cells lining the arterial wall destabilizing the plaque. HIF-knockout animal models and inhibitor studies were able to show beneficial effects on atherogenesis by counteracting the HIF-pathway in the cell wall. In this review the authors elaborate on the up-to-date literature on regulation of cells of the arterial wall through activation of HIF-1α and its effect on atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Stephen Norda
- Department of Cardiovascular Medicine, University Hospital Münster, Germany
| | - Rosa Papadantonaki
- Emergency Department, West Middlesex University Hospital, Chelsea and Westminster NHS Trust, London, United Kingdom
| |
Collapse
|
5
|
Hypoxia-Inducible Factor 1 and Mitochondria: An Intimate Connection. Biomolecules 2022; 13:biom13010050. [PMID: 36671435 PMCID: PMC9855368 DOI: 10.3390/biom13010050] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
The general objective of the review is to explain the interaction between HIF-1 and mitochondria. On the one hand, this review describes the effects of HIF-1 on mitochondrial structure, including quantity, distribution, and morphology, as well as on mitochondrial metabolism and respiratory function. On the other hand, various factors, including mitochondrial activation of enzymes, the respiratory chain, complex and decoupling proteins, affect the stability and activity of HIF-1. It is possible to develop future molecular therapeutic interventions by understanding the interrelationships between HIF-1 and mitochondria.
Collapse
|
6
|
Barzegari A, Aaboulhassanzadeh S, Landon R, Gueguen V, Meddahi-Pellé A, Parvizpour S, Anagnostou F, Pavon-Djavid G. Mitohormesis and mitochondrial dynamics in the regulation of stem cell fate. J Cell Physiol 2022; 237:3435-3448. [PMID: 35775725 DOI: 10.1002/jcp.30820] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/11/2022]
Abstract
The ability of stem cells for self-renewing, differentiation, and regeneration of injured tissues is believed to occur via the hormetic modulation of nuclear/mitochondrial signal transductions. The evidence now indicates that in damaged tissues, the mitochondria set off the alarm under oxidative stress conditions, hence they are the central regulators of stem cell fate decisions. This review aimed to provide an update to a broader concept of stem cell fate in stress conditions of damaged tissues, and insights for the mitochondrial hormesis (mitohormesis), including the integrated stress response (ISR), mitochondrial dynamics, mitochondria uncoupling, unfolded protein response, and mitokines, with implications for the control of stem cells programing in a successful clinical cell therapy.
Collapse
Affiliation(s)
- Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sobhan Aaboulhassanzadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rebecca Landon
- CNRS UMR7052-INSERM U1271, Laboratory of Osteoarticular Biology, Bioengineering and Bioimaging, Paris Diderot University, Paris, France
| | - Virginie Gueguen
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| | - Anne Meddahi-Pellé
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| | - Sepideh Parvizpour
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fani Anagnostou
- CNRS UMR7052-INSERM U1271, Laboratory of Osteoarticular Biology, Bioengineering and Bioimaging, Paris Diderot University, Paris, France
| | - Graciela Pavon-Djavid
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| |
Collapse
|
7
|
Navarrete S, Solar C, Tapia R, Pereira J, Fuentes E, Palomo I. Pathophysiology of deep vein thrombosis. Clin Exp Med 2022:10.1007/s10238-022-00829-w. [PMID: 35471714 DOI: 10.1007/s10238-022-00829-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 03/31/2022] [Indexed: 12/29/2022]
Abstract
Deep venous thrombosis is a frequent, multifactorial disease and a leading cause of morbidity and mortality. Most of the time deep venous thrombosis is triggered by the interaction between acquired risk factors, such as hip fracture, pregnancy, and immobility, and hereditary risk factors such as thrombophilias. The mechanisms underlying deep venous thrombosis are not fully elucidated; however, in recent years, important advances have shed light on the role of venous flow, endothelium, platelets, leukocytes, and the interaction between inflammation and hemostasis. It has been described that the alteration of venous blood flow produces endothelial activation, favoring the adhesion of platelets and leukocytes, which, through tissue factor expression and neutrophil extracellular traps formation, contribute to the activation of coagulation, trapping more cells, such as red blood cells. Thus, the concerted interaction of these phenomena allows the formation and growth of the thrombus. In this work, the main mechanisms involved in the pathophysiology of deep vein thrombosis will be described.
Collapse
Affiliation(s)
- Simón Navarrete
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Av. Lircay s/n, 3460000, Talca, Chile
| | - Carla Solar
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Av. Lircay s/n, 3460000, Talca, Chile
| | | | - Jaime Pereira
- Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Av. Lircay s/n, 3460000, Talca, Chile
| | - Iván Palomo
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Av. Lircay s/n, 3460000, Talca, Chile.
| |
Collapse
|
8
|
Xu K, Lu C, Ren X, Wang J, Xu P, Zhang Y. Overexpression of HIF-1α enhances the protective effect of mitophagy on steroid-induced osteocytes apoptosis. ENVIRONMENTAL TOXICOLOGY 2021; 36:2123-2137. [PMID: 34310007 DOI: 10.1002/tox.23327] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/01/2021] [Accepted: 07/07/2021] [Indexed: 06/13/2023]
Abstract
Glucocorticoid (GC; dexamethasone, DEX) -induced osteonecrosis of the femoral head (GIOFH) is a challenging orthopedic disease, and its underlying mechanism remains not clear. This study exposed murine long bone osteocyte-Y4 (MLO-Y4) cells to DEX below normoxic or hypoxic circumstances and found that cell autophagy have been reduced. At the same time, flow cytometry analysis showed increased apoptosis, which was more pronounced in hypoxic environments. Recent research also claimed that GC induces osteoporosis after osteocyte apoptosis, and subsequent microfractures lead to ischemia and hypoxia of the femoral head, resulted in GIOFH. Presently, we found that both mitophagy-related protein hypoxia-inducible factor-1α (HIF-1α) and BNIP3 were up-regulated in the hypoxic environment, and their expression was down-regulated when exposed to DEX. Besides, we demonstrated that overexpressing HIF-1α resisted DEX-induced apoptosis in a hypoxic environment. Here, we demonstrated that overexpression of HIF-1α, through its downstream marker BNIP3, reduced the suppression of DEX on mitophagy induced by hypoxia and protected bone cells from apoptosis. Also, these findings may provide a direction of the promising application for better GIOFH treatment shortly.
Collapse
Affiliation(s)
- Ke Xu
- Department of Orthopaedics of the First Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, China
| | - Chao Lu
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyu Ren
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jing Wang
- Department of Orthopaedics of the First Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, China
| | - Peng Xu
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yingang Zhang
- Department of Orthopaedics of the First Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
9
|
Kuwabara T, Ishikawa F, Ikeda M, Ide T, Kohwi-Shigematsu T, Tanaka Y, Kondo M. SATB1-dependent mitochondrial ROS production controls TCR signaling in CD4 T cells. Life Sci Alliance 2021; 4:4/11/e202101093. [PMID: 34583974 PMCID: PMC8500228 DOI: 10.26508/lsa.202101093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 11/24/2022] Open
Abstract
SATB1 regulates mitochondrial function and reactive oxygen species (ROS) production through the expression of mitochondrial transcription factor A. SATB1-mediated ROS production is necessary for TCR stimulation and T-cell function. Special AT-rich sequence binding protein-1 (SATB1) is localized to the nucleus and remodels chromatin structure in T cells. SATB1-deficient CD4 T cells cannot respond to TCR stimulation; however, the cause of this unresponsiveness is to be clarified. Here, we demonstrate that SATB1 is indispensable to proper mitochondrial functioning and necessary for the activation of signal cascades via the TCR in CD4 T cells. Naïve SATB1-deficient CD4 T cells contain fewer mitochondria than WT T cells, as the former do not express mitochondrial transcription factor A (TFAM). Impaired mitochondrial function in SATB1-deficient T cells subverts mitochondrial ROS production and SHP-1 inactivation by constitutive oxidization. Ectopic TFAM expression increases mitochondrial mass and mitochondrial ROS production and rescues defects in the antigen-specific response in the SATB1-deficient T cells. Thus, SATB1 is vital for maintaining mitochondrial mass and function by regulating TFAM expression, which is necessary for TCR signaling.
Collapse
Affiliation(s)
- Taku Kuwabara
- Department of Molecular Immunology, Toho University School of Medicine, Tokyo, Japan
| | - Fumio Ishikawa
- Department of Molecular Immunology, Toho University School of Medicine, Tokyo, Japan.,Faculty of Health Sciences, Tsukuba International University, Tsuchiura, Japan
| | - Masataka Ikeda
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Terumi Kohwi-Shigematsu
- Department of Orofacial Science, University of California San Francisco School of Dentistry, San Francisco, CA, USA
| | - Yuriko Tanaka
- Department of Molecular Immunology, Toho University School of Medicine, Tokyo, Japan
| | - Motonari Kondo
- Department of Molecular Immunology, Toho University School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Cloning of Mn-SOD gene and its mRNA expression difference and antioxidant enzyme activities under hypoxia stress of cobia Rachycentron canadum. Mol Biol Rep 2021; 48:6897-6909. [PMID: 34453674 DOI: 10.1007/s11033-021-06692-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/24/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Environmental hypoxia affects the survival and development of organisms. It is also an important environmental factor that leads to oxidative damage. Hypoxia is a condition in which tissues are deprived of oxygen; reoxygenation is the phenomenon in which hypoxic tissues are exposed to oxygen. Hypoxia-reoxygenation is vital in pathogenesis, where the production of reactive oxygen species and antioxidant disparity significantly contribute to disease progression, and it is one of the most common physiological stressors in the aquaculture industry. METHODS AND RESULTS In this study, the full length of complementary DNA (cDNA) of the manganese superoxide dismutase (Mn-SOD) gene of healthy cobia Rachycentron canadum was analysed using rapid amplification of cDNA ends. The real-time quantitative Polymerase Chain Reaction was used to measure the expression levels of Mn-SOD mRNAs in various tissues (heart, muscle, brain, liver, kidney, gill, intestine, and spleen). The 2-ΔΔCT method was used to performed the expression analysis. The experimental data were analysed using SPSS ver. 19.0 ( https://spss.software.informer.com/19.0/ ). P < 0.05 and P < 0.01 were set as significant differences. The values were articulated as mean ± standard deviation. The Mn-SOD gene cDNA sequence was 1209 bp long, including a 684 bp open reading frame, 42 bp 5'UTR and 483 bp 3'UTR, encoding 227 amino acids. Under hypoxia-reoxygen stress, the expression of Mn-SOD in brain tissue was significantly lower than in the control group after 8 h of reoxygenation and higher than the control group after 24 h. Hypoxia and subsequent reoxygenation triggered a disturbance in antioxidant homeostasis, displayed in the modification of GPx expression/activity in the liver: GPx was improved. CONCLUSIONS These results provide valuable information on the role of Mn-SOD regulation in oxidative stress caused by hypoxia.
