1
|
Nabiullina R, Golovin S, Kirichenko E, Petrushan M, Logvinov A, Kaplya M, Sedova D, Rodkin S. 3D Bioprinting of Cultivated Meat Followed by the Development of a Fine-Tuned YOLO Model for the Detection and Counting of Lipoblasts, Fibroblasts, and Myogenic Cells. FRONT BIOSCI-LANDMRK 2025; 30:36266. [PMID: 40152392 DOI: 10.31083/fbl36266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/16/2025] [Accepted: 01/31/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Cultured meat holds significant potential as a pivotal solution for producing safe, sustainable, and high-quality protein to meet the growing demands of the global population. However, scaling this technology requires innovative bioengineering approaches integrated with software methods to assess the growth of cell cultures. This study aims to develop a technology for 3D printing a hybrid meat product and subsequently design a finely tuned You Only Look Once (YOLO) model for detecting and counting lipoblasts, fibroblasts, and myogenic cells. METHODS Cultures of multipotent mesenchymal stem cells (MMSCs) and fibroblasts were obtained from the domestic rabbit Oryctolagus cuniculus domesticus. Standard protocols were employed to induce adipogenic and myogenic differentiation from MMSCs. Fibroblasts were isolated from skin biopsy samples. The 3D printing process utilized bioinks. The engineering approach involved the development of a unique print head integrated into a 3D printer. Confocal and transmission electron microscopy of the cells within the construct was performed. A dataset of digital images of lipoblasts, myogenic cells, and fibroblasts was created. Four models based on the YOLOv8-seg architecture were trained on annotated images, implemented in the Telegram bot. RESULTS Stable cultures of lipoblasts, myogenic cells, and fibroblasts were obtained. 3D-printed tissue constructs composed of rabbit cells, sodium alginate, and sunflower protein were successfully fabricated. A unique print head for a 3D printer was assembled. Confocal microscopy confirmed cell viability within the tissue construct. Ultrastructural analysis revealed dense intercellular contacts and high metabolic activity. The resulting product replicated the organoleptic and structural properties of natural meat. In the IT segment, the single-class model trained on lipoblasts achieved metrics of recall 85%, precision 77%, and mean Average Precision at IoU threshold 0.50 (mAP50) 79%, which improved in the multiclass model to recall 92%, precision 92%, and mAP50 81%. The IT solution was implemented in a Telegram bot capable of detecting and counting different cell types. CONCLUSIONS A 3D tissue construct was achieved. Detailed microscopic analysis demonstrated cell viability and high metabolic activity within the polymerized alginate hydrogel. The engineered tissue product presents a potential alternative to natural meat. Additionally, the trained neural network models, implemented in a Telegram bot, proved effective in monitoring culture growth and identifying cell types in digital images across three cell cultures. As a result, we developed four YOLOv8 models and demonstrated that the multiclass model outperforms the single-class model. However, all models exhibited reduced accuracy in high-density cultures, where overlapping cells led to undercounting.
Collapse
Affiliation(s)
- Rozaliia Nabiullina
- Research Laboratory "Medical Digital Images Based on the Basic Model", Department of Bioengineering, Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, 344000 Rostov-on-Don, Russia
| | - Sergey Golovin
- Research Laboratory "Medical Digital Images Based on the Basic Model", Department of Bioengineering, Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, 344000 Rostov-on-Don, Russia
| | - Evgeniya Kirichenko
- Research Laboratory "Medical Digital Images Based on the Basic Model", Department of Bioengineering, Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, 344000 Rostov-on-Don, Russia
| | | | - Alexander Logvinov
- Academy of Biology and Biotechnology, Southern Federal University, 344090 Rostov-on-Don, Russia
| | - Marya Kaplya
- Research Laboratory "Medical Digital Images Based on the Basic Model", Department of Bioengineering, Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, 344000 Rostov-on-Don, Russia
| | - Darya Sedova
- Research Laboratory "Medical Digital Images Based on the Basic Model", Department of Bioengineering, Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, 344000 Rostov-on-Don, Russia
| | - Stanislav Rodkin
- Research Laboratory "Medical Digital Images Based on the Basic Model", Department of Bioengineering, Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, 344000 Rostov-on-Don, Russia
| |
Collapse
|
2
|
Cho H, Huh KM, Shim MS, Cho YY, Lee JY, Lee HS, Kwon YJ, Kang HC. Selective delivery of imaging probes and therapeutics to the endoplasmic reticulum or Golgi apparatus: Current strategies and beyond. Adv Drug Deliv Rev 2024; 212:115386. [PMID: 38971180 DOI: 10.1016/j.addr.2024.115386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/14/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
To maximize therapeutic effects and minimize unwanted effects, the interest in drug targeting to the endoplasmic reticulum (ER) or Golgi apparatus (GA) has been recently growing because two organelles are distributing hubs of cellular building/signaling components (e.g., proteins, lipids, Ca2+) to other organelles and the plasma membrane. Their structural or functional damages induce organelle stress (i.e., ER or GA stress), and their aggravation is strongly related to diseases (e.g., cancers, liver diseases, brain diseases). Many efforts have been developed to image (patho)physiological functions (e.g., oxidative stress, protein/lipid-related processing) and characteristics (e.g., pH, temperature, biothiols, reactive oxygen species) in the target organelles and to deliver drugs for organelle disruption using organelle-targeting moieties. Therefore, this review will overview the structure, (patho)physiological functions/characteristics, and related diseases of the organelles of interest. Future direction on ER or GA targeting will be discussed by understanding current strategies and investigations on targeting, imaging/sensing, and therapeutic systems.
Collapse
Affiliation(s)
- Hana Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Kang Moo Huh
- Departments of Polymer Science and Engineering & Materials Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Yong-Yeon Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Joo Young Lee
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hye Suk Lee
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Young Jik Kwon
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA
| | - Han Chang Kang
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea.
| |
Collapse
|
3
|
Dutta T, Chakraborty B, Nigam A, Minocha S, Koner AL. A small-molecule probe to decipher stress-induced ER microenvironments and ER-Golgi communication. J Mater Chem B 2024; 12:7848-7857. [PMID: 38808376 DOI: 10.1039/d4tb00572d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Cellular stress is a crucial factor in regulating and maintaining both organismal and microenvironmental homeostasis. It induces a response that also affects the micropolarity of specific cellular compartments, which is essential for early disease diagnosis. In this contribution, we present a quantitative study of micropolarity changes inside the endoplasmic reticulum (ER) during the G1/S and G2/M phases, using a biocompatible small-molecule fluorophore called ER-Oct. This probe is selectively driven to the ER by its hydrophobicity, and it has the fastest diffusion properties among a series of analogous probes. We found that induced ER stress caused cell cycle arrests leading to an increase in ER micropolarity which is well supported by lambda scanning experiments and fluorescence lifetime imaging microscopy (FLIM) as well. ER-Oct is a versatile staining agent that could effectively stain the ER in various living/fixed mammalian cells, isolated ER, Caenorhabditis elegans, and mice tissues. Furthermore, we used this probe to visualize a well-known biological event, ER to Golgi transport, by live-cell fluorescence microscopy. Our exhaustive investigation of micropolarity using ER-staining dye provides a new way to study ER stress, which could provide a deeper understanding of proteostasis in model systems and even in fixed patient samples.
Collapse
Affiliation(s)
- Tanoy Dutta
- Bionanotechnology Lab, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh - 462066, India.
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Barsha Chakraborty
- Bionanotechnology Lab, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh - 462066, India.
| | - Aditya Nigam
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi - 110016, India
| | - Shilpi Minocha
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi - 110016, India
| | - Apurba Lal Koner
- Bionanotechnology Lab, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh - 462066, India.
| |
Collapse
|
4
|
Zhou X, Xiao B, Jiang M, Rui J. Pan-cancer analysis identifies EMC6 as a potential target for lung adenocarcinoma. iScience 2024; 27:108648. [PMID: 38155776 PMCID: PMC10753071 DOI: 10.1016/j.isci.2023.108648] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/14/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023] Open
Abstract
Endoplasmic reticulum membrane protein complex subunit 6 (EMC6) plays an important function in both physiological and pathological states of cells. Nevertheless, there are few studies focused on the role of EMC6 in tumors. At first, we performed a series of bioinformatics analyses on 33 kinds of cancers, including differential expression analysis, tumor mutational burden analysis, prognostic analysis, and clinicopathological staging analysis. Then, we corroborated the important role of EMC6 in lung cancer by cytological and in vivo experiments. We found that the reduction of EMC6 expression did effectively inhibit the proliferation, invasion, and metastasis of A549. Finally, EMC6 is indeed involved in the regulation of ferroptosis, cuproptosis, and immune response in LUAD. In a word, our study not only comprehensively analyzed the functional mechanisms of EMC6 in all cancers but also validated the regulatory role of EMC6 in lung cancer for the first time.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Urology, The Wuxi No.2 People’s Hospital, Wuxi, China
| | - Bowen Xiao
- Department of Interventional Vascular Department, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Manman Jiang
- Suzhou Center for Disease Control, Suzhou, China
| | - Jun Rui
- Department of Chest, The Wuxi No.2 People’s Hospital, Wuxi, China
| |
Collapse
|
5
|
Baum B, Spang A. On the origin of the nucleus: a hypothesis. Microbiol Mol Biol Rev 2023; 87:e0018621. [PMID: 38018971 PMCID: PMC10732040 DOI: 10.1128/mmbr.00186-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023] Open
Abstract
SUMMARYIn this hypothesis article, we explore the origin of the eukaryotic nucleus. In doing so, we first look afresh at the nature of this defining feature of the eukaryotic cell and its core functions-emphasizing the utility of seeing the eukaryotic nucleoplasm and cytoplasm as distinct regions of a common compartment. We then discuss recent progress in understanding the evolution of the eukaryotic cell from archaeal and bacterial ancestors, focusing on phylogenetic and experimental data which have revealed that many eukaryotic machines with nuclear activities have archaeal counterparts. In addition, we review the literature describing the cell biology of representatives of the TACK and Asgardarchaeaota - the closest known living archaeal relatives of eukaryotes. Finally, bringing these strands together, we propose a model for the archaeal origin of the nucleus that explains much of the current data, including predictions that can be used to put the model to the test.
Collapse
Affiliation(s)
- Buzz Baum
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Anja Spang
- Department of Marine Microbiology and Biogeochemistry, NIOZ, Royal Netherlands Institute for Sea Research, Den Burg, the Netherlands
- Department of Evolutionary & Population Biology, Institute for Biodiversity and Ecosystem Dynamics (IBED), University of Amsterdam, Amsterdam, the Netherlands
- Department of Marine Microbiology and Biogeochemistry, NIOZ, Royal Netherlands Institute for Sea Research, Den Burg, the Netherlands
| |
Collapse
|
6
|
Patronas EM, Balber T, Miller A, Geist BK, Michligk A, Vraka C, Krisch M, Rohr-Udilova N, Haschemi A, Viernstein H, Hacker M, Mitterhauser M. A fingerprint of 2-[ 18F]FDG radiometabolites - How tissue-specific metabolism beyond 2-[ 18F]FDG-6-P could affect tracer accumulation. iScience 2023; 26:108137. [PMID: 37867937 PMCID: PMC10585399 DOI: 10.1016/j.isci.2023.108137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/01/2023] [Accepted: 10/02/2023] [Indexed: 10/24/2023] Open
Abstract
Studies indicate that the radiotracer 2-[18F]fluoro-2-deoxy-D-glucose (2-[18F]FDG) can be metabolized beyond 2-[18F]FDG-6-phosphate (2-[18F]FDG-6-P), but its metabolism is incompletely understood. Most importantly, it remains unclear whether downstream metabolism affects tracer accumulation in vivo. Here we present a fingerprint of 2-[18F]FDG radiometabolites over time in cancer cells, corresponding tumor xenografts and murine organs. Strikingly, radiometabolites representing glycogen metabolism or the oxPPP correlated inversely with tracer accumulation across all examined tissues. Recent studies suggest that not only hexokinase, but also hexose-6-phosphate dehydrogenase (H6PD), an enzyme of the oxidative pentose phosphate pathway (oxPPP), determines 2-[18F]FDG accumulation. However, little is known about the corresponding enzyme glucose-6-phosphate dehydrogenase (G6PD). Our mechanistic in vitro experiments on the role of the oxPPP propose that 2-[18F]FDG can be metabolized via both G6PD and H6PD, but data from separate enzyme knockdown suggest diverging roles in downstream tracer metabolism. Overall, we propose that tissue-specific metabolism beyond 2-[18F]FDG-6-P could matter for imaging.
