151
|
Astrocyte strategies in the energy-efficient brain. Essays Biochem 2023; 67:3-16. [PMID: 36350053 DOI: 10.1042/ebc20220077] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022]
Abstract
Astrocytes generate ATP through glycolysis and mitochondrion respiration, using glucose, lactate, fatty acids, amino acids, and ketone bodies as metabolic fuels. Astrocytic mitochondria also participate in neuronal redox homeostasis and neurotransmitter recycling. In this essay, we aim to integrate the multifaceted evidence about astrocyte bioenergetics at the cellular and systems levels, with a focus on mitochondrial oxidation. At the cellular level, the use of fatty acid β-oxidation and the existence of molecular switches for the selection of metabolic mode and fuels are examined. At the systems level, we discuss energy audits of astrocytes and how astrocytic Ca2+ signaling might contribute to the higher performance and lower energy consumption of the brain as compared to engineered circuits. We finish by examining the neural-circuit dysregulation and behavior impairment associated with alterations of astrocytic mitochondria. We conclude that astrocytes may contribute to brain energy efficiency by coupling energy, redox, and computational homeostasis in neural circuits.
Collapse
|
152
|
Long-term plasticity of astrocytic phenotypes and their control by neurons in health and disease. Essays Biochem 2023; 67:39-47. [PMID: 36695493 PMCID: PMC10011399 DOI: 10.1042/ebc20220090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/26/2023]
Abstract
The brain is a complex organ even when viewed from a cell biological perspective. Neuronal networks are embedded in a dense milieu of diverse and specialised cell types, including several types of vascular, immune, and macroglial cells. To view each cell as a small cog in a highly complex machine is itself an oversimplification. Not only are they functionally coupled to enable the brain to operate, each cell type's functions are themselves influenced by each other, in development, maturity, and also in disease. Astrocytes are a type of macroglia that occupy a significant fraction of the human forebrain. They play a critical role in sustaining functional neuronal circuits across the lifespan through myriad homeostatic functions including the maintenance of redox balance, ionic gradients, neurotransmitter clearance, and bioenergetic support. It is becoming apparent that astrocytes' capacity to carry out these and other neurosupportive roles is not fixed, but is regulated by signals coming from the neurons themselves, both in the healthy brain but also in response to neuron-derived disease pathology. Here, we review mechanisms by which neurons control the properties of astrocytes long term in order to alter their homeostatic capacity both in development and maturity. Our working hypothesis is that these signals are designed to change and maintain the homeostatic capacity of local astrocytes to suit the needs of nearby neurons. Knowledge of the external signals that can control core aspects of a healthy astrocytic phenotype are being uncovered, raising the question as to whether this knowledge can be harnessed to promote astrocyte-mediated neurosupport in brain disorders.
Collapse
|
153
|
Cross-talk between energy and redox metabolism in astrocyte-neuron functional cooperation. Essays Biochem 2023; 67:17-26. [PMID: 36805653 PMCID: PMC10011404 DOI: 10.1042/ebc20220075] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/23/2023]
Abstract
Astrocytes show unique anatomical, morphological, and metabolic features to take up substrates from the blood and metabolize them for local delivery to active synapses to sustain neuron function. In the present review, we specifically focus on key molecular aspects of energy and redox metabolism that facilitate this astrocyte-neuronal coupling in a controlled manner. Basal glycolysis is co-ordinated by the anaphase-promoting complex/cyclosome (APC/C)-Cdh1, a ubiquitin ligase that targets the proglycolytic enzyme 6-phosphofructokinase-2,6-bisphosphastate-3 (PFKFB3) for degradation. APC/C-Cdh1 activity is more robust in neurons than in astrocytes, which determine that PFKFB3 abundance and glycolytic rate are weaker in neurons. The low PFKFB3 activity in neurons facilitates glucose-6-phosphate oxidation via the pentose-phosphate pathway, which promotes antioxidant protection. Conversely, the high PFKFB3 activity in astrocytes allows the production and release of glycolytic lactate, which is taken up by neurons that use it as an oxidizable substrate. Importantly, the mitochondrial respiratory chain is tighter assembled in neurons than in astrocytes, thus the bioenergetic efficiency of mitochondria is higher in neurons. Because of this, the production of reactive oxygen species (mROS) by mitochondrial complex I is very low in neurons and very high in astrocytes. Such a naturally occurring high abundance of mROS in astrocytes physiologically determines a specific transcriptional fingerprint that contributes to sustaining cognitive performance. We conclude that the energy and redox metabolism of astrocytes must complementarily match that of neurons to regulate brain function and animal welfare.