Collapse
|
11
|
Kurokawa H, Ito H, Matsui H. Porphylipoprotein Accumulation and Porphylipoprotein Photodynamic Therapy Effects Involving Cancer Cell-Specific Cytotoxicity. Int J Mol Sci 2021; 22:ijms22147306. [PMID: 34298933 PMCID: PMC8305091 DOI: 10.3390/ijms22147306] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/03/2021] [Accepted: 07/05/2021] [Indexed: 12/01/2022] Open
Abstract
In photodynamic therapy (PDT) for neoplasms, photosensitizers selectively accumulate in cancer tissue. Upon excitation with light of an optimal wavelength, the photosensitizer and surrounding molecules generate reactive oxygen species, resulting in cancer cell-specific cytotoxicity. Porphylipoprotein (PLP) has a porphyrin-based nanostructure. The porphyrin moiety of PLP is quenched because of its structure. When PLP is disrupted, the stacked porphyrins are separated into single molecules and act as photosensitizers. Unless PLP is disrupted, there is no photosensitive disorder in normal tissues. PLP can attenuate the photosensitive disorder compared with other photosensitizers and is ideal for use as a photosensitizer. However, the efficacy of PLP has not yet been evaluated. In this study, the mechanism of cancer cell-specific accumulation of PLP and its cytotoxic effect on cholangiocarcinoma cells were evaluated. The effects were investigated on normal and cancer-like mutant cells. The cytotoxicity effect of PLP PDT in cancer cells was significantly stronger than in normal cells. In addition, reactive oxygen species regulated intracellular PLP accumulation. The cytotoxic effects were also investigated using a cholangiocarcinoma cell line. The cytotoxicity of PLP PDT was significantly higher than that of laserphyrin-based PDT, a conventional type of PDT. PLP PDT could also inhibit tumor growth in vivo.
Collapse
Affiliation(s)
- Hiromi Kurokawa
- Faculty of Medicine, University of Tsukuba, Ibaraki 305-8577, Japan
- Correspondence: ; Tel.: +81-29-853-3466
| | - Hiromu Ito
- Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan; (H.I.); (H.M.)
| | - Hirofumi Matsui
- Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan; (H.I.); (H.M.)
| |
Collapse
|
12
|
Yang Y, Zhang G, Guo F, Li Q, Luo H, Shu Y, Shen Y, Gan J, Xu L, Yang H. Mitochondrial UQCC3 Modulates Hypoxia Adaptation by Orchestrating OXPHOS and Glycolysis in Hepatocellular Carcinoma. Cell Rep 2021; 33:108340. [PMID: 33147459 DOI: 10.1016/j.celrep.2020.108340] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 08/07/2020] [Accepted: 10/09/2020] [Indexed: 02/08/2023] Open
Abstract
Bioenergetic reprogramming during hypoxia adaption is critical to promote hepatocellular carcinoma (HCC) growth and progression. However, the mechanism underlying the orchestration of mitochondrial OXPHOS (oxidative phosphorylation) and glycolysis in hypoxia is not fully understood. Here, we report that mitochondrial UQCC3 (C11orf83) expression increases in hypoxia and correlates with the poor prognosis of HCC patients. Loss of UQCC3 impairs HCC cell proliferation in hypoxia in vitro and in vivo. Mechanistically, UQCC3 forms a positive feedback loop with mitochondrial reactive oxygen species (ROS) to sustain UQCC3 expression and ROS generation in hypoxic HCC cells and subsequently maintains mitochondrial structure and function and stabilizes HIF-1α expression to enhance glycolysis under hypoxia. Thus, UQCC3 plays an indispensable role for bioenergetic reprogramming of HCC cells during hypoxia adaption by simultaneously regulating OXPHOS and glycolysis. The positive feedback between UQCC3 and ROS indicates a self-modulating model within mitochondria that initiates the adaptation of HCC to hypoxic stress.
Collapse
Affiliation(s)
- Yun Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Guimin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Fengzhu Guo
- Lung Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qiqi Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Hui Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yuge Shen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Jia Gan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Lin Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Hanshuo Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China; Experimental and Research Animal Institute, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
Oxidative Stress: A Possible Trigger for Pelvic Organ Prolapse. J Immunol Res 2020; 2020:3791934. [PMID: 32953891 PMCID: PMC7481916 DOI: 10.1155/2020/3791934] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/06/2020] [Accepted: 08/21/2020] [Indexed: 01/23/2023] Open
Abstract
Pelvic organ prolapse is a frequent health problem in women, encountered worldwide, its physiopathology being still incompletely understood. The integrity of the pelvic-supportive structures is a key element that prevents the prolapse of the pelvic organs. Numerous researchers have underlined the role of connective tissue molecular changes in the pathogenesis of pelvic organ prolapse and have raised the attention upon oxidative stress as an important element involved in its appearance. The advancements made over the years in terms of molecular biology have allowed researchers to investigate how the constituent elements of the pelvic-supportive structures react in conditions of oxidative stress. The purpose of this paper is to underline the importance of oxidative stress in the pathogenesis of pelvic organ prolapse, as well as to highlight the main oxidative stress molecular changes that appear at the level of the pelvic-supportive structures. Sustained mechanical stress is proven to be a key factor in the appearance of pelvic organ prolapse, correlating with increased levels of free radicals production and mitochondrial-induced fibroblasts apoptosis, the rate of cellular apoptosis depending on the intensity of the mechanical stress, and the period of time the mechanical stress is applied. Oxidative stress hinders normal cellular signaling pathways, as well as different important cellular components like proteins, lipids, and cellular DNA, therefore significantly interfering with the process of collagen and elastin synthesis.
Collapse
|
14
|
Sharma P, Bharat, Dogra N, Singh S. Small Regulatory Molecules Acting Big in Cancer: Potential Role of Mito-miRs in Cancer. Curr Mol Med 2020; 19:621-631. [PMID: 31340735 DOI: 10.2174/1566524019666190723165357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/08/2019] [Accepted: 07/15/2019] [Indexed: 12/13/2022]
Abstract
MicroRNAs [miRNAs] are short, non-coding, single stranded RNA molecules regulating gene expression of their targets at the posttranscriptional level by either degrading mRNA or by inhibiting translation. Previously, miRNAs have been reported to be present inside the mitochondria and these miRNAs have been termed as mito-miRs. Origin of these mito-miRs may either be from mitochondrial genome or import from nucleus. The second class of mito-miRs makes it important to unravel the involvement of miRNAs in crosstalk between nucleus and mitochondria. Since miRNAs are involved in various physiological processes, their deregulation is often associated with disease progression, including cancer. The current review focuses on the involvement of miRNAs in different mitochondrial mediated processes. It also highlights the importance of exploring the interaction of miRNAs with mitochondrial genome, which may lead to the development of small regulatory RNA based therapeutic options.
Collapse
Affiliation(s)
- Praveen Sharma
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Mansa Road, Bathinda 151001, Punjab, India
| | - Bharat
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Mansa Road, Bathinda 151001, Punjab, India
| | - Nilambra Dogra
- Centre for Systems Biology and Bioinformatics, Panjab University, Sector-25, Chandigarh 160014, India
| | - Sandeep Singh
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Mansa Road, Bathinda 151001, Punjab, India
| |
Collapse
|
15
|
Baiula M, Greco R, Ferrazzano L, Caligiana A, Hoxha K, Bandini D, Longobardi P, Spampinato S, Tolomelli A. Integrin-mediated adhesive properties of neutrophils are reduced by hyperbaric oxygen therapy in patients with chronic non-healing wound. PLoS One 2020; 15:e0237746. [PMID: 32810144 PMCID: PMC7433869 DOI: 10.1371/journal.pone.0237746] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/31/2020] [Indexed: 12/30/2022] Open
Abstract
In recent years, several studies suggested that the ability of hyperbaric oxygen therapy (HBOT) to promote healing in patients with diabetic ulcers and chronic wounds is due to the reduction of inflammatory cytokines and to a significant decrease in neutrophils recruitment to the damaged area. α4 and β2 integrins are receptors mediating the neutrophil adhesion to the endothelium and the comprehension of the effects of hyperbaric oxygenation on their expression and functions in neutrophils could be of great importance for the design of novel therapeutic protocols focused on anti-inflammatory agents. In this study, the α4 and β2 integrins' expression and functions have been evaluated in human primary neutrophils obtained from patients with chronic non-healing wounds and undergoing a prolonged HBOT (150 kPa per 90 minutes). The effect of a peptidomimetic α4β1 integrin antagonist has been also analyzed under these conditions. A statistically significant decrease (68%) in β2 integrin expression on neutrophils was observed during the treatment with HBO and maintained one month after the last treatment, while α4 integrin levels remained unchanged. However, cell adhesion function of both neutrophilic integrins α4β1 and β2 was significantly reduced 70 and 67%, respectively), but α4β1 integrin was still sensitive to antagonist inhibition in the presence of fibronectin, suggesting that a combined therapy between HBOT and integrin antagonists could have greater antinflammatory efficacy.