Collapse
Affiliation(s)
- Eva-Maria Patronas
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
- Division of Pharmaceutical Technology and Biopharmaceutics, Department of Pharmaceutical Sciences, University of Vienna, Vienna 1090, Austria
| | - Theresa Balber
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna 1090, Austria
| | - Anne Miller
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna 1090, Austria
| | - Barbara Katharina Geist
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Antje Michligk
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Chrysoula Vraka
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Maximilian Krisch
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Nataliya Rohr-Udilova
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Helmut Viernstein
- Division of Pharmaceutical Technology and Biopharmaceutics, Department of Pharmaceutical Sciences, University of Vienna, Vienna 1090, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Markus Mitterhauser
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna 1090, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna 1090, Austria
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry, Vienna 1090, Austria
| |
Collapse
|
7
|
Zhao Y, Gao J, Fan Y, Xu H, Wang Y, Yao P. A risk score model based on endoplasmic reticulum stress related genes for predicting prognostic value of osteosarcoma. BMC Musculoskelet Disord 2023; 24:519. [PMID: 37353812 DOI: 10.1186/s12891-023-06629-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 06/12/2023] [Indexed: 06/25/2023] Open
Abstract
BACKGROUND We aimed to establish an osteosarcoma prognosis prediction model based on a signature of endoplasmic reticulum stress-related genes. METHODS Differentially expressed genes (DEGs) between osteosarcoma with and without metastasis from The Cancer Genome Atlas (TCGA) database were mapped to ERS genes retrieved from Gene Set Enrichment Analysis to select endoplasmic reticulum stress-related DEGs. Subsequently, we constructed a risk score model based on survival-related endoplasmic reticulum stress DEGs and a nomogram of independent survival prognostic factors. Based on the median risk score, we stratified the samples into high- and low-risk groups. The ability of the model was assessed by Kaplan-Meier, receiver operating characteristic curve, and functional analyses. Additionally, the expression of the identified prognostic endoplasmic reticulum stress-related DEGs was verified using real-time quantitative PCR (RT-qPCR). RESULTS In total, 41 endoplasmic reticulum stress-related DEGs were identified in patients with osteosarcoma with metastasis. A risk score model consisting of six prognostic endoplasmic reticulum stress-related DEGs (ATP2A3, ERMP1, FBXO6, ITPR1, NFE2L2, and USP13) was established, and the Kaplan-Meier and receiver operating characteristic curves validated their performance in the training and validation datasets. Age, tumor metastasis, and the risk score model were demonstrated to be independent prognostic clinical factors for osteosarcoma and were used to establish a nomogram survival model. The nomogram model showed similar performance of one, three, and five year-survival rate to the actual survival rates. Nine immune cell types in the high-risk group were found to be significantly different from those in the low-risk group. These survival-related genes were significantly enriched in nine Kyoto Encyclopedia of Genes and Genomes pathways, including cell adhesion molecule cascades, and chemokine signaling pathways. Further, RT-qPCR results demonstrated that the consistency rate of bioinformatics analysis was approximately 83.33%, suggesting the relatively high reliability of the bioinformatics analysis. CONCLUSION We established an osteosarcoma prediction model based on six prognostic endoplasmic reticulum stress-related DEGs that could be helpful in directing personalized treatment.
Collapse
Affiliation(s)
- Yong Zhao
- Department of Orthopaedic Surgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shaoxing, 312400, Zhejiang, China.
| | - Jijian Gao
- Department of Orthopaedic Surgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shaoxing, 312400, Zhejiang, China
| | - Yong Fan
- Department of Orthopaedic Surgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shaoxing, 312400, Zhejiang, China
| | - Hongyu Xu
- Department of Orthopaedic Surgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shaoxing, 312400, Zhejiang, China
| | - Yun Wang
- Department of Orthopaedic Surgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shaoxing, 312400, Zhejiang, China
| | - Pengjie Yao
- Department of Orthopaedic Surgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shaoxing, 312400, Zhejiang, China
| |
Collapse
|
8
|
Arruda AP, Parlakgül G. Endoplasmic Reticulum Architecture and Inter-Organelle Communication in Metabolic Health and Disease. Cold Spring Harb Perspect Biol 2023; 15:a041261. [PMID: 35940911 PMCID: PMC9899651 DOI: 10.1101/cshperspect.a041261] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The endoplasmic reticulum (ER) is a key organelle involved in the regulation of lipid and glucose metabolism, proteostasis, Ca2+ signaling, and detoxification. The structural organization of the ER is very dynamic and complex, with distinct subdomains such as the nuclear envelope and the peripheral ER organized into ER sheets and tubules. ER also forms physical contact sites with all other cellular organelles and with the plasma membrane. Both form and function of the ER are highly adaptive, with a potent capacity to respond to transient changes in environmental cues such as nutritional fluctuations. However, under obesity-induced chronic stress, the ER fails to adapt, leading to ER dysfunction and the development of metabolic pathologies such as insulin resistance and fatty liver disease. Here, we discuss how the remodeling of ER structure and contact sites with other organelles results in diversification of metabolic function and how perturbations to this structural flexibility by chronic overnutrition contribute to ER dysfunction and metabolic pathologies in obesity.
Collapse
Affiliation(s)
- Ana Paula Arruda
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California 94720, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| | - Güneş Parlakgül
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California 94720, USA
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| |
Collapse
|
9
|
Mitochondrial Generated Redox Stress Differently Affects the Endoplasmic Reticulum of Circulating Lymphocytes and Monocytes in Treatment-Naïve Hodgkin’s Lymphoma. Antioxidants (Basel) 2022; 11:antiox11040762. [PMID: 35453447 PMCID: PMC9024578 DOI: 10.3390/antiox11040762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 02/06/2023] Open
Abstract
Background. The redox stress caused by Hodgkin’s lymphoma (HL) also involves the peripheral blood mononucleated cells (PBMCs) even before chemotherapy. Here, we tested whether lymphocytes and monocytes show a different response to the increased mitochondrial generation of reactive oxygen species (ROS). Methods. PBMCs, isolated from the blood of treatment-naïve HL patients and control subjects, underwent assessment of malondialdehyde content and enzymatic activity of both hexose- and glucose-6P dehydrogenase (H6PD and G6PD) as well as flow cytometric analysis of mitochondrial ROS content. These data were complemented by evaluating the uptake of the fluorescent glucose analogue 2-NBDG that is selectively stored within the endoplasmic reticulum (ER). Results. Malondialdehyde content was increased in the whole population of HL PBMCs. The oxidative damage matched an increased activity of G6PD, and even more of H6PD, that trigger the cytosolic and ER pentose phosphate pathways, respectively. At flow cytometry, the number of recovered viable cells was selectively decreased in HL lymphocytes that also showed a more pronounced increase in mitochondrial ROS generation and 2-NBDG uptake, with respect to monocytes. Conclusions. PBMCs of HL patients display a selective mitochondrial and ER redox stress most evident in lymphocytes already before the exposure to chemotherapy toxicity.
Collapse
|
10
|
Caffeine and MDMA (Ecstasy) Exacerbate ER Stress Triggered by Hyperthermia. Int J Mol Sci 2022; 23:ijms23041974. [PMID: 35216090 PMCID: PMC8880705 DOI: 10.3390/ijms23041974] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Drugs of abuse can cause local and systemic hyperthermia, a known trigger of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). Another trigger of ER stress and UPR is ER calcium depletion, which causes ER exodosis, the secretion of ER-resident proteins. In rodent models, club drugs such as 3,4-methylenedioxymethamphetamine (MDMA, ‘ecstasy’) can create hyperthermic conditions in the brain and cause toxicity that is affected by the environmental temperature and the presence of other drugs, such as caffeine. In human studies, MDMA stimulated an acute, dose-dependent increase in core body temperature, but an examination of caffeine and MDMA in combination remains a topic for clinical research. Here we examine the secretion of ER-resident proteins and activation of the UPR under combined exposure to MDMA and caffeine in a cellular model of hyperthermia. We show that hyperthermia triggers the secretion of normally ER-resident proteins, and that this aberrant protein secretion is potentiated by the presence of MDMA, caffeine, or a combination of the two drugs. Hyperthermia activates the UPR but the addition of MDMA or caffeine does not alter the canonical UPR gene expression despite the drug effects on ER exodosis of UPR-related proteins. One exception was increased BiP/GRP78 mRNA levels in MDMA-treated cells exposed to hyperthermia. These findings suggest that club drug use under hyperthermic conditions exacerbates disruption of ER proteostasis, contributing to cellular toxicity.
Collapse
|
11
|
Marini C, Cossu V, Kumar M, Milanese M, Cortese K, Bruno S, Bellese G, Carta S, Zerbo RA, Torazza C, Bauckneht M, Venturi C, Raffa S, Orengo AM, Donegani MI, Chiola S, Ravera S, Castellani P, Morbelli S, Sambuceti G, Bonanno G. The Role of Endoplasmic Reticulum in the Differential Endurance against Redox Stress in Cortical and Spinal Astrocytes from the Newborn SOD1 G93A Mouse Model of Amyotrophic Lateral Sclerosis. Antioxidants (Basel) 2021; 10:antiox10091392. [PMID: 34573024 PMCID: PMC8472526 DOI: 10.3390/antiox10091392] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/19/2021] [Accepted: 08/27/2021] [Indexed: 12/01/2022] Open
Abstract
Recent studies reported that the uptake of [18F]-fluorodeoxyglucose (FDG) is increased in the spinal cord (SC) and decreased in the motor cortex (MC) of patients with ALS, suggesting that the disease might differently affect the two nervous districts with different time sequence or with different mechanisms. Here we show that MC and SC astrocytes harvested from newborn B6SJL-Tg (SOD1G93A) 1Gur mice could play different roles in the pathogenesis of the disease. Spectrophotometric and cytofluorimetric analyses showed an increase in redox stress, a decrease in antioxidant capacity and a relative mitochondria respiratory uncoupling in MC SOD1G93A astrocytes. By contrast, SC mutated cells showed a higher endurance against oxidative damage, through the increase in antioxidant defense, and a preserved respiratory function. FDG uptake reproduced the metabolic response observed in ALS patients: SOD1G93A mutation caused a selective enhancement in tracer retention only in mutated SC astrocytes, matching the activity of the reticular pentose phosphate pathway and, thus, of hexose-6P dehydrogenase. Finally, both MC and SC mutated astrocytes were characterized by an impressive ultrastructural enlargement of the endoplasmic reticulum (ER) and impairment in ER–mitochondria networking, more evident in mutated MC than in SC cells. Thus, SOD1G93A mutation differently impaired MC and SC astrocyte biology in a very early stage of life.
Collapse
Affiliation(s)
- Cecilia Marini
- CNR Institute of Molecular Bioimaging and Physiology (IBFM), Segrate, 20054 Milan, Italy
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (A.M.O.); (S.C.); (S.M.); (G.S.)
- Correspondence:
| | - Vanessa Cossu
- Department of Health Sciences, University of Genoa, 16132 Genoa, Italy; (V.C.); (S.R.); (M.I.D.)
| | - Mandeep Kumar
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genoa, 16148 Genoa, Italy; (M.K.); (M.M.); (R.A.Z.); (C.T.); (G.B.)
| | - Marco Milanese
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genoa, 16148 Genoa, Italy; (M.K.); (M.M.); (R.A.Z.); (C.T.); (G.B.)
| | - Katia Cortese
- Department of Experimental Medicine, Human Anatomy, University of Genoa, 16132 Genoa, Italy; (K.C.); (S.B.); (G.B.); (C.V.); (S.R.)
| | - Silvia Bruno
- Department of Experimental Medicine, Human Anatomy, University of Genoa, 16132 Genoa, Italy; (K.C.); (S.B.); (G.B.); (C.V.); (S.R.)
| | - Grazia Bellese
- Department of Experimental Medicine, Human Anatomy, University of Genoa, 16132 Genoa, Italy; (K.C.); (S.B.); (G.B.); (C.V.); (S.R.)
| | - Sonia Carta
- Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (S.C.); (P.C.)
| | - Roberta Arianna Zerbo
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genoa, 16148 Genoa, Italy; (M.K.); (M.M.); (R.A.Z.); (C.T.); (G.B.)
| | - Carola Torazza
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genoa, 16148 Genoa, Italy; (M.K.); (M.M.); (R.A.Z.); (C.T.); (G.B.)
| | - Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (A.M.O.); (S.C.); (S.M.); (G.S.)
| | - Consuelo Venturi
- Department of Experimental Medicine, Human Anatomy, University of Genoa, 16132 Genoa, Italy; (K.C.); (S.B.); (G.B.); (C.V.); (S.R.)
| | - Stefano Raffa
- Department of Health Sciences, University of Genoa, 16132 Genoa, Italy; (V.C.); (S.R.); (M.I.D.)
| | - Anna Maria Orengo
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (A.M.O.); (S.C.); (S.M.); (G.S.)
| | - Maria Isabella Donegani
- Department of Health Sciences, University of Genoa, 16132 Genoa, Italy; (V.C.); (S.R.); (M.I.D.)
| | - Silvia Chiola
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (A.M.O.); (S.C.); (S.M.); (G.S.)
| | - Silvia Ravera
- Department of Experimental Medicine, Human Anatomy, University of Genoa, 16132 Genoa, Italy; (K.C.); (S.B.); (G.B.); (C.V.); (S.R.)
| | - Patrizia Castellani
- Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (S.C.); (P.C.)
| | - Silvia Morbelli
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (A.M.O.); (S.C.); (S.M.); (G.S.)