Collapse
Affiliation(s)
- Monica Baiula
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Roberto Greco
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum -University of Bologna, Bologna, Italy
| | - Lucia Ferrazzano
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum -University of Bologna, Bologna, Italy
| | - Alberto Caligiana
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | | | | | | | - Santi Spampinato
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Alessandra Tolomelli
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum -University of Bologna, Bologna, Italy
| |
Collapse
|
16
|
Zhao H, Narasimhan P, Kalish F, Wong RJ, Stevenson DK. Dysregulation of hypoxia-inducible factor-1α (Hif1α) expression in the Hmox1-deficient placenta. Placenta 2020; 99:108-116. [PMID: 32784053 PMCID: PMC7549641 DOI: 10.1016/j.placenta.2020.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/15/2020] [Indexed: 12/19/2022]
Abstract
Introduction Severe hypoxia exists in placentas during early pregnancy, with reoxygenation during mid-gestation. Hypoxia-inducible factor-1α (Hif1α), an oxygen sensor, initiates placental vascular development. We have shown that the placental vasculature in Hmox1-deficient (Hmox1+/−, Het) pregnancies is impaired, with morphological defects similar to Hif1α-deficient placentas. Materials and methods Whole wild-type (WT) and Het mouse placentas were collected at E8.5 (1%–3% O2) and E9.5–15.5 (8%–10% O2). mRNA levels were determined using real-time RT-PCR or PCR arrays and protein levels using Western blot. Bone marrow-derived macrophages (BMDMs) from WT, Het, and Hmox1 knockout (KO) mice, representing different Hmox1 cellular levels, were generated to study the role of Hmox1 on Hif1α ′s response to hypoxia-reoxygenation and gestational age-specific placental lysates. Results Hif1α was expressed in WT and Het placentas throughout gestation, with protein levels peaking at E8.5 and mRNA levels significantly upregulated from E9.5–E13.5, but significantly lower in Het placentas. Genes associated with angiogenesis (Vegfa, Vegfr1, Mmp2, Cxcl12, Angpt1, Nos3), antioxidants (Sod1, Gpx1), and transcription factors (Ap2, Bach1, Nrf2) were significantly different in Het placentas. In response to in vitro hypoxia-reoxygenation and to WT or Het placental lysates, Hif1α transcription was lower in Het and Hmox1 KO BMDMs compared with WT BMDMs. Discussion These findings suggest that deficiencies in Hmox1 underlie the insufficient placental Hif1α response to hypoxia-reoxygenation during gestation and subsequently impair downstream placental vascular formation. Therefore, a dysregulation of Hif1α expression caused by any genetic defect or environmental influence in early pregnancy could be the root cause of pregnancy disorders. Expression of Hif1α in wild-type (WT) placentas is gestational age-dependent. Hif1α expression is reduced in Hmox1-deficient placentas. Expression of angiogenic genes is altered in Hmox1-deficient placentas. Hypoxia-reoxygenation induces a differential expression of Hif1α in cells. Adding placental lysates dysregulates expression of Hif1α in Hmox1-deficient cells.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Purnima Narasimhan
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Flora Kalish
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
17
|
Yun HR, Jo YH, Kim J, Shin Y, Kim SS, Choi TG. Roles of Autophagy in Oxidative Stress. Int J Mol Sci 2020; 21:ijms21093289. [PMID: 32384691 PMCID: PMC7246723 DOI: 10.3390/ijms21093289] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a catabolic process for unnecessary or dysfunctional cytoplasmic contents by lysosomal degradation pathways. Autophagy is implicated in various biological processes such as programmed cell death, stress responses, elimination of damaged organelles and development. The role of autophagy as a crucial mediator has been clarified and expanded in the pathological response to redox signalling. Autophagy is a major sensor of the redox signalling. Reactive oxygen species (ROS) are highly reactive molecules that are generated as by-products of cellular metabolism, principally by mitochondria. Mitochondrial ROS (mROS) are beneficial or detrimental to cells depending on their concentration and location. mROS function as redox messengers in intracellular signalling at physiologically low level, whereas excessive production of mROS causes oxidative damage to cellular constituents and thus incurs cell death. Hence, the balance of autophagy-related stress adaptation and cell death is important to comprehend redox signalling-related pathogenesis. In this review, we attempt to provide an overview the basic mechanism and function of autophagy in the context of response to oxidative stress and redox signalling in pathology.
Collapse
Affiliation(s)
- Hyeong Rok Yun
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
| | - Yong Hwa Jo
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Jieun Kim
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (S.S.K.); (T.G.C.); Tel.: +82-2-961-0524 (S.S.K.); +82-2-961-0287 (T.G.C.)
| | - Tae Gyu Choi
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (S.S.K.); (T.G.C.); Tel.: +82-2-961-0524 (S.S.K.); +82-2-961-0287 (T.G.C.)
| |
Collapse
|
18
|
Wang ZN, Su RN, Yang BY, Yang KX, Yang LF, Yan Y, Chen ZG. Potential Role of Cellular Senescence in Asthma. Front Cell Dev Biol 2020; 8:59. [PMID: 32117985 PMCID: PMC7026390 DOI: 10.3389/fcell.2020.00059] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/22/2020] [Indexed: 12/18/2022] Open
Abstract
Cellular senescence is a complicated process featured by irreversible cell cycle arrest and senescence-associated secreted phenotype (SASP), resulting in accumulation of senescent cells, and low-grade inflammation. Cellular senescence not only occurs during the natural aging of normal cells, but also can be accelerated by various pathological factors. Cumulative studies have shown the role of cellular senescence in the pathogenesis of chronic lung diseases including chronic obstructive pulmonary diseases (COPD) and idiopathic pulmonary fibrosis (IPF) by promoting airway inflammation and airway remodeling. Recently, great interest has been raised in the involvement of cellular senescence in asthma. Limited but valuable data has indicated accelerating cellular senescence in asthma. This review will compile current findings regarding the underlying relationship between cellular senescence and asthma, mainly through discussing the potential mechanisms of cellular senescence in asthma, the impact of senescent cells on the pathobiology of asthma, and the efficiency and feasibility of using anti-aging therapies in asthmatic patients.
Collapse
Affiliation(s)
- Zhao-Ni Wang
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ruo-Nan Su
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bi-Yuan Yang
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke-Xin Yang
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Fen Yang
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Yan
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Zhuang-Gui Chen
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
19
|
Mijiritsky E, Gardin C, Ferroni L, Lacza Z, Zavan B. Albumin-impregnated bone granules modulate the interactions between mesenchymal stem cells and monocytes under in vitro inflammatory conditions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110678. [PMID: 32204105 DOI: 10.1016/j.msec.2020.110678] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/22/2019] [Accepted: 01/18/2020] [Indexed: 12/17/2022]
Abstract
Bone regeneration around newly implanted biomaterials is a complex process, which in its early phases involves the interactions between Mesenchymal Stem Cells (MSCs) and immune cells. The response of these cells to the biomaterial depends both on the local microenvironment and on the characteristics of the inserted bone substitute. In this work, bone allografts impregnated with albumin are loaded with a co-culture of human MSCs and monocytes; bone granules without albumin are used for comparison. Co-cultures are contextually treated with pro-inflammatory cytokines to simulate the inflammatory milieu naturally present during the bone regeneration process. As revealed by microscopic images, albumin-impregnated bone granules promote adhesion and interactions between cells populations. Compared to control granules, albumin coating diminishes reactive species production by cells. This reduced oxidative stress may be attributable to antioxidant properties of albumin, and it is also reflected in the mitigated gene expression of mitochondrial electron transport chain complexes, where most intracellular reactive molecules are generated. MSCs-monocytes co-cultured onto albumin-impregnated bone granules additionally release higher amounts of immunomodulatory cytokines and growth factors. In summary, this work demonstrates that impregnation of bone granules with albumin positively modulates the interactions between MSCs and immune cells, consequently influencing their mutual activities and immunomodulatory functions.
Collapse
Affiliation(s)
- Eitan Mijiritsky
- Department of Otolaryngology, Head and Neck and Maxillofacial Surgery, Sackler Faculty of Medicine, Tel-Aviv Sourasky Medical Center, 64239 Tel Aviv, Israel
| | - Chiara Gardin
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy.
| | - Letizia Ferroni
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy
| | - Zsombor Lacza
- Institute of Clinical Experimental Research, Semmelweis University, 1094 Budapest, Hungary
| | - Barbara Zavan
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy.
| |
Collapse
|
20
|
Parmakhtiar B, Burger RA, Kim JH, Fruehauf JP. HIF Inactivation of p53 in Ovarian Cancer Can Be Reversed by Topotecan, Restoring Cisplatin and Paclitaxel Sensitivity. Mol Cancer Res 2019; 17:1675-1686. [PMID: 31088908 DOI: 10.1158/1541-7786.mcr-18-1109] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/04/2019] [Accepted: 05/10/2019] [Indexed: 11/16/2022]
Abstract
Ovarian cancer growth under hypoxic conditions results in hypoxia-inducible factor-1α (HIF1α) stabilization. HIF1α is an adverse prognostic factor that may contribute to worse outcomes via its capacity to bind to p53, potentially blocking p53-mediated apoptosis. We determined whether HIF1α-p53 binding occurred in hypoxic ovarian cancer cell lines, and if this blocked p53 transcriptional activity. Topotecan (TPT), used in the treatment of ovarian cancer, inhibits HIF1α translation via a topoisomerase-1 (TOPO1)-dependent mechanism. We examined if TPT knockdown of HIF1α restored p53 transcriptional function. TPT effects on HIF1α and p53-related transcriptional targets were assessed by PCR. Associations between TPT effects and TOPO1 expression levels were examined by Western blots and knockdown by siRNA. RNA-binding protein immunoprecipitation was used to assess if TOPO1 was resident on HIF1α mRNA. We determined if sublethal doses of TPT, used to knockdown HIF1α, reversed hypoxia-related cisplatin and paclitaxel resistance (XTT assay). Flow cytometry was used to assess HIF1α-mediated upregulation of ABCB1 and ABCB5 efflux pump expression. We found that HIF1α binding to, and inhibition of, p53 transcriptional activity in hypoxic ovarian cancer cells was associated with drug resistance. TPT-mediated downregulation of HIF1α in hypoxic cells required TOPO1 resident on HIF1α mRNA, restored p53 transcriptional activity, downregulated ABCB1/ABCB5 cell surface expression, and reversed hypoxia-related cisplatin and paclitaxel resistance. IMPLICATIONS: TPT-mediated reduction of HIF1α accumulation in hypoxic ovarian cancer cell lines restores p53 tumor-suppressor function, offering a novel approach to reverse chemoresistance. Further clinical investigation is warranted.
Collapse
Affiliation(s)
- Basmina Parmakhtiar
- Department of Biological Chemistry, University of California Irvine, Irvine, California
| | - Robert A Burger
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jai-Hyun Kim
- Chao Family Comprehensive Cancer Center, University of California Irvine, Orange, California
| | - John P Fruehauf
- Department of Biological Chemistry, University of California Irvine, Irvine, California. .,Chao Family Comprehensive Cancer Center, University of California Irvine, Orange, California
| |
Collapse
|
21
|
Molecular characterization of manganese superoxide dismutase (MnSOD) from sterlet Acipenser ruthenus and its responses to Aeromonas hydrophila challenge and hypoxia stress. Comp Biochem Physiol A Mol Integr Physiol 2019; 234:68-76. [PMID: 30999108 DOI: 10.1016/j.cbpa.2019.04.010] [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: 12/14/2018] [Revised: 02/26/2019] [Accepted: 04/11/2019] [Indexed: 01/14/2023]
Abstract
A novel gene encoding the mitochondrial manganese superoxide dismutase from sterlet Acipenser ruthenus (Ar-MnSOD) was cloned. The full-length cDNA of MnSOD was of 1040 bp with a 672 bp open reading frame encoding 224 amino acids and the deduced amino acid sequence was located in mitochondria. Sequence comparison analysis showed that Ar-MnSOD was highly similar to MnSODs of invertebrates and vertebrates, especially those of freshwater Cyprinidae fishes and mammals. Phylogenetic analysis revealed that Ar-MnSOD was distant from MnSODs of other fishes and belonged to the family of mitochondrial MnSODs (mMnSOD). Consistently, Ar-MnSOD was located in mitochondria. The 3D structure of Ar-MnSOD was predicted and the overall structure was similar to that of MnSODs of humans and the bay scallop Argopecten irradians. In addition, mRNA of Ar-MnSOD was detected to extensively express in all tissues, with the highest level in brain and liver. Spleen and head kidney inoculation of Aeromonas hydrophila led to a significant up-regulation of Ar-MnSOD transcript levels. Also, hypoxia induced a transient increase in transcription of Ar-MnSOD in the gills, but not in the heart and brain, suggesting metabolic depression in these vital organs. The results also implied the anti-hypoxia properties of Ar-MnSOD in the related tissues and proved that Ar-MnSOD was involved in the stress response and (anti) oxidative processes triggered by hypoxia. The results indicated that Ar-MnSOD is induced upon A. hydrophila infection and hypoxia, consistent with its role in host immune and stress-induced anti-oxidative responses.
Collapse
|
22
|
Abstract
SIGNIFICANCE Hydroxyproline is a structurally and physiologically important imino acid in animals. It is provided from diets and endogenous synthesis, and its conversion into glycine enhances the production of glutathione, DNA, heme, and protein. Furthermore, oxidation of hydroxyproline by hydroxyproline oxidase (OH-POX) plays an important role in cell antioxidative reactions, survival, and homeostasis. Understanding the mechanisms whereby hydroxyproline participates in metabolism and cell signaling can improve the nutrition and health of animals and humans. Recent Advances: Hydroxyproline is highly abundant in milk and is utilized for renal synthesis of glycine to support neonatal growth, development, and survival. The oxidation of hydroxyproline by mitochondrial OH-POX generates reactive oxygen species (ROS). Enhanced ROS production contributes to the regulation of oxidative defense, apoptosis, angiogenesis, tumorigenesis, hypoxic responses, and cell survival in animals. CRITICAL ISSUES Although dietary hydroxyproline enters the portal circulation, its utilization by the portal-drained viscera is unknown. Pathways for hydroxyproline metabolism and their regulation at the molecular, cellular, and whole-body levels remain to be defined. Furthermore, the mechanisms responsible for hydroxyproline-derived ROS and related metabolites to induce cell survival or apoptosis are unknown. FUTURE DIRECTIONS Interorgan metabolism of hydroxyproline (including synthesis, catabolism, and flux) in animals must be quantified using isotope technologies. Efforts should also be directed toward studying dietary, hormonal, and epigenetic regulation of OH-POX expression at transcriptional and translational levels. Another emerging research need is to understand the roles of cellular redox and signaling networks involving both ROS and Δ1-pyrroline-3-hydroxy-5-carboxylate in nutrition, health, and disease.
Collapse
Affiliation(s)
- Zhenlong Wu
- 1 State Key Laboratory of Animal Nutrition, China Agricultural University , Beijing, China
| | - Yongqing Hou
- 2 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University , Wuhan, China
| | - Zhaolai Dai
- 1 State Key Laboratory of Animal Nutrition, China Agricultural University , Beijing, China
| | - Chien-An A Hu
- 2 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University , Wuhan, China .,3 Department of Biochemistry and Molecular Biology, University of New Mexico , Health Sciences Center, Albuquerque, New Mexico
| | - Guoyao Wu
- 1 State Key Laboratory of Animal Nutrition, China Agricultural University , Beijing, China .,2 Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University , Wuhan, China .,4 Department of Animal Science, Texas A&M University , College Station, Texas
| |
Collapse
|
23
|
Al Tameemi W, Dale TP, Al-Jumaily RMK, Forsyth NR. Hypoxia-Modified Cancer Cell Metabolism. Front Cell Dev Biol 2019; 7:4. [PMID: 30761299 PMCID: PMC6362613 DOI: 10.3389/fcell.2019.00004] [Citation(s) in RCA: 301] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/10/2019] [Indexed: 12/20/2022] Open
Abstract
While oxygen is critical to the continued existence of complex organisms, extreme levels of oxygen within a system, known as hypoxia (low levels of oxygen) and hyperoxia (excessive levels of oxygen), potentially promote stress within a defined biological environment. The consequences of tissue hypoxia, a result of a defective oxygen supply, vary in response to the gravity, extent and environment of the malfunction. Persistent pathological hypoxia is incompatible with normal biological functions, and as a result, multicellular organisms have been compelled to develop both organism-wide and cellular-level hypoxia solutions. Both direct, including oxidative phosphorylation down-regulation and inhibition of fatty-acid desaturation, and indirect processes, including altered hypoxia-sensitive transcription factor expression, facilitate the metabolic modifications that occur in response to hypoxia. Due to the dysfunctional vasculature associated with large areas of some cancers, sections of these tumors continue to develop in hypoxic environments. Crucial to drug development, a robust understanding of the significance of these metabolism changes will facilitate our understanding of cancer cell survival. This review defines our current knowledge base of several of the hypoxia-instigated modifications in cancer cell metabolism and exemplifies the correlation between metabolic change and its support of the hypoxic-adapted malignancy.
Collapse
Affiliation(s)
- Wafaa Al Tameemi
- Faculty of Medicine and Health Sciences, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| | - Tina P. Dale
- Faculty of Medicine and Health Sciences, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| | - Rakad M. Kh Al-Jumaily
- Faculty of Medicine and Health Sciences, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
- Department of Biology, College of Science, University of Baghdad, Baghdad, Iraq
| | - Nicholas R. Forsyth
- Faculty of Medicine and Health Sciences, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| |
Collapse
|
24
|
Bailey PSJ, Nathan JA. Metabolic Regulation of Hypoxia-Inducible Transcription Factors: The Role of Small Molecule Metabolites and Iron. Biomedicines 2018; 6:biomedicines6020060. [PMID: 29772792 PMCID: PMC6027492 DOI: 10.3390/biomedicines6020060] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 02/02/2023] Open
Abstract
Hypoxia-inducible transcription factors (HIFs) facilitate cellular adaptations to low-oxygen environments. However, it is increasingly recognised that HIFs may be activated in response to metabolic stimuli, even when oxygen is present. Understanding the mechanisms for the crosstalk that exists between HIF signalling and metabolic pathways is therefore important. This review focuses on the metabolic regulation of HIFs by small molecule metabolites and iron, highlighting the latest studies that explore how tricarboxylic acid (TCA) cycle intermediates, 2-hydroxyglutarate (2-HG) and intracellular iron levels influence the HIF response through modulating the activity of prolyl hydroxylases (PHDs). We also discuss the relevance of these metabolic pathways in physiological and disease contexts. Lastly, as PHDs are members of a large family of 2-oxoglutarate (2-OG) dependent dioxygenases that can all respond to metabolic stimuli, we explore the broader role of TCA cycle metabolites and 2-HG in the regulation of 2-OG dependent dioxygenases, focusing on the enzymes involved in chromatin remodelling.
Collapse
Affiliation(s)
- Peter S J Bailey
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, Cambridge CB2 0XY, UK.
| | - James A Nathan
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, Cambridge CB2 0XY, UK.
| |
Collapse
|
25
|
Pak O, Sydykov A, Kosanovic D, Schermuly RT, Dietrich A, Schröder K, Brandes RP, Gudermann T, Sommer N, Weissmann N. Lung Ischaemia-Reperfusion Injury: The Role of Reactive Oxygen Species. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:195-225. [PMID: 29047088 DOI: 10.1007/978-3-319-63245-2_12] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Lung ischaemia-reperfusion injury (LIRI) occurs in many lung diseases and during surgical procedures such as lung transplantation. The re-establishment of blood flow and oxygen delivery into the previously ischaemic lung exacerbates the ischaemic injury and leads to increased microvascular permeability and pulmonary vascular resistance as well as to vigorous activation of the immune response. These events initiate the irreversible damage of the lung with subsequent oedema formation that can result in systemic hypoxaemia and multi-organ failure. Alterations in the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) have been suggested as crucial mediators of such responses during ischaemia-reperfusion in the lung. Among numerous potential sources of ROS/RNS within cells, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, xanthine oxidases, nitric oxide synthases and mitochondria have been investigated during LIRI. Against this background, we aim to review here the extensive literature about the ROS-mediated cellular signalling during LIRI, as well as the effectiveness of antioxidants as treatment option for LIRI.
Collapse
Affiliation(s)
- Oleg Pak
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Akylbek Sydykov
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Djuro Kosanovic
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Alexander Dietrich
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Goethestraße 33, 80336, Munich, Germany
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Thomas Gudermann
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Goethestraße 33, 80336, Munich, Germany
| | - Natascha Sommer
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany.
| |
Collapse
|
26
|
Ježek J, Cooper KF, Strich R. Reactive Oxygen Species and Mitochondrial Dynamics: The Yin and Yang of Mitochondrial Dysfunction and Cancer Progression. Antioxidants (Basel) 2018; 7:E13. [PMID: 29337889 PMCID: PMC5789323 DOI: 10.3390/antiox7010013] [Citation(s) in RCA: 295] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/02/2018] [Accepted: 01/09/2018] [Indexed: 12/11/2022] Open
Abstract
Mitochondria are organelles with a highly dynamic ultrastructure maintained by a delicate equilibrium between its fission and fusion rates. Understanding the factors influencing this balance is important as perturbations to mitochondrial dynamics can result in pathological states. As a terminal site of nutrient oxidation for the cell, mitochondrial powerhouses harness energy in the form of ATP in a process driven by the electron transport chain. Contemporaneously, electrons translocated within the electron transport chain undergo spontaneous side reactions with oxygen, giving rise to superoxide and a variety of other downstream reactive oxygen species (ROS). Mitochondrially-derived ROS can mediate redox signaling or, in excess, cause cell injury and even cell death. Recent evidence suggests that mitochondrial ultrastructure is tightly coupled to ROS generation depending on the physiological status of the cell. Yet, the mechanism by which changes in mitochondrial shape modulate mitochondrial function and redox homeostasis is less clear. Aberrant mitochondrial morphology may lead to enhanced ROS formation, which, in turn, may deteriorate mitochondrial health and further exacerbate oxidative stress in a self-perpetuating vicious cycle. Here, we review the latest findings on the intricate relationship between mitochondrial dynamics and ROS production, focusing mainly on its role in malignant disease.
Collapse
Affiliation(s)
- Jan Ježek
- Department of Molecular Biology, Rowan University Graduate School of Biomedical Sciences, Stratford, NJ 08084, USA.
| | - Katrina F Cooper
- Department of Molecular Biology, Rowan University Graduate School of Biomedical Sciences, Stratford, NJ 08084, USA.
| | - Randy Strich
- Department of Molecular Biology, Rowan University Graduate School of Biomedical Sciences, Stratford, NJ 08084, USA.
| |
Collapse
|
27
|
Abstract
Mitochondria are the power stations of the eukaryotic cell, using the energy released by the oxidation of glucose and other sugars to produce ATP. Electrons are transferred from NADH, produced in the citric acid cycle in the mitochondrial matrix, to oxygen by a series of large protein complexes in the inner mitochondrial membrane, which create a transmembrane electrochemical gradient by pumping protons across the membrane. The flow of protons back into the matrix via a proton channel in the ATP synthase leads to conformational changes in the nucleotide binding pockets and the formation of ATP. The three proton pumping complexes of the electron transfer chain are NADH-ubiquinone oxidoreductase or complex I, ubiquinone-cytochrome c oxidoreductase or complex III, and cytochrome c oxidase or complex IV. Succinate dehydrogenase or complex II does not pump protons, but contributes reduced ubiquinone. The structures of complex II, III and IV were determined by x-ray crystallography several decades ago, but complex I and ATP synthase have only recently started to reveal their secrets by advances in x-ray crystallography and cryo-electron microscopy. The complexes I, III and IV occur to a certain extent as supercomplexes in the membrane, the so-called respirasomes. Several hypotheses exist about their function. Recent cryo-electron microscopy structures show the architecture of the respirasome with near-atomic detail. ATP synthase occurs as dimers in the inner mitochondrial membrane, which by their curvature are responsible for the folding of the membrane into cristae and thus for the huge increase in available surface that makes mitochondria the efficient energy plants of the eukaryotic cell.