- Department of Health Sciences, University of Genoa, 16132 Genoa, Italy; (V.C.); (S.R.); (M.I.D.)
| | - Gianmario Sambuceti
- CNR Institute of Molecular Bioimaging and Physiology (IBFM), Segrate, 20054 Milan, Italy
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (A.M.O.); (S.C.); (S.M.); (G.S.)
| | - Giambattista Bonanno
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genoa, 16148 Genoa, Italy; (M.K.); (M.M.); (R.A.Z.); (C.T.); (G.B.)
- Pharmacology and Toxycology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
12
|
Marini C, Cossu V, Bauckneht M, Lanfranchi F, Raffa S, Orengo AM, Ravera S, Bruno S, Sambuceti G. Metformin and Cancer Glucose Metabolism: At the Bench or at the Bedside? Biomolecules 2021; 11:biom11081231. [PMID: 34439897 PMCID: PMC8392176 DOI: 10.3390/biom11081231] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
Several studies reported that metformin, the most widely used drug for type 2 diabetes, might affect cancer aggressiveness. The biguanide seems to directly impair cancer energy asset, with the consequent phosphorylation of AMP-activated protein kinase (AMPK) inhibiting cell proliferation and tumor growth. This action is most often attributed to a well-documented blockage of oxidative phosphorylation (OXPHOS) caused by a direct interference of metformin on Complex I function. Nevertheless, several other pleiotropic actions seem to contribute to the anticancer potential of this biguanide. In particular, in vitro and in vivo experimental studies recently documented that metformin selectively inhibits the uptake of 2-[18F]-Fluoro-2-Deoxy-D-Glucose (FDG), via an impaired catalytic function of the enzyme hexose-6P-dehydrogenase (H6PD). H6PD triggers a still largely uncharacterized pentose-phosphate pathway (PPP) within the endoplasmic reticulum (ER) that has been found to play a pivotal role in feeding the NADPH reductive power for both cellular proliferation and antioxidant responses. Regardless of its exploitability in the clinical setting, this metformin action might configure the ER metabolism as a potential target for innovative therapeutic strategies in patients with solid cancers and potentially modifies the current interpretative model of FDG uptake, attributing PET/CT capability to predict cancer aggressiveness to the activation of H6PD catalytic function.
Collapse
Affiliation(s)
- Cecilia Marini
- CNR Institute of Molecular Bioimaging and Physiology (IBFM), 20054 Milan, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (A.M.O.)
- Correspondence: ; Tel.: +39-010-555-4812
| | - Vanessa Cossu
- Department of Health Sciences, University of Genoa, 16132 Genoa, Italy; (V.C.); (F.L.); (S.R.)
| | - Matteo Bauckneht
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (A.M.O.)
- Department of Health Sciences, University of Genoa, 16132 Genoa, Italy; (V.C.); (F.L.); (S.R.)
| | - Francesco Lanfranchi
- Department of Health Sciences, University of Genoa, 16132 Genoa, Italy; (V.C.); (F.L.); (S.R.)
| | - Stefano Raffa
- Department of Health Sciences, University of Genoa, 16132 Genoa, Italy; (V.C.); (F.L.); (S.R.)
| | - Anna Maria Orengo
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (A.M.O.)
| | - Silvia Ravera
- Department of Experimental Medicine, Human Anatomy, University of Genoa, 16132 Genoa, Italy; (S.R.); (S.B.)
| | - Silvia Bruno
- Department of Experimental Medicine, Human Anatomy, University of Genoa, 16132 Genoa, Italy; (S.R.); (S.B.)
| | - Gianmario Sambuceti
- CNR Institute of Molecular Bioimaging and Physiology (IBFM), 20054 Milan, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (A.M.O.)
| |
Collapse
|
13
|
Almeida C. A potential third-order role of the host endoplasmic reticulum as a contact site in interkingdom microbial endosymbiosis and viral infection. ENVIRONMENTAL MICROBIOLOGY REPORTS 2021; 13:255-271. [PMID: 33559322 DOI: 10.1111/1758-2229.12938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 06/12/2023]
Abstract
The normal functioning of eukaryotic cells depends on the compartmentalization of metabolic processes within specific organelles. Interactions among organelles, such as those between the endoplasmic reticulum (ER) - considered the largest single structure in eukaryotic cells - and other organelles at membrane contact sites (MCSs) have also been suggested to trigger synergisms, including intracellular immune responses against pathogens. In addition to the ER-endogenous functions and ER-organelle MCSs, we present the perspective of a third-order role of the ER as a host contact site for endosymbiotic microbial non-pathogens and pathogens, from endosymbiont bacteria to parasitic protists and viruses. Although understudied, ER-endosymbiont interactions have been observed in a range of eukaryotic hosts, including protists, plants, algae, and metazoans. Host ER interactions with endosymbionts could be an ER function built from ancient, conserved mechanisms selected for communicating with mutualistic endosymbionts in specific life cycle stages, and they may be exploited by pathogens and parasites. The host ER-'guest' interactome and traits in endosymbiotic biology are briefly discussed. The acknowledgment and understanding of these possible mechanisms might reveal novel evolutionary perspectives, uncover the causes of unexplained cellular disorders and suggest new pharmacological targets.
Collapse
Affiliation(s)
- Celso Almeida
- ENDOBIOS Biotech®, Praceta Progresso Clube n° 6, 2725-110 Mem-Martins, Portugal
| |
Collapse
|
14
|
Sambuceti G, Cossu V, Bauckneht M, Morbelli S, Orengo A, Carta S, Ravera S, Bruno S, Marini C. 18F-fluoro-2-deoxy-d-glucose (FDG) uptake. What are we looking at? Eur J Nucl Med Mol Imaging 2021; 48:1278-1286. [PMID: 33864142 DOI: 10.1007/s00259-021-05368-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Gianmario Sambuceti
- IRCCS Ospedale Policlinico San Martino, Nuclear Medicine, Largo Rosanna Benzi 10, 16132, Genoa, Italy. .,CNR Institute of Molecular Bioimaging and Physiology (IBFM), Milan, Italy.
| | - Vanessa Cossu
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Matteo Bauckneht
- IRCCS Ospedale Policlinico San Martino, Nuclear Medicine, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Silvia Morbelli
- IRCCS Ospedale Policlinico San Martino, Nuclear Medicine, Largo Rosanna Benzi 10, 16132, Genoa, Italy.,Department of Health Sciences, University of Genoa, Genoa, Italy
| | - AnnaMaria Orengo
- IRCCS Ospedale Policlinico San Martino, Nuclear Medicine, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Sonia Carta
- IRCCS Ospedale Policlinico San Martino, Nuclear Medicine, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Silvia Ravera
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genoa, Italy
| | - Silvia Bruno
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genoa, Italy
| | - Cecilia Marini
- IRCCS Ospedale Policlinico San Martino, Nuclear Medicine, Largo Rosanna Benzi 10, 16132, Genoa, Italy.,CNR Institute of Molecular Bioimaging and Physiology (IBFM), Milan, Italy
| |
Collapse
|
15
|
Two high-rate pentose-phosphate pathways in cancer cells. Sci Rep 2020; 10:22111. [PMID: 33335166 PMCID: PMC7746718 DOI: 10.1038/s41598-020-79185-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 11/30/2020] [Indexed: 11/24/2022] Open
Abstract
The relevant role of pentose phosphate pathway (PPP) in cancer metabolic reprogramming has been usually outlined by studying glucose-6-phosphate dehydrogenase (G6PD). However, recent evidence suggests an unexpected role for a less characterized PPP, triggered by hexose-6-phosphate dehydrogenase (H6PD) within the endoplasmic reticulum (ER). Studying H6PD biological role in breast and lung cancer, here we show that gene silencing of this reticular enzyme decreases cell content of PPP intermediates and d-ribose, to a similar extent as G6PD silencing. Decrease in overall NADPH content and increase in cell oxidative status are also comparable. Finally, either gene silencing impairs at a similar degree cell proliferating activity. This unexpected response occurs despite the absence of any cross-interference between the expression of both G6PD and H6PD. Thus, overall cancer PPP reflects the contribution of two different pathways located in the cytosol and ER, respectively. Disregarding the reticular pathway might hamper our comprehension of PPP role in cancer cell biology.
Collapse
|
16
|
Wang DD, Zou LW, Jin Q, Guan XQ, Yu Y, Zhu YD, Huang J, Gao P, Wang P, Ge GB, Yang L. Bioluminescent Sensor Reveals that Carboxylesterase 1A is a Novel Endoplasmic Reticulum-Derived Serologic Indicator for Hepatocyte Injury. ACS Sens 2020; 5:1987-1995. [PMID: 32529833 DOI: 10.1021/acssensors.0c00384] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Discovery of novel liver injury indicators and development of practical assays to detect target indicator(s) would strongly facilitate the diagnosis of liver disorders. Herein, an alternative biomarker discovery strategy was applied to find suitable endoplasmic reticulum-resident protein(s) as serologic indicator(s) for hepatocyte injury via analysis of the human proteome database among plasma and various organs. Both database searching and preliminary experiments suggested that human carboxylesterase 1A (CES1A), one of the most abundant and hepatic-restricted proteins, could serve as a good serologic indicator for hepatocyte injury. Then, a highly selective and practical bioluminescent sensor was developed for real-time sensing of CES1A in various biological systems including plasma. With the help of this bioluminescent sensor, the release of hepatic CES1A into the extracellular medium or the circulation system could be directly monitored. Further investigations demonstrated that serum activity levels of CES1A were elevated dramatically in mice with liver injury or patients with liver diseases. Collectively, this study provided solid evidence to support that CES1A was a novel serological indicator for hepatocyte injury. Furthermore, the strategy used in this study paved a new way for the rational discovery of practical indicators to monitor the dynamic progression of injury in a given tissue or organ.
Collapse
Affiliation(s)
- Dan-Dan Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li-Wei Zou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qiang Jin
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao-Qing Guan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yang Yu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ya-Di Zhu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jian Huang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Institute of Food and Drug Control, Shanghai 201203, China
| | - Peng Gao
- Dalian Sixth Peoples Hospital Affiliated of Dalian Medical University, Dalian 116001, China
| | - Ping Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ling Yang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
17
|
Marini C, Cossu V, Bonifacino T, Bauckneht M, Torazza C, Bruno S, Castellani P, Ravera S, Milanese M, Venturi C, Carlone S, Piccioli P, Emionite L, Morbelli S, Orengo AM, Donegani MI, Miceli A, Raffa S, Marra S, Signori A, Cortese K, Grillo F, Fiocca R, Bonanno G, Sambuceti G. Mechanisms underlying the predictive power of high skeletal muscle uptake of FDG in amyotrophic lateral sclerosis. EJNMMI Res 2020; 10:76. [PMID: 32638178 PMCID: PMC7340686 DOI: 10.1186/s13550-020-00666-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023] Open
Abstract
Background We recently reported that enhanced [18F]-fluorodeoxyglucose (FDG) uptake in skeletal muscles predicts disease aggressiveness in patients with amyotrophic lateral sclerosis (ALS). The present experimental study aimed to assess whether this predictive potential reflects the link between FDG uptake and redox stress that has been previously reported in different tissues and disease models. Methods The study included 15 SOD1G93A mice (as experimental ALS model) and 15 wildtype mice (around 120 days old). Mice were submitted to micro-PET imaging. Enzymatic pathways and response to oxidative stress were evaluated in harvested quadriceps and hearts by biochemical, immunohistochemical, and immunofluorescence analysis. Colocalization between the endoplasmic reticulum (ER) and the fluorescent FDG analog 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxyglucose (2-NBDG) was performed in fresh skeletal muscle sections. Finally, mitochondrial ultrastructure and bioenergetics were evaluated in harvested quadriceps and hearts. Results FDG retention was significantly higher in hindlimb skeletal muscles of symptomatic SOD1G93A mice with respect to control ones. This difference was not explained by any acceleration in glucose degradation through glycolysis or cytosolic pentose phosphate pathway (PPP). Similarly, it was independent of inflammatory infiltration. Rather, the high FDG retention in SOD1G93A skeletal muscle was associated with an accelerated generation of reactive oxygen species. This redox stress selectively involved the ER and the local PPP triggered by hexose-6P-dehydrogenase. ER involvement was confirmed by the colocalization of the 2-NBDG with a vital ER tracker. The oxidative damage in transgenic skeletal muscle was associated with a severe impairment in the crosstalk between ER and mitochondria combined with alterations in mitochondrial ultrastructure and fusion/fission balance. The expected respiratory damage was confirmed by a deceleration in ATP synthesis and oxygen consumption rate. These same abnormalities were represented to a markedly lower degree in the myocardium, as a sample of non-voluntary striated muscle. Conclusion Skeletal muscle of SOD1G93A mice reproduces the increased FDG uptake observed in ALS patients. This finding reflects the selective activation of the ER-PPP in response to significant redox stress associated with alterations of mitochondrial ultrastructure, networking, and connection with the ER itself. This scenario is less severe in cardiomyocytes suggesting a relevant role for either communication with synaptic plaque or contraction dynamics.