Collapse
Affiliation(s)
- Joana S Sousa
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Edoardo D'Imprima
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Janet Vonck
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
| |
Collapse
|
28
|
Bullone M, Lavoie JP. The Contribution of Oxidative Stress and Inflamm-Aging in Human and Equine Asthma. Int J Mol Sci 2017; 18:ijms18122612. [PMID: 29206130 PMCID: PMC5751215 DOI: 10.3390/ijms18122612] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
Aging is associated with a dysregulation of the immune system, leading to a general pro-inflammatory state of the organism, a process that has been named inflamm-aging. Oxidative stress has an important role in aging and in the regulation of immune responses, probably playing a role in the development of age-related diseases. The respiratory system function physiologically declines with the advancement of age. In elderly asthmatic patients, this may contribute to disease expression. In this review, we will focus on age-related changes affecting the immune system and in respiratory structure and function that could contribute to asthma occurrence, and/or clinical presentation in the elderly. Also, naturally occurring equine asthma will be discussed as a possible model for studying the importance of oxidative stress and immun-aging/inflamm-aging in humans.
Collapse
Affiliation(s)
- Michela Bullone
- Department of Clinical and Biological Sciences, University of Turin, AUO San Luigi Gonzaga, Regione Gonzole 10, 10043 Orbassano, Italy.
| | - Jean-Pierre Lavoie
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, 3200 Rue Sicotte, St-Hyacinthe, QC J2S 2M2, Canada.
| |
Collapse
|
29
|
Taylor CT, Colgan SP. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat Rev Immunol 2017; 17:774-785. [PMID: 28972206 PMCID: PMC5799081 DOI: 10.1038/nri.2017.103] [Citation(s) in RCA: 425] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immunological niches are focal sites of immune activity that can have varying microenvironmental features. Hypoxia is a feature of physiological and pathological immunological niches. The impact of hypoxia on immunity and inflammation can vary depending on the microenvironment and immune processes occurring in a given niche. In physiological immunological niches, such as the bone marrow, lymphoid tissue, placenta and intestinal mucosa, physiological hypoxia controls innate and adaptive immunity by modulating immune cell proliferation, development and effector function, largely via transcriptional changes driven by hypoxia-inducible factor (HIF). By contrast, in pathological immunological niches, such as tumours and chronically inflamed, infected or ischaemic tissues, pathological hypoxia can drive tissue dysfunction and disease development through immune cell dysregulation. Here, we differentiate between the effects of physiological and pathological hypoxia on immune cells and the consequences for immunity and inflammation in different immunological niches. Furthermore, we discuss the possibility of targeting hypoxia-sensitive pathways in immune cells for the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Cormac T Taylor
- UCD Conway Institute, Systems Biology Ireland and the School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sean P Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, 80045 Colorado, USA
| |
Collapse
|
30
|
Xu T, Pang Q, Wang Y, Yan X. Betulinic acid induces apoptosis by regulating PI3K/Akt signaling and mitochondrial pathways in human cervical cancer cells. Int J Mol Med 2017; 40:1669-1678. [PMID: 29039440 PMCID: PMC5716432 DOI: 10.3892/ijmm.2017.3163] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 09/19/2017] [Indexed: 12/29/2022] Open
Abstract
Betulinic acid (BA), a potential anticancer compound, could induce apoptosis in human cervical cancer (HeLa) cells, but its mechanism has yet to be fully elucidated. The present study was focused on deciphering the detailed molecular mechanism of BA-induced apoptosis. In the present study, results indicated that BA was highly effective against HeLa cells via induction of time-dependent apoptosis, and the authors demonstrated that the BA treatment acted through downregulating a phosphatidylinositol 3-kinase (PI3K) subunit and suppressing the Akt phosphorylation at Thr308 and Ser473 after increasing the generation of intracellular reactive oxygen species. Then, BA induced cell cycle arrest at the G0/G1 phase, which was consistent with the cell cycle-related protein results in which BA significantly enhanced the expression of p27Kip and p21Waf1/Cip1 in HeLa cells. This target-specific inhibition was associated with mitochondrial apoptosis, as reflected by the increased expression of Bad and caspase-9, the generation of reactive oxygen species (ROS) and the decline in mitochondrial membrane potential. Moreover, preincubation of the cells with glutathione (antioxidant) blocked the process of apoptosis, prevented the phosphorylation of downstream substrates. These results established that ROS acted as a key factor to effect apoptosis by BA treatment in HeLa cells. Therefore, these findings demonstrated that BA induced apoptosis in HeLa cells by downregulating the expression of PI3K/Akt signaling molecules via ROS, and triggering a mitochondrial pathway.
Collapse
Affiliation(s)
- Tao Xu
- Alkali Soil Natural Environmental Science Center, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Qiuying Pang
- Alkali Soil Natural Environmental Science Center, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Yang Wang
- Alkali Soil Natural Environmental Science Center, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Xiufeng Yan
- Alkali Soil Natural Environmental Science Center, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
31
|
Baulies A, Montero J, Matías N, Insausti N, Terrones O, Basañez G, Vallejo C, Conde de La Rosa L, Martinez L, Robles D, Morales A, Abian J, Carrascal M, Machida K, Kumar DBU, Tsukamoto H, Kaplowitz N, Garcia-Ruiz C, Fernández-Checa JC. The 2-oxoglutarate carrier promotes liver cancer by sustaining mitochondrial GSH despite cholesterol loading. Redox Biol 2017; 14:164-177. [PMID: 28942194 PMCID: PMC5609874 DOI: 10.1016/j.redox.2017.08.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/21/2017] [Accepted: 08/24/2017] [Indexed: 12/15/2022] Open
Abstract
Cancer cells exhibit mitochondrial cholesterol (mt-cholesterol) accumulation, which contributes to cell death resistance by antagonizing mitochondrial outer membrane (MOM) permeabilization. Hepatocellular mt-cholesterol loading, however, promotes steatohepatitis, an advanced stage of chronic liver disease that precedes hepatocellular carcinoma (HCC), by depleting mitochondrial GSH (mGSH) due to a cholesterol-mediated impairment in mGSH transport. Whether and how HCC cells overcome the restriction of mGSH transport imposed by mt-cholesterol loading to support mGSH uptake remains unknown. Although the transport of mGSH is not fully understood, SLC25A10 (dicarboxylate carrier, DIC) and SLC25A11 (2-oxoglutarate carrier, OGC) have been involved in mGSH transport, and therefore we examined their expression and role in HCC. Unexpectedly, HCC cells and liver explants from patients with HCC exhibit divergent expression of these mitochondrial carriers, with selective OGC upregulation, which contributes to mGSH maintenance. OGC but not DIC downregulation by siRNA depleted mGSH levels and sensitized HCC cells to hypoxia-induced ROS generation and cell death as well as impaired cell growth in three-dimensional multicellular HCC spheroids, effects that were reversible upon mGSH replenishment by GSH ethyl ester, a membrane permeable GSH precursor. We also show that OGC regulates mitochondrial respiration and glycolysis. Moreover, OGC silencing promoted hypoxia-induced cardiolipin peroxidation, which reversed the inhibition of cholesterol on the permeabilization of MOM-like liposomes induced by Bax or Bak. Genetic OGC knockdown reduced the ability of tumor-initiating stem-like cells to induce liver cancer. These findings underscore the selective overexpression of OGC as an adaptive mechanism of HCC to provide adequate mGSH levels in the face of mt-cholesterol loading and suggest that OGC may be a novel therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Anna Baulies
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Joan Montero
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Nuria Matías
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Naroa Insausti
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Oihana Terrones
- Unidad de Biofísica (Centro Mixto Consejo Superior de Investigaciones Científicas-Universidad del País Vasco/Euskal Herriko Unibertsitatea), Universidad del País Vasco/Euskal Herriko Unibertsitatea, 48080 Bilbao, Spain
| | - Gorka Basañez
- Unidad de Biofísica (Centro Mixto Consejo Superior de Investigaciones Científicas-Universidad del País Vasco/Euskal Herriko Unibertsitatea), Universidad del País Vasco/Euskal Herriko Unibertsitatea, 48080 Bilbao, Spain
| | - Carmen Vallejo
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Laura Conde de La Rosa
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Laura Martinez
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - David Robles
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain
| | - Albert Morales
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain
| | - Joaquin Abian
- CSIC/UAB Proteomics Laboratory, IIBB-CSIC, 08036 Barcelona, Spain
| | | | - Keigo Machida
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA
| | - Dinesh B U Kumar
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA
| | - Hidekazu Tsukamoto
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA; Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Neil Kaplowitz
- University of Southern California Research Center for Liver Diseases, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain; Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA; University of Southern California Research Center for Liver Diseases, Keck School of Medicine, USC, Los Angeles, CA, USA.
| | - José C Fernández-Checa
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain; Liver Unit and Hospital Clínic i Provincial, IDIBAPS, and Centro de Investigación Biomédica en Red (CIBERehd), Spain; Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA; University of Southern California Research Center for Liver Diseases, Keck School of Medicine, USC, Los Angeles, CA, USA.
| |
Collapse
|
32
|
Blajszczak C, Bonini MG. Mitochondria targeting by environmental stressors: Implications for redox cellular signaling. Toxicology 2017; 391:84-89. [PMID: 28750850 DOI: 10.1016/j.tox.2017.07.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/22/2017] [Accepted: 07/21/2017] [Indexed: 01/07/2023]
Abstract
Mitochondria are cellular powerhouses as well as metabolic and signaling hubs regulating diverse cellular functions, from basic physiology to phenotypic fate determination. It is widely accepted that reactive oxygen species (ROS) generated in mitochondria participate in the regulation of cellular signaling, and that some mitochondria chronically operate at a high ROS baseline. However, it is not completely understood how mitochondria adapt to persistently high ROS states and to environmental stressors that disturb the redox balance. Here we will review some of the current concepts regarding how mitochondria resist oxidative damage, how they are replaced when excessive oxidative damage compromises function, and the effect of environmental toxicants (i.e. heavy metals) on the regulation of mitochondrial ROS (mtROS) production and subsequent impact.
Collapse
Affiliation(s)
- Chuck Blajszczak
- Departments of Medicine and Pathology, University of Illinois College of Medicine at Chicago, IL, USA
| | - Marcelo G Bonini
- Departments of Medicine and Pathology, University of Illinois College of Medicine at Chicago, IL, USA.
| |
Collapse
|
33
|
Huang C, Kuo W, Huang C, Lee T, Chen C, Peng W, Lu K, Yang C, Yu LC. Distinct cytoprotective roles of pyruvate and ATP by glucose metabolism on epithelial necroptosis and crypt proliferation in ischaemic gut. J Physiol 2017; 595:505-521. [PMID: 27121603 PMCID: PMC5233659 DOI: 10.1113/jp272208] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/24/2016] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS Intestinal ischaemia causes epithelial death and crypt dysfunction, leading to barrier defects and gut bacteria-derived septic complications. Enteral glucose protects against ischaemic injury; however, the roles played by glucose metabolites such as pyruvate and ATP on epithelial death and crypt dysfunction remain elusive. A novel form of necrotic death that involves the assembly and phosphorylation of receptor interacting protein kinase 1/3 complex was found in ischaemic enterocytes. Pyruvate suppressed epithelial cell death in an ATP-independent manner and failed to maintain crypt function. Conversely, replenishment of ATP partly restored crypt proliferation but had no effect on epithelial necroptosis in ischaemic gut. Our data argue against the traditional view of ATP as the main cytoprotective factor by glucose metabolism, and indicate a novel anti-necroptotic role of glycolytic pyruvate under ischaemic stress. ABSTRACT Mesenteric ischaemia/reperfusion induces epithelial death in both forms of apoptosis and necrosis, leading to villus denudation and gut barrier damage. It remains unclear whether programmed cell necrosis [i.e. receptor-interacting protein kinase (RIP)-dependent necroptosis] is involved in ischaemic injury. Previous studies have demonstrated that enteral glucose uptake by sodium-glucose transporter 1 ameliorated ischaemia/reperfusion-induced epithelial injury, partly via anti-apoptotic signalling and maintenance of crypt proliferation. Glucose metabolism is generally assumed to be cytoprotective; however, the roles played by glucose metabolites (e.g. pyruvate and ATP) on epithelial cell death and crypt dysfunction remain elusive. The present study aimed to investigate the cytoprotective effects exerted by distinct glycolytic metabolites in ischaemic gut. Wistar rats subjected to mesenteric ischaemia were enterally instilled glucose, pyruvate or liposomal ATP. The results showed that intestinal ischaemia caused RIP1-dependent epithelial necroptosis and villus destruction accompanied by a reduction in crypt proliferation. Enteral glucose uptake decreased epithelial cell death and increased crypt proliferation, and ameliorated mucosal histological damage. Instillation of cell-permeable pyruvate suppressed epithelial cell death in an ATP-independent manner and improved the villus morphology but failed to maintain crypt function. Conversely, the administration of liposomal ATP partly restored crypt proliferation but did not reduce epithelial necroptosis and histopathological injury. Lastly, glucose and pyruvate attenuated mucosal-to-serosal macromolecular flux and prevented enteric bacterial translocation upon blood reperfusion. In conclusion, glucose metabolites protect against ischaemic injury through distinct modes and sites, including inhibition of epithelial necroptosis by pyruvate and the promotion of crypt proliferation by ATP.
Collapse
Affiliation(s)
- Ching‐Ying Huang
- Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Wei‐Ting Kuo
- Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Chung‐Yen Huang
- Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Tsung‐Chun Lee
- Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
- Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Chin‐Tin Chen
- Department of Biochemical Science and TechnologyNational Taiwan UniversityTaipeiTaiwan
| | - Wei‐Hao Peng
- Graduate Institute of Anatomy and Cell BiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Kuo‐Shyan Lu
- Graduate Institute of Anatomy and Cell BiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Chung‐Yi Yang
- Department of Medical Imaging, E‐Da HospitalI‐Shou UniversityKaohsiungTaiwan
- Department of Medical ImagingNational Taiwan University HospitalTaipeiTaiwan
| | - Linda Chia‐Hui Yu
- Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| |
Collapse
|
34
|
Feuer S, Liu X, Donjacour A, Simbulan R, Maltepe E, Rinaudo P. Common and specific transcriptional signatures in mouse embryos and adult tissues induced by in vitro procedures. Reproduction 2016; 153:REP-16-0473. [PMID: 27799627 PMCID: PMC5411347 DOI: 10.1530/rep-16-0473] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 10/26/2016] [Indexed: 12/17/2022]
Abstract
Stressful environmental exposures incurred early in development can affect postnatal metabolic health and susceptibility to non-communicable diseases in adulthood, although the molecular mechanisms by which this occurs have yet to be elucidated. Here we use a mouse model to investigate how assorted in vitro exposures restricted exclusively to the preimplantation period affect transcription both acutely in embryos and long-term in subsequent offspring adult tissues, to determine if reliable transcriptional markers of in vitro stress are present at specific developmental time points and throughout development. Each in vitro fertilization or embryo culture environment led to a specific and unique blastocyst transcriptional profile, but we identified a common 18-gene and 9-pathway signature of preimplantation embryo manipulation that was present in all in vitro embryos irrespective of culture condition or method of fertilization. This fingerprint did not persist throughout development and there was no clear transcriptional cohesion between adult IVF offspring tissues or compared to their preceding embryos, indicating a tissue-specific impact of in vitro stress on gene expression. However, the transcriptional changes present in each IVF tissue were targeted by the same upstream transcriptional regulators, which provide insight as to how acute transcriptional responses to stressful environmental exposures might be preserved throughout development to influence adult gene expression.
Collapse
Affiliation(s)
- Sky Feuer
- S Feuer, Obstetrics and Gynecology, University of California, San Francisco, San Francisco, United States
| | - Xiaowei Liu
- X Liu, Obstetrics and Gynecology, University of California, San Francisco, San Francisco, United States
| | - Annemarie Donjacour
- A Donjacour, Obstetrics and Gynecology, University of California, San Francisco, San Francisco, United States
| | - Rhodel Simbulan
- R Simbulan, Obstetrics and Gynecology, University of California, San Francisco, San Francisco, United States
| | - Emin Maltepe
- E Maltepe, Obstetrics and Gynecology, University of California, San Francisco, San Francisco, United States
| | - Paolo Rinaudo
- P Rinaudo, Obstetrics and Gynecology, University of California, San Francisco, San Francisco, 94115, United States
| |
Collapse
|
35
|
Kamal M, D'Amora DR, Kubiseski TJ. Loss of hif-1 promotes resistance to the exogenous mitochondrial stressor ethidium bromide in Caenorhabditis elegans. BMC Cell Biol 2016; 17 Suppl 1:34. [PMID: 27618966 PMCID: PMC5020483 DOI: 10.1186/s12860-016-0112-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 09/06/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Mitochondrial dysfunction is one of the leading causes of neurological disorders in humans. Mitochondrial perturbations lead to adaptive mechanisms that include HIF-1 stabilization, though the consequences of increased levels of HIF-1 following mitochondrial stress remain poorly understood. RESULTS Using Caenorhabditis elegans, we show that a hif-1 loss-of-function mutation confers resistance towards the mitochondrial toxin ethidium bromide (EtBr) and suppresses EtBr-induced production of ROS. In mammals, the PD-related gene DJ-1 is known to act as a redox sensor to confer protection against antioxidants and mitochondrial inhibitors. A deletion mutant of the C. elegans homolog djr-1.1 also showed increased resistance to EtBr. Furthermore, our data implicates p38 MAP kinase as an indispensable factor for survival against mitochondrial stress in both hif-1 and djr-1.1 mutants. CONCLUSIONS We propose that EtBr-induced HIF-1 activates pathways that are antagonistic in conferring protection against EtBr toxicity and that blocking HIF-1 activity may promote survival in cells with compromised mitochondrial function.
Collapse
Affiliation(s)
- Muntasir Kamal
- Department of Biology, York University, Toronto, Canada.,Present address: Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | | | - Terrance J Kubiseski
- Department of Biology, York University, Toronto, Canada. .,Department of Neuroscience, York University, Toronto, Canada.
| |
Collapse
|
36
|
Cribbet MR, Logan RW, Edwards MD, Hanlon E, Bien Peek C, Stubblefield JJ, Vasudevan S, Ritchey F, Frank E. Circadian rhythms and metabolism: from the brain to the gut and back again. Ann N Y Acad Sci 2016; 1385:21-40. [PMID: 27589593 PMCID: PMC5428740 DOI: 10.1111/nyas.13188] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 07/05/2016] [Indexed: 12/21/2022]
Abstract
This paper focuses on the relationship between the circadian system and glucose metabolism. Research across the translational spectrum confirms the importance of the circadian system for glucose metabolism and offers promising clues as to when and why these systems go awry. In particular, basic research has started to clarify the molecular and genetic mechanisms through which the circadian system regulates metabolism. The study of human behavior, especially in the context of psychiatric disorders, such as bipolar disorder and major depression, forces us to see how inextricably linked mental health and metabolic health are. We also emphasize the remarkable opportunities for advancing circadian science through big data and advanced analytics. Advances in circadian research have translated into environmental and pharmacological interventions with tremendous therapeutic potential.
Collapse
Affiliation(s)
- Matthew R Cribbet
- Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ryan W Logan
- Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Mathew D Edwards
- Division of Neurobiology, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Erin Hanlon
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Clara Bien Peek
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jeremy J Stubblefield
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Fiona Ritchey
- Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ellen Frank
- Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
37
|
Zhang G, Mao J, Liang F, Chen J, Zhao C, Yin S, Wang L, Tang Z, Chen S. Modulated expression and enzymatic activities of Darkbarbel catfish, Pelteobagrus vachelli for oxidative stress induced by acute hypoxia and reoxygenation. CHEMOSPHERE 2016; 151:271-9. [PMID: 26945243 DOI: 10.1016/j.chemosphere.2016.02.072] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 05/14/2023]
Abstract
Large changes in oxygen availability in aquatic environments, ranging from anoxia through to hyperoxia, can lead to corresponding wide variation in the production of reactive oxygen species (ROS) by fish with aquatic respiration. In order to evaluate the effects of hypoxia and reoxygenation on oxidative stress in fish, the mRNA and protein expression of SODs (Cu/Zn-SOD and Mn-SOD) as well as indices (CP, LPO and MDA) and enzymatic activities (SOD, CAT, GPx, GR and GST) were analyzed in liver and brain tissues of Pelteobagrus vachelli. Predominant expression of PvSOD2 was detected in heart, brain, and liver. In contrast, PvSOD1 was highly expressed in liver. Based on the expression patterns of above parameters, we inferred that brain tissue of P. vachelli under 0.7 mg/L degree of acute hypoxia condition could experience hypometabolic states or no suffering stress, but brain tissue has effective mechanisms to minimize or prevent oxidative stress during the transition from hypoxia to reoxygenation. Our results also demonstrated an increased expression of SODs and enzymatic activities for oxidative stress in liver under hypoxic conditions, which supports the hypothesis that anticipatory preparation takes place in order to deal with the encountered oxidative stress during the recovery from hypoxia as proposed by M. Hermes-Lima. Therefore, this study will provide a clue to better understand the action mode of antioxidant genes and enzymes under oxidative stress in fish.