Collapse
Affiliation(s)
- Cecilia Marini
- CNR Institute of Molecular Bioimaging and Physiology (IBFM), Milano, Italy. .,Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy.
| | - Vanessa Cossu
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy.,Department of Health Sciences, University of Genoa, Genova, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Section of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy.,Department of Health Sciences, University of Genoa, Genova, Italy
| | - Carola Torazza
- Department of Pharmacy, Section of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Silvia Bruno
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genova, Italy
| | | | - Silvia Ravera
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genova, Italy
| | - Marco Milanese
- Department of Pharmacy, Section of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Consuelo Venturi
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genova, Italy
| | | | | | - Laura Emionite
- Animal Facility, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Silvia Morbelli
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy.,Department of Health Sciences, University of Genoa, Genova, Italy
| | - Anna Maria Orengo
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy
| | | | - Alberto Miceli
- Department of Health Sciences, University of Genoa, Genova, Italy
| | - Stefano Raffa
- Department of Health Sciences, University of Genoa, Genova, Italy
| | - Stefano Marra
- Department of Health Sciences, University of Genoa, Genova, Italy
| | - Alessio Signori
- Department of Health Sciences, University of Genoa, Genova, Italy
| | - Katia Cortese
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genova, Italy
| | - Federica Grillo
- Department of Surgical Sciences and Integrated Diagnostics, Pathology Unit, University of Genoa, Genova, Italy
| | - Roberto Fiocca
- Department of Surgical Sciences and Integrated Diagnostics, Pathology Unit, University of Genoa, Genova, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Section of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, Genova, Italy.,Pharmacology and Toxicology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy.,Department of Health Sciences, University of Genoa, Genova, Italy
| |
Collapse
|
18
|
Cossu V, Bauckneht M, Bruno S, Orengo AM, Emionite L, Balza E, Castellani P, Piccioli P, Miceli A, Raffa S, Borra A, Donegani MI, Carlone S, Morbelli S, Ravera S, Sambuceti G, Marini C. The Elusive Link Between Cancer FDG Uptake and Glycolytic Flux Explains the Preserved Diagnostic Accuracy of PET/CT in Diabetes. Transl Oncol 2020; 13:100752. [PMID: 32302773 PMCID: PMC7163080 DOI: 10.1016/j.tranon.2020.100752] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/05/2020] [Accepted: 02/26/2020] [Indexed: 01/21/2023] Open
Abstract
This study aims to verify in experimental models of hyperglycemia induced by streptozotocin (STZ-DM) to what degree the high competition between unlabeled glucose and metformin (MET) treatment might affect the accuracy of cancer FDG imaging. The study included 36 “control” and 36 “STZ-DM” Balb/c mice, undergoing intraperitoneal injection of saline or streptozotocin, respectively. Two-weeks later, mice were subcutaneously implanted with breast (4 T1) or colon (CT26) cancer cells and subdivided in three subgroups for treatment with water or with MET at 10 or 750 mg/Kg/day. Two weeks after, mice were submitted to micro-PET imaging. Enzymatic pathways and response to oxidative stress were evaluated in harvested tumors. Finally, competition by glucose, 2-deoxyglucose (2DG) and the fluorescent analog 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxyglucose (2-NBDG) on FDG uptake was studied in 4 T1 and CT26 cultured cells. STZ-DM slightly decreased cancer volume and FDG uptake rate (MRF). More importantly, it also abolished MET capability to decelerate lesion growth and MRF. This metabolic reprogramming closely agreed with the activity of hexose-6-phosphate dehydrogenase within the endoplasmic reticulum. Finally, co-incubation with 2DG virtually abolished FDG and 2-NBDG uptake within the endoplasmic reticulum in cultured cells. These data challenge the current dogma linking FDG uptake to glycolytic flux and introduce a new model to explain the relation between glucose analogue uptake and hexoses reticular metabolism. This selective fate of FDG contributes to the preserved sensitivity of PET imaging in oncology even in chronic moderate hyperglycemic conditions.
Collapse
Affiliation(s)
- Vanessa Cossu
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Health Sciences, University of Genoa, Italy
| | - Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Health Sciences, University of Genoa, Italy
| | - Silvia Bruno
- Department Experimental Medicine, University of Genoa, Italy
| | - Anna Maria Orengo
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Laura Emionite
- Animal Facility, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Enrica Balza
- Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Patrizia Piccioli
- Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alberto Miceli
- Department of Health Sciences, University of Genoa, Italy
| | - Stefano Raffa
- Department of Health Sciences, University of Genoa, Italy
| | - Anna Borra
- Department of Health Sciences, University of Genoa, Italy
| | | | | | - Silvia Morbelli
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Silvia Ravera
- Department Experimental Medicine, University of Genoa, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Health Sciences, University of Genoa, Italy
| | - Cecilia Marini
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genova, Italy; CNR Institute of Molecular Bioimaging and Physiology (IBFM), Milan, Italy.
| |
Collapse
|
19
|
Bauckneht M, Cossu V, Castellani P, Piccioli P, Orengo AM, Emionite L, Di Giulio F, Donegani MI, Miceli A, Raffa S, Borra A, Capitanio S, Morbelli S, Caviglia G, Bruno S, Ravera S, Maggi D, Sambuceti G, Marini C. FDG uptake tracks the oxidative damage in diabetic skeletal muscle: An experimental study. Mol Metab 2019; 31:98-108. [PMID: 31918925 PMCID: PMC6920267 DOI: 10.1016/j.molmet.2019.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/29/2019] [Accepted: 11/03/2019] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES The present study aims to verify the relationship between glucose consumption and uptake of 18F-2-deoxy-glucose (FDG) in the skeletal muscle (SM) of experimental models of streptozotocin-induced diabetes mellitus (STZ-DM). METHODS The study included 36 Balb/c mice. Two weeks after intraperitoneal administration of saline (control group, n = 18) or 150 mg streptozotocin (STZ-DM group, n = 18), the two cohorts were submitted to an oral glucose tolerance test and were further subdivided into three groups (n = 6 each): untreated and treated with metformin (MTF) at low or high doses (10 or 750 mg/kg daily, respectively). Two weeks thereafter, all mice were submitted to dynamic micro-positron emission tomography (PET) imaging after prolonged fasting. After sacrifice, enzymatic pathways and response to oxidative stress were evaluated in harvested SM. RESULTS On PET imaging, the FDG uptake rate in hindlimb SM was significantly lower in nondiabetic mice as compared with STZ-DM-untreated mice. MTF had no significant effect on SM FDG uptake in untreated mice; however, its high dose induced a significant decrease in STZ-DM animals. Upon conventional analysis, the SM standard uptake value was higher in STZ-DM mice, while MTF was virtually ineffective in either control or STZ-DM models. This metabolic reprogramming was not explained by any change in cytosolic glucose metabolism. By contrast, it closely agreed with the catalytic function of hexose-6P-dehydrogenase (H6PD; i.e., the trigger of a specific pentose phosphate pathway selectively located within the endoplasmic reticulum). In agreement with this role, the H6PD enzymatic response to both STZ-DM and MTF matched the activation of the NADPH-dependent antioxidant responses to the increased generation of reactive oxygen species caused by chronic hyperglycemia. Ex vivo analysis of tracer kinetics confirmed that the enhanced SM avidity for FDG occurred despite a significant reduction in glucose consumption, while it was associated with increased radioactivity transfer to the endoplasmic reticulum. CONCLUSIONS These data challenge the current dogma linking FDG uptake to the glycolytic rate. They instead introduce a new model considering a strict link between the uptake of this glucose analog, H6PD reticular activity, and oxidative damage in diabetes, at least under fasting condition.
Collapse
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genoa, Italy; Department of Health Sciences, University of Genoa, Via Pastore 1, 16132 Genoa, Italy
| | - Vanessa Cossu
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genoa, Italy; Department of Health Sciences, University of Genoa, Via Pastore 1, 16132 Genoa, Italy
| | - Patrizia Castellani
- Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genoa, Italy
| | - Patrizia Piccioli
- Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genoa, Italy
| | - Anna Maria Orengo
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genoa, Italy
| | - Laura Emionite
- Animal Facility, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genoa, Italy
| | - Francesco Di Giulio
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genoa, Italy
| | | | - Alberto Miceli
- Department of Health Sciences, University of Genoa, Via Pastore 1, 16132 Genoa, Italy
| | - Stefano Raffa
- Department of Health Sciences, University of Genoa, Via Pastore 1, 16132 Genoa, Italy
| | - Anna Borra
- Department of Health Sciences, University of Genoa, Via Pastore 1, 16132 Genoa, Italy
| | - Selene Capitanio
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genoa, Italy
| | - Silvia Morbelli
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genoa, Italy; Department of Health Sciences, University of Genoa, Via Pastore 1, 16132 Genoa, Italy
| | - Giacomo Caviglia
- Department Experimental Medicine, University of Genoa, Len Battista Alberti 2, 16132 Genoa, Italy
| | - Silvia Bruno
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy
| | - Silvia Ravera
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy
| | - Davide Maggi
- Diabetes Unit, IRCCS Ospedale Policlinico San Martino Genoa, Largo Benzi 10, 16132 Genoa, Italy; Department of Mathematics (DIMA), University of Genoa, Via Dodecaneso 35, 16146 Genoa, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genoa, Italy; Department of Health Sciences, University of Genoa, Via Pastore 1, 16132 Genoa, Italy
| | - Cecilia Marini
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132 Genoa, Italy; CNR Institute of Molecular Bioimaging and Physiology (IBFM), Via Fratelli Cervi 93, 20090 Segrate (MI), Italy.
| |
Collapse
|
20
|
Moreno-Altamirano MMB, Kolstoe SE, Sánchez-García FJ. Virus Control of Cell Metabolism for Replication and Evasion of Host Immune Responses. Front Cell Infect Microbiol 2019; 9:95. [PMID: 31058096 PMCID: PMC6482253 DOI: 10.3389/fcimb.2019.00095] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/22/2019] [Indexed: 12/11/2022] Open
Abstract
Over the last decade, there has been significant advances in the understanding of the cross-talk between metabolism and immune responses. It is now evident that immune cell effector function strongly depends on the metabolic pathway in which cells are engaged in at a particular point in time, the activation conditions, and the cell microenvironment. It is also clear that some metabolic intermediates have signaling as well as effector properties and, hence, topics such as immunometabolism, metabolic reprograming, and metabolic symbiosis (among others) have emerged. Viruses completely rely on their host's cell energy and molecular machinery to enter, multiply, and exit for a new round of infection. This review explores how viruses mimic, exploit or interfere with host cell metabolic pathways and how, in doing so, they may evade immune responses. It offers a brief outline of key metabolic pathways, mitochondrial function and metabolism-related signaling pathways, followed by examples of the mechanisms by which several viral proteins regulate host cell metabolic activity.