Collapse
Affiliation(s)
- Guosong Zhang
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, Nanjing, Jiangsu 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Jianqiang Mao
- Nanjing Institute of Fisheries Science, Nanjing, Jiangsu 210036, China
| | - Fenfei Liang
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, Nanjing, Jiangsu 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Jiawei Chen
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, Nanjing, Jiangsu 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Cheng Zhao
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, Nanjing, Jiangsu 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Shaowu Yin
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, Nanjing, Jiangsu 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China.
| | - Li Wang
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, Nanjing, Jiangsu 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, China
| | - Zhonglin Tang
- Nanjing Institute of Fisheries Science, Nanjing, Jiangsu 210036, China
| | - Shuqiao Chen
- Nanjing Institute of Fisheries Science, Nanjing, Jiangsu 210036, China
| |
Collapse
|
38
|
Pacini N, Borziani F. Oncostatic-Cytoprotective Effect of Melatonin and Other Bioactive Molecules: A Common Target in Mitochondrial Respiration. Int J Mol Sci 2016; 17:341. [PMID: 26959015 PMCID: PMC4813203 DOI: 10.3390/ijms17030341] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 11/27/2015] [Accepted: 12/03/2015] [Indexed: 12/27/2022] Open
Abstract
For several years, oncostatic and antiproliferative properties, as well as thoses of cell death induction through 5-methoxy-N-acetiltryptamine or melatonin treatment, have been known. Paradoxically, its remarkable scavenger, cytoprotective and anti-apoptotic characteristics in neurodegeneration models, such as Alzheimer’s disease and Parkinson’s disease are known too. Analogous results have been confirmed by a large literature to be associated to the use of many other bioactive molecules such as resveratrol, tocopherol derivatives or vitamin E and others. It is interesting to note that the two opposite situations, namely the neoplastic pathology and the neurodegeneration, are characterized by deep alterations of the metabolome, of mitochondrial function and of oxygen consumption, so that the oncostatic and cytoprotective action can find a potential rationalization because of the different metabolic and mitochondrial situations, and in the effect that these molecules exercise on the mitochondrial function. In this review we discuss historical and general aspects of melatonin, relations between cancers and the metabolome and between neurodegeneration and the metabolome, and the possible effects of melatonin and of other bioactive molecules on metabolic and mitochondrial dynamics. Finally, we suggest a common general mechanism as responsible for the oncostatic/cytoprotective effect of melatonin and of other molecules examined.
Collapse
Affiliation(s)
- Nicola Pacini
- Laboratorio Privato di Biochimica F. Pacini, via trabocchetto 10, 89126 Reggio Calabria, Italy.
| | - Fabio Borziani
- Laboratorio Privato di Biochimica F. Pacini, via trabocchetto 10, 89126 Reggio Calabria, Italy.
| |
Collapse
|
39
|
Lv J, Wang J, Chang S, Liu M, Pang X. The greedy nature of mutant RAS: a boon for drug discovery targeting cancer metabolism? Acta Biochim Biophys Sin (Shanghai) 2016; 48:17-26. [PMID: 26487443 DOI: 10.1093/abbs/gmv102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 07/20/2015] [Indexed: 12/13/2022] Open
Abstract
RAS oncogene mutations are frequently detected in human cancers. Among RAS-mediated tumorigenesis, KRAS-driven cancers are the most frequently diagnosed and resistant to current therapies. Despite more than three decades of intensive efforts, there are still no specific therapies for mutant RAS proteins. While trying to block those well-established downstream pathways, such as the RAF-MAPK pathway and the PI3K-AKT pathway, attentions have been paid to potential effects of RAS on metabolic pathways and the feasibility for targeting these pathways. Recent studies have proved that RAS not only promotes aerobic glycolysis and glutamine metabolism reprograming to provide energy, but it also facilitates branched metabolism pathways, autophagy, and macropinocytosis. These alterations generate building blocks for tumor growth and strengthen antioxidant defense in tumor cells. All of these metabolic changes meet different demands of RAS-driven cancers, making them distinct from normal cells. Indeed, some achievements have been made to inhibit tumor growth through targeting specific metabolism rewiring in preclinical models. Although there is still a long way to elucidate the landscape of altered metabolism, we believe that specific metabolic enzymes or pathways could be therapeutically targeted for selective inhibition of RAS-driven cancers.
Collapse
Affiliation(s)
- Jing Lv
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jieqiong Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Siyu Chang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China Department of Molecular and Cellular Medicine, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA
| | - Xiufeng Pang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
40
|
Mechanisms of Superoxide Generation and Signaling in Cytochrome bc Complexes. ADVANCES IN PHOTOSYNTHESIS AND RESPIRATION 2016. [DOI: 10.1007/978-94-017-7481-9_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
41
|
NADPH oxidases—do they play a role in TRPC regulation under hypoxia? Pflugers Arch 2015; 468:23-41. [DOI: 10.1007/s00424-015-1731-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/23/2015] [Accepted: 08/25/2015] [Indexed: 12/25/2022]
|
42
|
Hwang HJ, Lynn SG, Vengellur A, Saini Y, Grier EA, Ferguson-Miller SM, LaPres JJ. Hypoxia Inducible Factors Modulate Mitochondrial Oxygen Consumption and Transcriptional Regulation of Nuclear-Encoded Electron Transport Chain Genes. Biochemistry 2015; 54:3739-48. [PMID: 26030260 PMCID: PMC5957085 DOI: 10.1021/bi5012892] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Hypoxia inducible factor-1 (HIF1) is a stress-responsive nuclear transcription factor that is activated with a decrease in oxygen availability. HIF1 regulates the expression of genes involved in a cell's adaptation to hypoxic stress, including those with mitochondrial specific function. To gain a more comprehensive understanding of the role of HIF1 in mitochondrial homeostasis, we studied the link between hypoxia, HIF1 transactivation, and electron transport chain (ETC) function. We established immortalized mouse embryonic fibroblasts (MEFs) for HIF1α wild-type (WT) and null cells and tested whether HIF1α regulates mitochondrial respiration by modulating gene expressions of nuclear-encoded ETC components. High-throughput quantitative real-time polymerase chain reaction was performed to screen nuclear-encoded mitochondrial genes related to the ETC to identify those whose regulation was HIF1α-dependent. Our data suggest that HIF1α regulates transcription of cytochrome c oxidase (CcO) heart/muscle isoform 7a1 (Cox7a1) under hypoxia, where it is induced 1.5-2.5-fold, whereas Cox4i2 hypoxic induction was HIF1α-independent. We propose that adaptation to hypoxic stress of CcO as the main cellular oxygen consumer is mediated by induction of hypoxia-sensitive tissue-specific isoforms. We suggest that HIF1 plays a central role in maintaining homeostasis in cellular respiration during hypoxic stress via regulation of CcO activity.
Collapse
Affiliation(s)
- Hye Jin Hwang
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319, United States
- Center for Mitochondrial Science and Medicine, Michigan State University, East Lansing, Michigan 48824-1319, United States
| | - Scott G. Lynn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319, United States
- Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824-1319, United States
| | - Ajith Vengellur
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319, United States
| | - Yogesh Saini
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319, United States
- Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824-1319, United States
| | - Elizabeth A. Grier
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319, United States
| | - Shelagh M. Ferguson-Miller
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319, United States
- Center for Mitochondrial Science and Medicine, Michigan State University, East Lansing, Michigan 48824-1319, United States
| | - John J. LaPres
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319, United States
- Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824-1319, United States
- Center for Mitochondrial Science and Medicine, Michigan State University, East Lansing, Michigan 48824-1319, United States
| |
Collapse
|
43
|
Oxidative stress and aging: a non-invasive EPR investigation in human volunteers. Aging Clin Exp Res 2015; 27:235-8. [PMID: 25080128 DOI: 10.1007/s40520-014-0265-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 07/23/2014] [Indexed: 10/25/2022]
Abstract
The oxidative stress theory of aging has brought to the implicit expectation that oxidative stress increases with aging. Unfortunately, a broad investigation in humans is missing due to limitations of conventional oxidative stress status (OSS) analyses. Here we show that the OSS measured in peripheral blood of 247 healthy volunteers, aged 2 days-104 years, using the electron paramagnetic resonance "EPR-radical probe" technique, negatively correlated with age (-1.1 %/year; p < 0.0001) both by simple and multiple linear regression analyses and that it was only marginally affected by sex. These findings stimulate further mechanistic studies.
Collapse
|
44
|
Sunkari VG, Lind F, Botusan IR, Kashif A, Liu ZJ, Ylä-Herttuala S, Brismar K, Velazquez O, Catrina SB. Hyperbaric oxygen therapy activates hypoxia-inducible factor 1 (HIF-1), which contributes to improved wound healing in diabetic mice. Wound Repair Regen 2015; 23:98-103. [DOI: 10.1111/wrr.12253] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/05/2014] [Indexed: 12/30/2022]
Affiliation(s)
| | - Folke Lind
- Department of Anesthesiology Surgical Services and Intensive Care; Karolinska Hospital; Stockholm Sweden
| | | | - Abad Kashif
- Department of Surgery; Miller School of Medicine; University of Miami; Miami Florida
| | - Zhao-Jun Liu
- Department of Surgery; Miller School of Medicine; University of Miami; Miami Florida
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences; University of Eastern Finland; Kuopio Finland
| | - Kerstin Brismar
- Department of Molecular Medicine and Surgery; Karolinska Institute; Stockholm Sweden
| | - Omaida Velazquez
- Department of Surgery; Miller School of Medicine; University of Miami; Miami Florida
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery; Karolinska Institute; Stockholm Sweden
| |
Collapse
|
45
|
Bleier L, Wittig I, Heide H, Steger M, Brandt U, Dröse S. Generator-specific targets of mitochondrial reactive oxygen species. Free Radic Biol Med 2015; 78:1-10. [PMID: 25451644 DOI: 10.1016/j.freeradbiomed.2014.10.511] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 10/13/2014] [Accepted: 10/14/2014] [Indexed: 10/24/2022]
Abstract
To understand the role of reactive oxygen species (ROS) in oxidative stress and redox signaling it is necessary to link their site of generation to the oxidative modification of specific targets. Here we have studied the selective modification of protein thiols by mitochondrial ROS that have been implicated as deleterious agents in a number of degenerative diseases and in the process of biological aging, but also as important players in cellular signal transduction. We hypothesized that this bipartite role might be based on different generator sites for "signaling" and "damaging" ROS and a directed release into different mitochondrial compartments. Because two main mitochondrial ROS generators, complex I (NADH:ubiquinone oxidoreductase) and complex III (ubiquinol:cytochrome c oxidoreductase; cytochrome bc1 complex), are known to predominantly release superoxide and the derived hydrogen peroxide (H2O2) into the mitochondrial matrix and the intermembrane space, respectively, we investigated whether these ROS generators selectively oxidize specific protein thiols. We used redox fluorescence difference gel electrophoresis analysis to identify redox-sensitive targets in the mitochondrial proteome of intact rat heart mitochondria. We observed that the modified target proteins were distinctly different when complex I or complex III was employed as the source of ROS. These proteins are potential targets involved in mitochondrial redox signaling and may serve as biomarkers to study the generator-dependent dual role of mitochondrial ROS in redox signaling and oxidative stress.