Collapse
Affiliation(s)
- María Maximina B Moreno-Altamirano
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Simon E Kolstoe
- School of Health Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Francisco Javier Sánchez-García
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
21
|
Szelényi P, Somogyi A, Sarnyai F, Zámbó V, Simon-Szabó L, Kereszturi É, Csala M. Microsomal pre-receptor cortisol production is inhibited by resveratrol and epigallocatechin gallate through different mechanisms. Biofactors 2019; 45:236-243. [PMID: 30496642 DOI: 10.1002/biof.1477] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/24/2018] [Accepted: 10/22/2018] [Indexed: 01/11/2023]
Abstract
Local activation of cortisol in hormone target tissues is a major determinant of glucocorticoid effect. Disorders in this peripheral cortisol metabolism play an important role in the development of metabolic diseases, such as obesity or type 2 diabetes mellitus. Hence, dietary factors influencing the activity of the involved enzymes can have major impacts on the risk of the above diseases. Resveratrol and epigallocatechin gallate (EGCG), two natural polyphenols found in several nutriments and in green tea, respectively, are well-known for their antiobesity and antidiabetic activities. EGCG has been shown to interfere with microsomal cortisol production through decreasing the luminal NADPH:NADP+ ratio. The aim of this study was to clarify if resveratrol also induces such a redox shift or causes any direct enzyme inhibition that influences local cortisol production. Cortisone-cortisol conversions and changes in NADPH levels were monitored in rat liver microsomal vesicles. Cortisol production was inhibited by resveratrol in a concentration dependent manner while the intrinsic reducing and oxidizing capacity as well as the NADPH level inside the ER-derived vesicles remained unaffected. Activity measurements performed in permeabilized microsomes confirmed that resveratrol, unlike EGCG, inhibits 11β-hydroxysteroid dehydrogenase type 1 directly. Long-term moderation of pre-receptor cortisol production likely contributes to the beneficial health effects of both polyphenols. © 2018 BioFactors, 45(2):236-243, 2019.
Collapse
Affiliation(s)
- Péter Szelényi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Anna Somogyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Farkas Sarnyai
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Veronika Zámbó
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Laura Simon-Szabó
- Pathobiochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University (MTA-SE), Budapest, Hungary
| | - Éva Kereszturi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Miklós Csala
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| |
Collapse
|
22
|
Goodman RP, Calvo SE, Mootha VK. Spatiotemporal compartmentalization of hepatic NADH and NADPH metabolism. J Biol Chem 2018; 293:7508-7516. [PMID: 29514978 PMCID: PMC5961030 DOI: 10.1074/jbc.tm117.000258] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Compartmentalization is a fundamental design principle of eukaryotic metabolism. Here, we review the compartmentalization of NAD+/NADH and NADP+/NADPH with a focus on the liver, an organ that experiences the extremes of biochemical physiology each day. Historical studies of the liver, using classical biochemical fractionation and measurements of redox-coupled metabolites, have given rise to the prevailing view that mitochondrial NAD(H) pools tend to be oxidized and important for energy homeostasis, whereas cytosolic NADP(H) pools tend to be highly reduced for reductive biosynthesis. Despite this textbook view, many questions still remain as to the relative size of these subcellular pools and their redox ratios in different physiological states, and to what extent such redox ratios are simply indicators versus drivers of metabolism. By performing a bioinformatic survey, we find that the liver expresses 352 known or predicted enzymes composing the hepatic NAD(P)ome, i.e. the union of all predicted enzymes producing or consuming NADP(H) or NAD(H) or using them as a redox co-factor. Notably, less than half are predicted to be localized within the cytosol or mitochondria, and a very large fraction of these genes exhibit gene expression patterns that vary during the time of day or in response to fasting or feeding. A future challenge lies in applying emerging new genetic tools to measure and manipulate in vivo hepatic NADP(H) and NAD(H) with subcellular and temporal resolution. Insights from such fundamental studies will be crucial in deciphering the pathogenesis of very common diseases known to involve alterations in hepatic NAD(P)H, such as diabetes and fatty liver disease.
Collapse
Affiliation(s)
- Russell P Goodman
- From the Division of Gastroenterology and
- Howard Hughes Medical Institute, and Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114 and
| | - Sarah E Calvo
- Howard Hughes Medical Institute, and Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114 and
- the Broad Institute, Cambridge, Massachusetts 02142
| | - Vamsi K Mootha
- Howard Hughes Medical Institute, and Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114 and
- the Broad Institute, Cambridge, Massachusetts 02142
| |
Collapse
|
23
|
Miller ML, Porollo A, Wert S. Ultrastructure of Highly Ordered Granules in Alveolar Type II Cells in Several Species. Anat Rec (Hoboken) 2018; 301:1290-1302. [PMID: 29544026 DOI: 10.1002/ar.23805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 11/30/2017] [Accepted: 02/02/2018] [Indexed: 01/26/2023]
Abstract
Alveolar Type II cells from seven mammalian species were examined for a protein in the rough endoplasmic reticulum (RER), which showed a multilayered, repeating motif. Each motif, 100 nm in width, comprised two parallel outer dense layers, a less dense central layer, and often 1-3 faint layers on either side of the latter. Outer layers showed periodicities at 3-4 densities/100 nm of width, while layers on either side of the central layer showed 5-7 densities/100 nm of width. RER membranes were ribosome-free when parallel to these layers, but showed four ribosomes per motif at the growing ends: one ribosome at each outer dense layer, and one on either side of the less dense central layer. Granules appeared as single or as multiple motifs, stacked, curved, folded, or branching together within the same RER profile. Hexagons of around 30 nm in diameter with central densities were seen in tangential cuts of outer dense layers. Granule incidence varied: guinea pig > ferret > dog. Possible homologous structures occurred in rabbit and cat, but not in rat or mouse. Surfactant protein A (SP-A), a C-type lectin produced in Type II cells, forms trimers and bouquet-like 18-mer and can oligomerize further. Two pairs of SP-A 18-mers with carbohydrate recognition domains pointing inwardly and outwardly, stacked vertically as a column of four molecules, then repeated side by side in rows, approximated the size and layering patterns observed in these granules. Sequence analyses of SP-A from these species showed phylogenetic distances consistent with the observed occurrence and frequency of patterned granules. Anat Rec, 301:1290-1302, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Marian L Miller
- Department of Environmental Health, University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Aleksey Porollo
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio.,Center for Autoimmune Genomics and Etiology, University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Susan Wert
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio.,Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
24
|
Zeng X, Cai D, Zeng Q, Chen Z, Zhong G, Zhuo J, Gan H, Huang X, Zhao Z, Yao N, Huang D, Zhang C, Sun D, Chen Y. Selective reduction in the expression of UGTs and SULTs, a novel mechanism by which piperine enhances the bioavailability of curcumin in rat. Biopharm Drug Dispos 2017; 38:3-19. [DOI: 10.1002/bdd.2049] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/10/2016] [Accepted: 11/16/2016] [Indexed: 11/12/2022]
Affiliation(s)
- Xiaohui Zeng
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
| | - Dake Cai
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
| | - Qiaohuang Zeng
- Department of Nephrology, The Second Clinical College; Guangzhou University of Chinese Medicine and Guangdong Provincial Academy of Chinese Medical Sciences; Guangzhou Guangdong PR China
- Guangzhou University of Chinese Medicine; Guangzhou Guangdong 510006 PR China
| | - Zhao Chen
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
| | - Guoping Zhong
- School of Pharmaceutical Science, Sun Yat-sen University; Guangzhou 510006 PR China
| | - Juncheng Zhuo
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangzhou University of Chinese Medicine; Guangzhou Guangdong 510006 PR China
| | - Haining Gan
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
| | - Xuejun Huang
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
| | - Ziming Zhao
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
| | - Nan Yao
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
| | - Dane Huang
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
| | - Chengzhe Zhang
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangzhou University of Chinese Medicine; Guangzhou Guangdong 510006 PR China
| | - Dongmei Sun
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
| | - Yuxing Chen
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
- Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine; Guangzhou Guangdong 510095 PR China
| |
Collapse
|
25
|
Marini C, Ravera S, Buschiazzo A, Bianchi G, Orengo AM, Bruno S, Bottoni G, Emionite L, Pastorino F, Monteverde E, Garaboldi L, Martella R, Salani B, Maggi D, Ponzoni M, Fais F, Raffaghello L, Sambuceti G. Discovery of a novel glucose metabolism in cancer: The role of endoplasmic reticulum beyond glycolysis and pentose phosphate shunt. Sci Rep 2016; 6:25092. [PMID: 27121192 PMCID: PMC4848551 DOI: 10.1038/srep25092] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/07/2016] [Indexed: 12/25/2022] Open
Abstract
Cancer metabolism is characterized by an accelerated glycolytic rate facing reduced activity of oxidative phosphorylation. This “Warburg effect” represents a standard to diagnose and monitor tumor aggressiveness with 18F-fluorodeoxyglucose whose uptake is currently regarded as an accurate index of total glucose consumption. Studying cancer metabolic response to respiratory chain inhibition by metformin, we repeatedly observed a reduction of tracer uptake facing a marked increase in glucose consumption. This puzzling discordance brought us to discover that 18F-fluorodeoxyglucose preferentially accumulates within endoplasmic reticulum by exploiting the catalytic function of hexose-6-phosphate-dehydrogenase. Silencing enzyme expression and activity decreased both tracer uptake and glucose consumption, caused severe energy depletion and decreased NADPH content without altering mitochondrial function. These data document the existence of an unknown glucose metabolism triggered by hexose-6-phosphate-dehydrogenase within endoplasmic reticulum of cancer cells. Besides its basic relevance, this finding can improve clinical cancer diagnosis and might represent potential target for therapy.
Collapse
Affiliation(s)
- Cecilia Marini
- CNR Institute of Molecular Bioimaging and Physiology (IBFM), Milan, Section of Genoa, Genoa, Italy.,Nuclear Medicine Unit, Department of Health Sciences, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Ambra Buschiazzo
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Anna Maria Orengo
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Silvia Bruno
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Gianluca Bottoni
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Laura Emionite
- Animal facility, IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Elena Monteverde
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Lucia Garaboldi
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Barbara Salani
- Department of Internal Medicine, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Davide Maggi
- Department of Internal Medicine, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Mirco Ponzoni
- Laboratorio di Oncologia, IRCCS G. Gaslini, Genoa, Italy
| | - Franco Fais
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Molecular Pathology, IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Gianmario Sambuceti
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| |
Collapse
|
26
|
Wang Z, Mick GJ, Xie R, Wang X, Xie X, Li G, McCormick KL. Cortisol promotes endoplasmic glucose production via pyridine nucleotide redox. J Endocrinol 2016; 229:25-36. [PMID: 26860459 DOI: 10.1530/joe-16-0006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 02/09/2016] [Indexed: 01/23/2023]
Abstract
Both increased adrenal and peripheral cortisol production, the latter governed by 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), contribute to the maintenance of fasting blood glucose. In the endoplasmic reticulum (ER), the pyridine nucleotide redox state (NADP/NADPH) is dictated by the concentration of glucose-6-phosphate (G6P) and the coordinated activities of two enzymes, hexose-6-phosphate dehydrogenase (H6PDH) and 11β-HSD1. However, luminal G6P may similarly serve as a substrate for hepatic glucose-6-phophatase (G6Pase). A tacit belief is that the G6P pool in the ER is equally accessible to both H6PDH and G6Pase. Based on our inhibition studies and kinetic analysis in isolated rat liver microsomes, these two aforesaid luminal enzymes do share the G6P pool in the ER, but not equally. Based on the kinetic modeling of G6P flux, the ER transporter for G6P (T1) preferentially delivers this substrate to G6Pase; hence, the luminal enzymes do not share G6P equally. Moreover, cortisol, acting through 11β-HSD1, begets a more reduced pyridine redox ratio. By altering this luminal redox ratio, G6P flux through H6PDH is restrained, allowing more G6P for the competing enzyme G6Pase. And, at low G6P concentrations in the ER lumen, which occur during fasting, this acute cortisol-induced redox adjustment promotes glucose production. This reproducible cortisol-driven mechanism has been heretofore unrecognized.
Collapse
Affiliation(s)
- Zengmin Wang
- Department of PediatricsShandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, PR China Division of Pediatric EndocrinologyUniversity of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Gail J Mick
- Division of Pediatric EndocrinologyUniversity of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rongrong Xie
- Division of Pediatric EndocrinologyUniversity of Alabama at Birmingham, Birmingham, Alabama, USA Department of EndocrinologyChildren's Hospital of Soochow University, Suzhou, Jiangsu, PR China
| | - Xudong Wang
- Division of Pediatric EndocrinologyUniversity of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xuemei Xie
- Division of Pediatric EndocrinologyUniversity of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Guimei Li
- Department of PediatricsShandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, PR China
| | - Kenneth L McCormick
- Division of Pediatric EndocrinologyUniversity of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
27
|
Zámbó V, Tóth M, Schlachter K, Szelényi P, Sarnyai F, Lotz G, Csala M, Kereszturi É. Cytosolic localization of NADH cytochrome b₅ oxidoreductase (Ncb5or). FEBS Lett 2016; 590:661-71. [PMID: 26878259 DOI: 10.1002/1873-3468.12097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 01/29/2016] [Accepted: 02/04/2016] [Indexed: 11/10/2022]
Abstract
Acyl-CoA desaturation in the endoplasmic reticulum (ER) membrane depends on cytosolic NADH or NADPH, whereas NADPH in the ER lumen is utilized by prereceptor glucocorticoid production. It was assumed that NADH cytochrome b5 oxidoreductase (Ncb5or) might connect Acyl-CoA desaturation to ER luminal redox. We aimed to clarify the ambiguous compartmentalization of Ncb5or and test the possible effect of stearoyl-CoA on microsomal NADPH level. Amino acid sequence analysis, fluorescence microscopy of GFP-tagged protein, immunocytochemistry, and western blot analysis of subcellular fractions unequivocally demonstrated that Ncb5or, either endogenous or exogenous, is localized in the cytoplasm and not in the ER lumen in cultured cells and liver tissue. Moreover, the involvement of ER-luminal reducing equivalents in stearoyl-CoA desaturation was excluded.