Collapse
Affiliation(s)
- Lea Bleier
- Molecular Bioenergetics Group, Goethe-University, D-60590 Frankfurt am Main, Germany
| | - Ilka Wittig
- Molecular Bioenergetics Group, Goethe-University, D-60590 Frankfurt am Main, Germany; Functional Proteomics, SFB815 Core Unit, Medical School, Goethe-University, D-60590 Frankfurt am Main, Germany
| | - Heinrich Heide
- Molecular Bioenergetics Group, Goethe-University, D-60590 Frankfurt am Main, Germany
| | - Mirco Steger
- Molecular Bioenergetics Group, Goethe-University, D-60590 Frankfurt am Main, Germany
| | - Ulrich Brandt
- Molecular Bioenergetics Group, Goethe-University, D-60590 Frankfurt am Main, Germany; Cluster of Excellence Frankfurt "Macromolecular Complexes," Goethe-University, D-60590 Frankfurt am Main, Germany; Radboud University Medical Center, Nijmegen Center for Mitochondrial Disorders, 6500 GA Nijmegen, The Netherlands
| | - Stefan Dröse
- Molecular Bioenergetics Group, Goethe-University, D-60590 Frankfurt am Main, Germany; Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy, Goethe-University Hospital, Frankfurt am Main, Germany.
| |
Collapse
|
46
|
Bandarra D, Biddlestone J, Mudie S, Müller HAJ, Rocha S. HIF-1α restricts NF-κB-dependent gene expression to control innate immunity signals. Dis Model Mech 2014; 8:169-81. [PMID: 25510503 PMCID: PMC4314782 DOI: 10.1242/dmm.017285] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hypoxia and inflammation are intimately linked. It is known that nuclear factor κB (NF-κB) regulates the hypoxia-inducible factor (HIF) system, but little is known about how HIF regulates NF-κB. Here, we show that HIF-1α represses NF-κB-dependent gene expression. HIF-1α depletion results in increased NF-κB transcriptional activity both in mammalian cells and in the model organism Drosophila melanogaster. HIF-1α depletion enhances the NF-κB response, and this required not only the TAK-IKK complex, but also CDK6. Loss of HIF-1α results in an increased angiogenic response in mammalian cancer cells and increased mortality in Drosophila following infection. These results indicate that HIF-1α is required to restrain the NF-κB response, and thus prevents excessive and damaging pro-inflammatory responses.
Collapse
Affiliation(s)
- Daniel Bandarra
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - John Biddlestone
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - Sharon Mudie
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - H-Arno J Müller
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK
| | - Sonia Rocha
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH, UK.
| |
Collapse
|
47
|
Ito H, Matsui H, Tamura M, Majima HJ, Indo HP, Hyodo I. Mitochondrial reactive oxygen species accelerate the expression of heme carrier protein 1 and enhance photodynamic cancer therapy effect. J Clin Biochem Nutr 2014; 55:67-71. [PMID: 25120282 PMCID: PMC4078070 DOI: 10.3164/jcbn.14-27] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 02/27/2014] [Indexed: 12/26/2022] Open
Abstract
Photodynamic therapy using hematoporphyrin and its derivatives is clinically useful for cancer treatments. It has been reported that cancer cells incorporate hematoporphyrin and its derivatives via heme carrier protein 1, which is a proton-coupled folate transporter. However, the mechanism of this protein expression has not been elucidated. In general, the concentration of reactive oxygen species in cancer cells is higher than that in normal cells. We previously reported that reactive oxygen species from mitochondria involved in the expression of peptide transporter 1 and accelerate the uptake of 5-aminolevulinic acid, which is a precursor of protoporphyrin IX. We suggested mitochondrial reactive oxygen species also regulated the expression of heme carrier protein 1. In this study, we used a rat gastric mucosal cell line RGM1 and its cancer-like mutated cell line RGK1. We clarified the expression of heme carrier protein 1 increased in cancer cells and it decreased in manganese superoxide dismutase expressed cancer cells. In addition, the uptake level of hematoporphyrin and photodynamic therapeutic effect were also decreased in manganese superoxide dismutase expressed cancer cells in comparison with cancer cells. Thus, we concluded that mitochondrial reactive oxygen species regulated heme carrier protein 1 expression and photodynamic therapeutic effect.
Collapse
Affiliation(s)
- Hiromu Ito
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Ten-nohdai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hirofumi Matsui
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Ten-nohdai, Tsukuba, Ibaraki 305-8575, Japan
| | - Masato Tamura
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Ten-nohdai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hideyuki J Majima
- Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, Japan
| | - Hiroko P Indo
- Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, Japan
| | - Ichinosuke Hyodo
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Ten-nohdai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
48
|
Catrina SB. Impaired hypoxia-inducible factor (HIF) regulation by hyperglycemia. J Mol Med (Berl) 2014; 92:1025-34. [PMID: 25027070 DOI: 10.1007/s00109-014-1166-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 05/06/2014] [Accepted: 05/08/2014] [Indexed: 12/13/2022]
Abstract
The mechanisms that contribute to the development of diabetes complications remain unclear. A defective reaction of tissues to hypoxia has recently emerged as a new pathogenic mechanism and consists of a complex repression of hypoxia-inducible factor (HIF), which is the main regulator of the adaptive response to hypoxia. This paper discusses the mechanisms by which hyperglycaemia contributes to HIF repression in diabetes. Furthermore, a comprehensive analysis of the functional relevance of these new findings to the development of chronic diabetes complications is provided, along with examples from animal models and clinics.
Collapse
Affiliation(s)
- Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska University Hospital, L1:01, 17176, Stockholm, Sweden,
| |
Collapse
|
49
|
Bioenergetic analysis of ovarian cancer cell lines: profiling of histological subtypes and identification of a mitochondria-defective cell line. PLoS One 2014; 9:e98479. [PMID: 24858344 PMCID: PMC4032324 DOI: 10.1371/journal.pone.0098479] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 05/02/2014] [Indexed: 12/17/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal of all gynecological cancers, and encompasses distinct histological subtypes that have specific genetic and tissues-of-origin differences. Ovarian clear cell carcinoma (OCCC) represents approximately 10% of cases and has been termed a stress responsive cancer. OCCC is characterized by increased expression of oxidative stress and glycolysis-related genes. In the present study, we hypothesized that bioenergetic profiling might uniquely distinguish OCCC from other EOC histological subtypes. Using an extracellular flux analyzer, OCCC lines (ES-2, TOV-21-G) were shown to be highly metabolically active, with high oxygen consumption rate (OCR) and high extracellular acidification rate (ECAR), indicative of enhanced mitochondrial oxidative phosphorylation and glycolytic rate, respectively. A high bioenergetics profile was associated with the cell lines' ability to form anchorage independent spheroids. Given their high glycolytic and mitochondrial activity, OCCC cells displayed strong sensitivity to 2-deoxy-D-glucose and Rotenone growth inhibition, although this chemosensitivity profile was not specific to only OCCC cells. Bioenergetic profiling also identified a non-OCCC cell line, OVCA420, to have severely compromised mitochondrial function, based on low OCR and a lack of stimulation of maximal respiration following application of the uncoupler FCCP. This was accompanied by mitochondrial morphology changes indicative of enhanced fission, increased expression of the mitochondrial fission protein Drp1, a loss of mitochondrial membrane potential and dependence on glycolysis. Importantly, this loss of mitochondrial function was accompanied by the inability of OVCA420 cells to cope with hypoxic stress, and a compromised ability to stabilize HIF-1α in response to 1% O2 hypoxia. This knowledge may be imperative for researchers planning to utilize this cell line for further studies of metabolism and hypoxia, and suggests that altered mitochondrial fission dynamics represents a phenotype of a subpopulation of EOCs.
Collapse
|
50
|
Zhao L, Yang YF, Gao YB, Wang SM, Wang LF, Zuo HY, Dong J, Xu XP, Su ZT, Zhou HM, Zhu LL, Peng RY. Upregulation of HIF-1α via activation of ERK and PI3K pathway mediated protective response to microwave-induced mitochondrial injury in neuron-like cells. Mol Neurobiol 2014; 50:1024-34. [PMID: 24627260 DOI: 10.1007/s12035-014-8667-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 02/20/2014] [Indexed: 01/01/2023]
Abstract
Microwave-induced learning and memory deficits in animal models have been gaining attention in recent years, largely because of increasing public concerns on growing environmental influences. The data from our group and others have showed that the injury of mitochondria, the major source of cellular adenosine triphosphate (ATP) in primary neurons, could be detected in the neuron cells of microwave-exposed rats. In this study, we provided some insights into the cellular and molecular mechanisms behind mitochondrial injury in PC12 cell-derived neuron-like cells. PC12 cell-derived neuron-like cells were exposed to 30 mW/cm(2) microwave for 5 min, and damages of mitochondrial ultrastructure could be observed by using transmission electron microscopy. Impairments of mitochondrial function, indicated by decrease of ATP content, reduction of succinate dehydrogenase (SDH) and cytochrome c oxidase (COX) activities, decrease of mitochondrial membrane potential (MMP), and increase of reactive oxygen species (ROS) production, could be detected. We also found that hypoxia-inducible factor-1 (HIF-1α), a key regulator responsible for hypoxic response of the mammalian cells, was upregulated in microwave-exposed neuron-like cells. Furthermore, HIF-1α overexpression protected mitochondria from injury by increasing the ATP contents and MMP, while HIF-1α silence promoted microwave-induced mitochondrial damage. Finally, we demonstrated that both ERK and PI3K signaling activation are required in microwave-induced HIF-1α activation and protective response. In conclusion, we elucidated a regulatory connection between impairments of mitochondrial function and HIF-1α activation in microwave-exposed neuron-like cells. By modulating mitochondrial function and protecting neuron-like cells against microwave-induced mitochondrial injury, HIF-1α represents a promising therapeutic target for microwave radiation injury.
Collapse
Affiliation(s)
- Li Zhao
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Haidian District, 27 Taiping Road, Beijing, 100850, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|