Collapse
Affiliation(s)
- Veronika Zámbó
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Mónika Tóth
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | | | - Péter Szelényi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Farkas Sarnyai
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Gábor Lotz
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Miklós Csala
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Éva Kereszturi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| |
Collapse
|
28
|
Abstract
Embryogenesis is a fascinating event during the plant life cycle encompassing several steps whereby the zygote develops into a fully developed embryo which, in angiosperms, is composed of an axis separating the apical meristems, and two cotyledons. Recapitulation of embryogenesis can also occur in vitro through somatic embryogenesis, where somatic cells are induced to form embryos, and androgenesis, in which embryos originate from immature male gametophytes. Besides cell division and differentiation, embryo patterning in vivo and in vitro requires the dismantling and selective elimination of cells and tissues via programmed cell death (PCD). While the manifestation of the death program has long been acknowledged in vivo, especially in relation to the elimination of the suspensor during the late phases of embryo development, PCD during in vitro embryogenesis has only been described in more recent years. Independent studies using the gymnosperm Norway spruce and the angiosperm maize have shown that the death program is crucial for the proper formation and further development of immature somatic embryos. This chapter summarizes the recent advances in the field of PCD during embryogenesis and proposes novel regulatory mechanisms activating the death program in plants.
Collapse
Affiliation(s)
- Shuanglong Huang
- Department of Plant Science, University of Manitoba, 222 Agriculture Building, Winnipeg, Canada, R3T2N2
| | - Mohamed M Mira
- Department of Botany, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Claudio Stasolla
- Department of Plant Science, University of Manitoba, 222 Agriculture Building, Winnipeg, Canada, R3T2N2.
| |
Collapse
|
29
|
Stone S, Lin W. The unfolded protein response in multiple sclerosis. Front Neurosci 2015; 9:264. [PMID: 26283904 PMCID: PMC4518158 DOI: 10.3389/fnins.2015.00264] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/14/2015] [Indexed: 01/08/2023] Open
Abstract
The unfolded protein response (UPR) occurs in response to endoplasmic reticulum (ER) stress caused by the accumulation of unfolded or misfolded proteins in the ER. The UPR is comprised of three signaling pathways that promote cytoprotective functions to correct ER stress; however, if ER stress cannot be resolved the UPR results in apoptosis of affected cells. The UPR is an important feature of various human diseases, including multiple sclerosis (MS). Recent studies have shown several components of the UPR are upregulated in the multiple cell types in MS lesions, including oligodendrocytes, T cells, microglia/macrophages, and astrocytes. Data from animal model studies, particularly studies of experimental autoimmune encephalomyelitis (EAE) and the cuprizone model, imply an important role of the UPR activation in oligodendrocytes in the development of MS. In this review we will cover current literature on the UPR and the evidence for its role in the development of MS.
Collapse
Affiliation(s)
- Sarrabeth Stone
- Department of Neuroscience, University of Minnesota Minneapolis, MN, USA ; Institute for Translational Neuroscience, University of Minnesota Minneapolis, MN, USA
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota Minneapolis, MN, USA ; Institute for Translational Neuroscience, University of Minnesota Minneapolis, MN, USA
| |
Collapse
|
30
|
Veit F, Pak O, Brandes RP, Weissmann N. Hypoxia-dependent reactive oxygen species signaling in the pulmonary circulation: focus on ion channels. Antioxid Redox Signal 2015; 22:537-52. [PMID: 25545236 PMCID: PMC4322788 DOI: 10.1089/ars.2014.6234] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE An acute lack of oxygen in the lung causes hypoxic pulmonary vasoconstriction, which optimizes gas exchange. In contrast, chronic hypoxia triggers a pathological vascular remodeling causing pulmonary hypertension, and ischemia can cause vascular damage culminating in lung edema. RECENT ADVANCES Regulation of ion channel expression and gating by cellular redox state is a widely accepted mechanism; however, it remains a matter of debate whether an increase or a decrease in reactive oxygen species (ROS) occurs under hypoxic conditions. Ion channel redox regulation has been described in detail for some ion channels, such as Kv channels or TRPC6. However, in general, information on ion channel redox regulation remains scant. CRITICAL ISSUES AND FUTURE DIRECTIONS In addition to the debate of increased versus decreased ROS production during hypoxia, we aim here at describing and deciphering why different oxidants, under different conditions, can cause both activation and inhibition of channel activity. While the upstream pathways affecting channel gating are often well described, we need a better understanding of redox protein modifications to be able to determine the complexity of ion channel redox regulation. Against this background, we summarize the current knowledge on hypoxia-induced ROS-mediated ion channel signaling in the pulmonary circulation.
Collapse
Affiliation(s)
- Florian Veit
- 1 Excellence Cluster Cardiopulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL) , Giessen, Germany
| | | | | | | |
Collapse
|
31
|
Petrov V, Hille J, Mueller-Roeber B, Gechev TS. ROS-mediated abiotic stress-induced programmed cell death in plants. FRONTIERS IN PLANT SCIENCE 2015; 6:69. [PMID: 25741354 PMCID: PMC4332301 DOI: 10.3389/fpls.2015.00069] [Citation(s) in RCA: 403] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/26/2015] [Indexed: 05/18/2023]
Abstract
During the course of their ontogenesis plants are continuously exposed to a large variety of abiotic stress factors which can damage tissues and jeopardize the survival of the organism unless properly countered. While animals can simply escape and thus evade stressors, plants as sessile organisms have developed complex strategies to withstand them. When the intensity of a detrimental factor is high, one of the defense programs employed by plants is the induction of programmed cell death (PCD). This is an active, genetically controlled process which is initiated to isolate and remove damaged tissues thereby ensuring the survival of the organism. The mechanism of PCD induction usually includes an increase in the levels of reactive oxygen species (ROS) which are utilized as mediators of the stress signal. Abiotic stress-induced PCD is not only a process of fundamental biological importance, but also of considerable interest to agricultural practice as it has the potential to significantly influence crop yield. Therefore, numerous scientific enterprises have focused on elucidating the mechanisms leading to and controlling PCD in response to adverse conditions in plants. This knowledge may help develop novel strategies to obtain more resilient crop varieties with improved tolerance and enhanced productivity. The aim of the present review is to summarize the recent advances in research on ROS-induced PCD related to abiotic stress and the role of the organelles in the process.
Collapse
Affiliation(s)
- Veselin Petrov
- Institute of Molecular Biology and Biotechnology, PlovdivBulgaria
| | - Jacques Hille
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Groningen, GroningenNetherlands
| | - Bernd Mueller-Roeber
- Department of Molecular Biology, Institute of Biochemistry and Biology, University of Potsdam, Potsdam-GolmGermany
| | - Tsanko S. Gechev
- Institute of Molecular Biology and Biotechnology, PlovdivBulgaria
- Department of Molecular Biology, Institute of Biochemistry and Biology, University of Potsdam, Potsdam-GolmGermany
| |
Collapse
|
32
|
Csala M, Kardon T, Legeza B, Lizák B, Mandl J, Margittai É, Puskás F, Száraz P, Szelényi P, Bánhegyi G. On the role of 4-hydroxynonenal in health and disease. Biochim Biophys Acta Mol Basis Dis 2015; 1852:826-38. [PMID: 25643868 DOI: 10.1016/j.bbadis.2015.01.015] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/16/2014] [Accepted: 01/23/2015] [Indexed: 02/08/2023]
Abstract
Polyunsaturated fatty acids are susceptible to peroxidation and they yield various degradation products, including the main α,β-unsaturated hydroxyalkenal, 4-hydroxy-2,3-trans-nonenal (HNE) in oxidative stress. Due to its high reactivity, HNE interacts with various macromolecules of the cell, and this general toxicity clearly contributes to a wide variety of pathological conditions. In addition, growing evidence suggests a more specific function of HNE in electrophilic signaling as a second messenger of oxidative/electrophilic stress. It can induce antioxidant defense mechanisms to restrain its own production and to enhance the cellular protection against oxidative stress. Moreover, HNE-mediated signaling can largely influence the fate of the cell through modulating major cellular processes, such as autophagy, proliferation and apoptosis. This review focuses on the molecular mechanisms underlying the signaling and regulatory functions of HNE. The role of HNE in the pathophysiology of cancer, cardiovascular and neurodegenerative diseases is also discussed.
Collapse
Affiliation(s)
- Miklós Csala
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary
| | - Tamás Kardon
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary
| | - Balázs Legeza
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Beáta Lizák
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary
| | - József Mandl
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary
| | - Éva Margittai
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Ferenc Puskás
- Department of Anesthesiology, University of Colorado, Denver, CO, USA
| | - Péter Száraz
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Péter Szelényi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary
| | - Gábor Bánhegyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary.
| |
Collapse
|
33
|
Hooper SL, Burstein HJ. Minimization of extracellular space as a driving force in prokaryote association and the origin of eukaryotes. Biol Direct 2014; 9:24. [PMID: 25406691 PMCID: PMC4289276 DOI: 10.1186/1745-6150-9-24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 11/03/2014] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Internalization-based hypotheses of eukaryotic origin require close physical association of host and symbiont. Prior hypotheses of how these associations arose include chance, specific metabolic couplings between partners, and prey-predator/parasite interactions. Since these hypotheses were proposed, it has become apparent that mixed-species, close-association assemblages (biofilms) are widespread and predominant components of prokaryotic ecology. Which forces drove prokaryotes to evolve the ability to form these assemblages are uncertain. Bacteria and archaea have also been found to form membrane-lined interconnections (nanotubes) through which proteins and RNA pass. These observations, combined with the structure of the nuclear envelope and an energetic benefit of close association (see below), lead us to propose a novel hypothesis of the driving force underlying prokaryotic close association and the origin of eukaryotes. RESULTS Respiratory proton transport does not alter external pH when external volume is effectively infinite. Close physical association decreases external volume. For small external volumes, proton transport decreases external pH, resulting in each transported proton increasing proton motor force to a greater extent. We calculate here that in biofilms this effect could substantially decrease how many protons need to be transported to achieve a given proton motor force. Based as it is solely on geometry, this energetic benefit would occur for all prokaryotes using proton-based respiration. CONCLUSIONS This benefit may be a driving force in biofilm formation. Under this hypothesis a very wide range of prokaryotic species combinations could serve as eukaryotic progenitors. We use this observation and the discovery of prokaryotic nanotubes to propose that eukaryotes arose from physically distinct, functionally specialized (energy factory, protein factory, DNA repository/RNA factory), obligatorily symbiotic prokaryotes in which the protein factory and DNA repository/RNA factory cells were coupled by nanotubes and the protein factory ultimately internalized the other two. This hypothesis naturally explains many aspects of eukaryotic physiology, including the nuclear envelope being a folded single membrane repeatedly pierced by membrane-bound tubules (the nuclear pores), suggests that species analogous or homologous to eukaryotic progenitors are likely unculturable as monocultures, and makes a large number of testable predictions. REVIEWERS This article was reviewed by Purificación López-García and Toni Gabaldón.
Collapse
Affiliation(s)
- Scott L Hooper
- Department of Biological Sciences, Ohio University, Athens, OH 45701 USA
| | - Helaine J Burstein
- Department of Biological Sciences, Ohio University, Athens, OH 45701 USA
| |
Collapse
|
34
|
Kaludercic N, Deshwal S, Di Lisa F. Reactive oxygen species and redox compartmentalization. Front Physiol 2014; 5:285. [PMID: 25161621 PMCID: PMC4130307 DOI: 10.3389/fphys.2014.00285] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/11/2014] [Indexed: 01/01/2023] Open
Abstract
Reactive oxygen species (ROS) formation and signaling are of major importance and regulate a number of processes in physiological conditions. A disruption in redox status regulation, however, has been associated with numerous pathological conditions. In recent years it has become increasingly clear that oxidative and reductive modifications are confined in a spatio-temporal manner. This makes ROS signaling similar to that of Ca(2+) or other second messengers. Some subcellular compartments are more oxidizing (such as lysosomes or peroxisomes) whereas others are more reducing (mitochondria, nuclei). Moreover, although more reducing, mitochondria are especially susceptible to oxidation, most likely due to the high number of exposed thiols present in that compartment. Recent advances in the development of redox probes allow specific measurement of defined ROS in different cellular compartments in intact living cells or organisms. The availability of these tools now allows simultaneous spatio-temporal measurements and correlation between ROS generation and organelle and/or cellular function. The study of ROS compartmentalization and microdomains will help elucidate their role in physiology and disease. Here we will examine redox probes currently available and how ROS generation may vary between subcellular compartments. Furthermore, we will discuss ROS compartmentalization in physiological and pathological conditions focusing our attention on mitochondria, since their vulnerability to oxidative stress is likely at the basis of several diseases.
Collapse
Affiliation(s)
- Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR) Padova, Italy
| | - Soni Deshwal
- Department of Biomedical Sciences, University of Padova Padova, Italy
| | - Fabio Di Lisa
- Neuroscience Institute, National Research Council of Italy (CNR) Padova, Italy ; Department of Biomedical Sciences, University of Padova Padova, Italy
| |
Collapse
|
35
|
Halperin L, Jung J, Michalak M. The many functions of the endoplasmic reticulum chaperones and folding enzymes. IUBMB Life 2014; 66:318-26. [PMID: 24839203 DOI: 10.1002/iub.1272] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 04/24/2014] [Indexed: 12/12/2022]
Abstract
Endoplasmic reticulum (ER) is an essential sub-cellular compartment of the eukaryotic cell performing many diverse functions essential for the cell and the whole organism. ER molecular chaperones and folding enzymes are multidomain proteins that are designed to support nascent proteins entering ER lumen to achieve their native conformation, mediate post-translational modification, prevent misfolded protein aggregation, and facilitate exit from the ER. Typically the role of ER chaperones expands beyond protein folding. Here, we illustrate the multifunctional nature of many ER associated molecular chaperones and folding enzymes and unique functional overlap of these proteins all designed to support the many functions of the ER membrane.
Collapse
Affiliation(s)
- Laura Halperin
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
36
|
|
37
|
Depletion of luminal pyridine nucleotides in the endoplasmic reticulum activates autophagy with the involvement of mTOR pathway. BIOMED RESEARCH INTERNATIONAL 2013; 2013:942431. [PMID: 24350295 PMCID: PMC3847967 DOI: 10.1155/2013/942431] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 10/03/2013] [Accepted: 10/07/2013] [Indexed: 11/17/2022]
Abstract
It has been recently shown that redox imbalance of luminal pyridine nucleotides in the endoplasmic reticulum (ER) together with oxidative stress results in the activation of autophagy. In the present study we demonstrated that decrease of luminal NADPH/NADP(+) ratio alone by metyrapone was sufficient to promote the mechanism of "self-eating" detected by the activation of LC3. Depletion of luminal NADPH had also significant effect on the key proteins of mTOR pathway, which got inactivated by dephosphorylation. These findings were also confirmed by silencing the proteins (glucose-6-phosphate transporter and hexose-6-phosphate dehydrogenase) responsible for NADPH generation in the ER lumen. However, silencing the key components and addition of metyrapone had different effects on downstream substrates 4EBP1 and p70S6K of mTOR. The applied treatments did not compromise the viability of the cells. Our data suggest that ER stress caused by luminal NADPH depletion activates a pro-survival autophagic mechanism firmly coupled to the activation of mTOR pathway.
Collapse
|
38
|
Zámbó V, Simon-Szabó L, Szelényi P, Kereszturi &E, Bánhegyi G, Csala M. Lipotoxicity in the liver. World J Hepatol 2013; 5:550-557. [PMID: 24179614 PMCID: PMC3812457 DOI: 10.4254/wjh.v5.i10.550] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 09/27/2013] [Accepted: 10/16/2013] [Indexed: 02/06/2023] Open
Abstract
Obesity due to excessive food intake and the lack of physical activity is becoming one of the most serious public health problems of the 21st century. With the increasing prevalence of obesity, non-alcoholic fatty liver disease is also emerging as a pandemic. While previously this pathophysiological condition was mainly attributed to triglyceride accumulation in hepatocytes, recent data show that the development of oxidative stress, lipid peroxidation, cell death, inflammation and fibrosis are mostly due to accumulation of fatty acids, and the altered composition of membrane phospholipids. In fact, triglyceride accumulation might play a protective role, and the higher toxicity of saturated or trans fatty acids seems to be the consequence of a blockade in triglyceride synthesis. Increased membrane saturation can profoundly disturb cellular homeostasis by impairing the function of membrane receptors, channels and transporters. However, it also induces endoplasmic reticulum stress via novel sensing mechanisms of the organelle’s stress receptors. The triggered signaling pathways in turn largely contribute to the development of insulin resistance and apoptosis. These findings have substantiated the lipotoxic liver injury hypothesis for the pathomechanism of hepatosteatosis. This minireview focuses on the metabolic and redox aspects of lipotoxicity and lipoapoptosis, with special regards on the involvement of endoplasmic reticulum stress responses.
Collapse
|
39
|
Szelényi P, Révész K, Konta L, Tüttõ A, Mandl J, Kereszturi É, Csala M. Inhibition of microsomal cortisol production by (-)-epigallocatechin-3-gallate through a redox shift in the endoplasmic reticulum--a potential new target for treating obesity-related diseases. Biofactors 2013; 39:534-41. [PMID: 23554216 DOI: 10.1002/biof.1095] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 01/09/2013] [Indexed: 01/18/2023]
Abstract
Conversion of cortisone to cortisol by 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1) in the endoplasmic reticulum (ER) of the target cells is a major determinant of glucocorticoid action, and plays an important role in the development of obesity-related diseases. Inhibition of 11βHSD1 activity is, therefore, considered as a promising novel strategy for the treatment of metabolic syndrome and diabetes. Tea flavanols and their major representative, epigallocatechin gallate are known as antiobesity and antidiabetic agents. Their impacts on blood glucose level, hepatic glucose production, and insulin responsiveness resemble those observed on inhibition or depletion of 11βHSD1. We aimed to study the effect of epigallocatechin gallate on 11βHSD1 activity in ER-derived rat liver microsomes by measuring cortisone and cortisol with HPLC. Cortisol production was efficiently suppressed in a concentration dependent manner in intact microsomal vesicles. However, this effect was abolished by membrane permeabilization; and the three proteins involved in the overall process (11βHSD1, hexose 6-phosphate dehydrogenase, and glucose 6-phosphate transporter) were not or only mildly affected. Further investigation revealed the oxidation of luminal NADPH to NADP⁺, which attenuates cortisone reduction and favors cortisol oxidation in this compartment. Such a redox shift in the ER lumen might contribute to the beneficial health effects of tea flavanols and should be regarded as a promising strategy for the development of novel selective 11βHSD1 inhibitors to treat obesity-related diseases.
Collapse
Affiliation(s)
- Péter Szelényi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
40
|
Révész K, Tóth B, Staines AG, Coughtrie MWH, Mandl J, Csala M. Luminal accumulation of newly synthesized morphine-3-glucuronide in rat liver microsomal vesicles. Biofactors 2013; 39:271-8. [PMID: 23281118 DOI: 10.1002/biof.1067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 10/16/2012] [Indexed: 01/23/2023]
Abstract
Morphine is converted to morphine 3-β-D-glucuronide (M3G) by the UDP-glucuronosyltransferase Ugt2b1 in the endoplasmic reticulum (ER) of rat liver. Because of its luminal localization, UGT activity requires UDP-glucuronate import and glucuronide export across the ER membrane. The former transport is generally considered to be rate limiting and to explain the latency of UGT activities in intact microsomal vesicles. However, some observations indicate that the release of bulky glucuronides, such as M3G, might also be rate limiting for glucuronidation. This assumption was tested by characterizing the transport of M3G and its distribution between the intra- and extravesicular spaces during synthesis in rat liver microsomes. The amount of vesicle-associated M3G was measured using rapid filtration and LC-MS measurement. Our results reveal a remarkable accumulation of newly synthesized M3G in the microsomal lumen above the equilibrium. The transport showed a linear concentration-dependence in a wide range (5-200 μM). Therefore, the build-up of high (about 20 μM) luminal M3G concentration could adjust the rate of release to that of synthesis (44.85 ± 4.08 pmol/min/mg protein) during the conjugation of 100 μM morphine. These data can explain earlier findings indicative of separate intracellular pools of M3G in rat liver. Accumulation of bulky glucuronides in the ER lumen might also play an important role in their targeting and in the control of biliary excretion.
Collapse
Affiliation(s)
- Katalin Révész
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | |
Collapse
|
41
|
Breitwieser GE. Minireview: the intimate link between calcium sensing receptor trafficking and signaling: implications for disorders of calcium homeostasis. Mol Endocrinol 2012; 26:1482-95. [PMID: 22745192 DOI: 10.1210/me.2011-1370] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The calcium-sensing receptor (CaSR) regulates organismal Ca(2+) homeostasis. Dysregulation of CaSR expression or mutations in the CASR gene cause disorders of Ca(2+) homeostasis and contribute to the progression or severity of cancers and cardiovascular disease. This brief review highlights recent findings that define the CaSR life cycle, which controls the cellular abundance of CaSR and CaSR signaling. A novel mechanism, termed agonist-driven insertional signaling (ADIS), contributes to the unique hallmarks of CaSR signaling, including the high degree of cooperativity and the lack of functional desensitization. Agonist-mediated activation of plasma membrane-localized CaSR increases the rate of insertion of CaSR at the plasma membrane without altering the constitutive endocytosis rate, thereby acutely increasing the maximum signaling response. Prolonged CaSR signaling requires a large intracellular ADIS-mobilizable pool of CaSR, which is maintained by signaling-mediated increases in biosynthesis. This model provides a rational framework for characterizing the defects caused by CaSR mutations and the altered functional expression of wild-type CaSR in disease states. Mechanistic dissection of ADIS of CaSR should lead to optimized pharmacological approaches to normalize CaSR signaling in disorders of Ca(2+) homeostasis.
Collapse
Affiliation(s)
- Gerda E Breitwieser
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania 17822-2604, USA.
| |
Collapse
|
42
|
Csala M, Kereszturi É, Mandl J, Bánhegyi G. The endoplasmic reticulum as the extracellular space inside the cell: role in protein folding and glycosylation. Antioxid Redox Signal 2012; 16:1100-8. [PMID: 22149109 DOI: 10.1089/ars.2011.4227] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE Proteins destined to secretion and exposure on the cell surface are synthesized and processed in the extracellular-like environment of the endoplasmic reticulum (ER) of higher eukaryotic cells. Compartmentation plays a crucial role in the post-translational modifications, such as oxidative folding and N-glycosylation in the ER lumen. Transport of the required intermediates across the ER membrane and maintenance of the luminal redox conditions and Ca(2+) ion concentration are indispensable for appropriate protein maturation. RECENT ADVANCES Cooperation of enzymes and transporters to maintain a thiol-oxidizing milieu in the ER lumen has been recently elucidated. Ca(2+)-dependence of certain ER chaperones is a subject of intensive research. CRITICAL ISSUES Mounting evidence supports the existence of a real barrier between the ER lumen and the cytosol. The unique set of enzymes, selection of metabolites, and characteristic ion and redox milieu of the luminal compartment strongly argue against the general permeability of the ER membrane. FUTURE DIRECTIONS Alterations in the luminal environment can trigger the unfolded protein response, a common event in a variety of pathological conditions. Therefore, redox and calcium homeostasis and protein glycosylation in the ER provide novel drug-targets for medical treatment in a wide array of diseases.
Collapse
Affiliation(s)
- Miklós Csala
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary.
| | | | | | | |
Collapse
|
43
|
Huang Y, Cavanaugh A, Breitwieser GE. Regulation of stability and trafficking of calcium-sensing receptors by pharmacologic chaperones. ADVANCES IN PHARMACOLOGY 2012; 62:143-73. [PMID: 21907909 DOI: 10.1016/b978-0-12-385952-5.00007-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gain- or loss-of-function mutations and polymorphisms of the calcium-sensing receptor (CaSR) cause Ca(2+) handling diseases. Altered expression and/or signaling of wild-type CaSR can also contribute to pathology. Recent studies have demonstrated that a significant proportion of mutations cause altered targeting and/or trafficking of CaSR to the plasma membrane. Pharmacological approaches to rescue of CaSR function include treatment with allosteric modulators, which potentiate the effects of the orthosteric agonist Ca(2+). Dissection of the mechanism(s) contributing to allosteric agonist-mediated rescue of loss-of-function CaSR mutants has demonstrated pharmacologic chaperone actions coincident with CaSR biosynthesis. The distinctive responses to the allosteric agonist (NPS R-568), which promotes CaSR stability, and the allosteric antagonist (NPS 2143), which promotes CaSR degradation, have led to a model for a conformational checkpoint during CaSR biosynthesis. The conformational checkpoint would "tune" CaSR biosynthesis to cellular signaling state. Navigation of a distinct checkpoint for endoplasmic release can also be augmented by pharmacologic chaperones. The diverse, post-endoplasmic reticulum quality control site(s) for pharmacologic chaperone modulation of CaSR stability and trafficking redefines the role(s) of allosteric modulators in regulation of overall GPCR function.
Collapse
Affiliation(s)
- Ying Huang
- Cancer Drug Research Laboratory, McGill University, Royal Victoria Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
44
|
Jia J, Wang Z, Li X, Wang Z, Wang X. Morphological characteristics and co-stimulatory molecule (CD80, CD86, CD40) expression in tumor infiltrating dendritic cells in human endometrioid adenocarcinoma. Eur J Obstet Gynecol Reprod Biol 2011; 160:223-7. [PMID: 22142817 DOI: 10.1016/j.ejogrb.2011.11.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 07/24/2011] [Accepted: 11/12/2011] [Indexed: 12/29/2022]
Abstract
OBJECTIVE The purpose of this study was to investigate changes of the antigen-presenting function of tumor infiltrating dendritic cells (TIDCs) in human endometrioid adenocarcinoma. STUDY DESIGN The TIDCs from 45 cases of endometrioid adenocarcinoma were compared with 20 cases of normal human endometrial tissue, using transmission electron microscopic examination, and the expression of CD80, CD86, and CD40 was analyzed by flow cytometry. RESULTS In comparison with the control group, the ultrastructure of TIDCs in human endometrioid adenocarcinoma showed the following differences: numerous TIDCs were small in volume and round in shape but some were oval and multi-angular. The cytoplasmic processes were obviously decreased in number and stubbed. Round primary lysosomes with high electron-dense granules, and secondary lysosomes with high or low electron-dense granules were seen frequently in the cytoplasm. TIDCs contained much rough endoplasmic reticulum (RER). Vacuoles with flocculent electron-dense granules were rare. High electron-dense contents in the granules were near one side and the other side was bright. The nucleus became markedly small in volume, nephroid or hoofed in shape. The nucleus had little euchromatin and lots of heterochromatin under the nuclear membrane. The levels of expression of CD80, CD86 and CD40 on TIDCs were low or even nonexistent. The expression levels of CD80, CD86 and CD40 on DCs in human normal endometrium were significantly higher than those on TIDCs in endometrioid adenocarcinoma. CONCLUSION It is suggested that morphological differences and low expression of co-stimulatory molecules on TIDCs in endometrioid adenocarcinoma reflected the functional changes of the TIDCs in uptake, processing and presenting antigen, which may lead to the occurrence of tumor immune escape.
Collapse
Affiliation(s)
- Jianjun Jia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Jinan University, China.
| | | | | | | | | |
Collapse
|
45
|
Live diatom silica immobilization of multimeric and redox-active enzymes. Appl Environ Microbiol 2011; 78:211-8. [PMID: 22057862 DOI: 10.1128/aem.06698-11] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Living organisms are adept in forming inorganic materials (biominerals) with unique structures and properties that exceed the capabilities of engineered materials. Biomimetic materials syntheses are being developed that aim at replicating the advantageous properties of biominerals in vitro and endow them with additional functionalities. Recently, proof-of-concept was provided for an alternative approach that allows for the production of biomineral-based functional materials in vivo. In this approach, the cellular machinery for the biosynthesis of nano-/micropatterned SiO₂ (silica) structures in diatoms was genetically engineered to incorporate a monomeric, cofactor-independent ("simple") enzyme, HabB, into diatom silica. In the present work, it is demonstrated that this approach is also applicable for enzymes with "complex" activity requirements, including oligomerization, metal ions, organic redox cofactors, and posttranslational modifications. Functional expression of the enzymes β-glucuronidase, glucose oxidase, galactose oxidase, and horseradish peroxidase in the diatom Thalassiosira pseudonana was accomplished, and 66 to 78% of the expressed enzymes were stably incorporated into the biosilica. The in vivo incorporated enzymes represent approximately 0.1% (wt/wt) of the diatom biosilica and are stabilized against denaturation and proteolytic degradation. Furthermore, it is demonstrated that the gene construct for in vivo immobilization of glucose oxidase can be utilized as the first negative selection marker for diatom genetic engineering.
Collapse
|
46
|
Révész K, Tüttő A, Szelényi P, Konta L. Tea flavan-3-ols as modulating factors in endoplasmic reticulum function. Nutr Res 2011; 31:731-40. [DOI: 10.1016/j.nutres.2011.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 09/07/2011] [Accepted: 09/15/2011] [Indexed: 01/04/2023]
|
47
|
Manifold effects of palmitoylcarnitine on endoplasmic reticulum metabolism: 11β-hydroxysteroid dehydrogenase 1, flux through hexose-6-phosphate dehydrogenase and NADPH concentration. Biochem J 2011; 437:109-15. [PMID: 21492096 DOI: 10.1042/bj20102069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
With the exception of the oxidation of G6P (glucose 6-phosphate) by H6PDH (hexose-6-phosphate dehydrogenase), scant information is available about other endogenous substrates affecting the redox state or the regulation of key enzymes which govern the ratio of the pyridine nucleotide NADPH/NADP. In isolated rat liver microsomes, NADPH production was increased, as anticipated, by G6P; however, this was strikingly amplified by palmitoylcarnitine. Subsequent experiments revealed that the latter compound, well within its physiological concentration range, inhibited 11β-HSD1 (11β-hydroxysteroid dehydrogenase 1), the bidirectional enzyme which interconnects inactive 11-oxo steroids and their active 11-hydroxy derivatives. Notably, palmitoylcarnitine also stimulated the antithetical direction of 11β-HSD1 reductase, namely dehydrogenase. This stimulation of H6PDH may have likewise contributed to the NADPH accretion. All told, the result of these enzyme modifications is, in a conjoint fashion, a sharp amplification of microsomal NADPH production. Neither the purified 11β-HSD1 nor that obtained following microsomal sonification were sensitive to palmitoylcarnitine inhibition. This suggests that the long-chain amphipathic acylcarnitines, given their favourable partitioning into the membrane lipid bilayer, disrupt the proficient kinetic and physical interplay between 11β-HSD1 and H6PDH. Finally, although IDH (isocitrate dehydrogenase) and malic enzyme are present in microsomes and increase NADPH concentration akin to that of G6P, neither had an effect on 11β-HSD1 reductase, evidence that the NADPH pool in the endoplasmic reticulum shared by the H6PDH/11β-HSD1 alliance is uncoupled from that governed by IDH and malic enzyme.
Collapse
|
48
|
Proulx-Bonneau S, Pratt J, Annabi B. A role for MT1-MMP as a cell death sensor/effector through the regulation of endoplasmic reticulum stress in U87 glioblastoma cells. J Neurooncol 2011; 104:33-43. [PMID: 21088866 DOI: 10.1007/s11060-010-0468-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 11/08/2010] [Indexed: 12/30/2022]
Abstract
Recent findings in cell death signalling show that membrane type 1 matrix metalloproteinase (MT1-MMP), an MMP known for its involvement in cancer cell invasion and metastasis, can act as a "bioswitch" in the invasion versus cell death decision in brain tumour cells. Given that the endoplasmic reticulum (ER) is a subcellular compartment involved in metabolic control and cell death signalling and that cytoskeleton disruption, as encountered during cancer cell invasion, can lead to ER stress, we questioned whether MT1-MMP contributes to ER stress. We found that MT1-MMP gene silencing or pharmacological inhibition of vesicular trafficking with Brefeldin-A abrogated MT1-MMP cell surface-mediated proMMP-2 activation by the lectin Concanavalin-A (ConA) in U87 glioblastoma cells. ConA, also known to trigger the expression of pro-inflammatory cyclooxygenase (COX)-2 through MT1-MMP signalling from the plasma membrane, failed to do so when MT1-MMP was prevented from reaching the cell surface by Brefeldin-A. Gene silencing of MT1-MMP antagonized the expression of ConA-induced COX-2 and of the ER stress marker glucose-related protein 78 (GRP78), further suggesting that plasma membrane localization of MT1-MMP contributes to signalling ER stress. MT1-MMP maturation, which partially occurs during its trafficking from the ER to the plasma membrane, showed correlation of the 60 kDa MT1-MMP with GRP78 expression. Finally, Brefeldin-A treatment of glioblastoma cells led to Akt dephosphorylation; this effect was reversed when MT1-MMP was silenced. Collectively, our results provide a molecular rationale for a new role for MT1-MMP in the regulation of cancer cell death processes through ER stress signalling.
Collapse
Affiliation(s)
- Sébastien Proulx-Bonneau
- Laboratoire d'Oncologie Moléculaire, Département de Chimie, Centre de Recherche BioMED, Université du Québec à Montréal, C.P. 8888, Succ. Centre-ville, Montreal, QC, H3C 3P8, Canada
| | | | | |
Collapse
|
49
|
Zielinska AE, Walker EA, Stewart PM, Lavery GG. Biochemistry and physiology of hexose-6-phosphate knockout mice. Mol Cell Endocrinol 2011; 336:213-8. [PMID: 21146583 DOI: 10.1016/j.mce.2010.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 12/01/2010] [Accepted: 12/02/2010] [Indexed: 10/18/2022]
Abstract
Hexose-6-phosphate dehydrogenase (H6PDH) has emerged as an important factor in setting the redox status of the endoplasmic reticulum (ER) lumen. An important role of H6PDH is to generate a high NADPH/NADP(+) ratio which permits 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) to act as an oxo-reductase, catalyzing the activation of glucocorticoids (GCs). In H6PDH knockout mice 11β-HSD1 assumes dehydrogenase activity and inactivates GCs, rendering the target cell relatively GC insensitive. Consequently, H6PDHKO mice have a phenotype consistent with defects in the permissive and adaptive actions of GCs upon physiology. H6PDHKO mice have also offered an insight into muscle physiology as they also present with a severe vacuolating myopathy, abnormalities of glucose homeostasis and activation of the unfolded protein response due to ER stress, and a number of mechanisms driving this phenotype are thought to be involved. This article will review what we understand of the redox control of GC hormone metabolism regulated by H6PDH, and how H6PDHKO mice have allowed an in-depth understanding of its potentially novel, GC-independent roles in muscle physiology.
Collapse
Affiliation(s)
- Agnieszka E Zielinska
- Centre for Endocrinology, Diabetes and Metabolism, School of Clinical and Experimental Medicine, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | | | | |
Collapse
|
50
|
Abraham P, Isaac B. Ultrastructural changes in the rat kidney after single dose of cyclophosphamide—Possible roles for peroxisome proliferation and lysosomal dysfunction in cyclophosphamide-induced renal damage. Hum Exp Toxicol 2011; 30:1924-30. [DOI: 10.1177/0960327111402240] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Electron microscopy was used to examine changes in the subcellular organelles of the rat kidney at different time intervals after a single exposure to cyclophosphamide (CP). The morphological changes were studied at different time points (6 hrs, 16 hrs and 24 hrs) after a single-dose administration of CP. Six rats were killed at each time intervals after the administration of CP. Saline-treated rats served as controls. CP administration resulted in alterations in various subcellular organelles including peroxisomes, lysosomes, mitochondria, and the endoplasmic reticulum (ER) of the renal tubular epithelium as well as damage to the glomerulus. The basement membrane of the glomerulus was thickened. Many podocytes were destroyed. The nucleoplasm of the endothelial cell showed fewer granularities. The tubules were distorted and the brush border was destroyed. Two striking features in the renal tubular cells are increase in number and size of the peroxisomes (peroxisome proliferation) and decrease in the number of lysosomes. The mitochondria were elongated and the number was increased in the tubules of CP-treated rats. The ER was dilated. Cell necrosis was also seen. This study is an evidence of changes in morphology of rat kidney after induction of renal damage by a single dose of CP. Since transmission electron microscopy is the highest magnification tool at present, it can be useful in estimating the degree of injury and outcome of alternative treatment strategies in the management of CP-induced renal damage after establishing a scoring system.
Collapse
Affiliation(s)
- Premila Abraham
- Department of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, India
| | - Bina Isaac
- Department of Anatomy, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|