Collapse
|
154
|
Brymer KJ, Hurley EP, Barron JC, Mukherjee B, Barnes JR, Nafar F, Parsons MP. Asymmetric dysregulation of glutamate dynamics across the synaptic cleft in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2023; 11:27. [PMID: 36788598 PMCID: PMC9926626 DOI: 10.1186/s40478-023-01524-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/28/2023] [Indexed: 02/16/2023] Open
Abstract
Most research on glutamate spillover focuses on the deleterious consequences of postsynaptic glutamate receptor overactivation. However, two decades ago, it was noted that the glial coverage of hippocampal synapses is asymmetric: astrocytic coverage of postsynaptic sites exceeds coverage of presynaptic sites by a factor of four. The fundamental relevance of this glial asymmetry remains poorly understood. Here, we used the glutamate biosensor iGluSnFR, and restricted its expression to either CA3 or CA1 neurons to visualize glutamate dynamics at pre- and postsynaptic microenvironments, respectively. We demonstrate that inhibition of the primarily astrocytic glutamate transporter-1 (GLT-1) slows glutamate clearance to a greater extent at presynaptic compared to postsynaptic membranes. GLT-1 expression was reduced early in a mouse model of AD, resulting in slower glutamate clearance rates at presynaptic but not postsynaptic membranes that opposed presynaptic short-term plasticity. Overall, our data demonstrate that the presynapse is particularly vulnerable to GLT-1 dysfunction and may have implications for presynaptic impairments in a variety of brain diseases.
Collapse
Affiliation(s)
- Kyle J. Brymer
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Emily P. Hurley
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Jessica C. Barron
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Bandhan Mukherjee
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Jocelyn R. Barnes
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Firoozeh Nafar
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Matthew P. Parsons
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| |
Collapse
|
155
|
Saranya J, Saminathan P, Ankireddy SR, Shaik MR, Khan M, Khan M, Shaik B. Cerium Oxide/Graphene Oxide Hybrid: Synthesis, Characterization, and Evaluation of Anticancer Activity in a Breast Cancer Cell Line (MCF-7). Biomedicines 2023; 11:biomedicines11020531. [PMID: 36831067 PMCID: PMC9952927 DOI: 10.3390/biomedicines11020531] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
In the present study, we used a simple ultrasonic approach to develop a Cerium oxide/Graphene oxide hybrid (CeO2/GO hybrid) nanocomposite system. Particle size analysis, Fourier Transform Infrared Spectroscopy (FTIR), Scanning Electron Microscopy (SEM), and X-ray Diffraction (XRD) have been used to analyze the physio-chemical characteristics of the developed nanocomposite. The synthesized hybrid system has also been examined to assess its anticancer capability against MCF-7 cell lines and normal cell lines at different sample concentrations, pH values, and incubation intervals using an antiproliferative assay test. The test results demonstrate that as sample concentration rises, the apoptotic behavior of the CeO2/GO hybrid in the MCF-7 cell line also rises. The IC50 was 62.5 µg/mL after 72 h of incubation. Cytotoxicity of cisplatin bound CeO2/GO hybrid was also tested in MCF-7 cell lines. To identify apoptosis-associated alterations of cell membranes during the process of apoptosis, a dual acridine orange/ethidium bromide (AO/EB) fluorescence staining was carried out at three specified doses (i.e., 1000 µg/mL, 250 µg/mL, and 62.5 µg/mL of CeO2/GO hybrid). The color variations from both live (green) and dead (red) cells were examined using fluorescence microscopy under in vitro conditions. The quantitative analysis was performed using flow cytometry to identify the cell cycle at which the maximum number of MCF-7 cells had been destroyed as a result of interaction with the developed CeO2/GO hybrid (FACS study). According to the results of the FACS investigation, the majority of cancer cells were inhibited at the R3 (G2/M) phase. Therefore, the CeO2/GO hybrid has successfully showed enhanced anticancer efficacy against the MCF-7 cell line at the IC50 concentration. According to the current study, the CeO2/GO platform can be used as a therapeutic platform for breast cancer. The synergetic effects of the developed CeO2/GO hybrid with the MCF-7 cell line are presented.
Collapse
Affiliation(s)
- J. Saranya
- Department of Electronics and Communication Engineering, Rajalakshmi Engineering College, Chennai 602105, Tamil Nadu, India
- Correspondence: (J.S.); (M.R.S.); Tel.: +966-11-4670439 (M.R.S.)
| | - P. Saminathan
- Sasaam Biologicals Lab Services, Ashok Nagar, Chennai 600083, Tamil Nadu, India
| | - Seshadri Reddy Ankireddy
- Dr. Buddolla’s Institute of Life Sciences, Renigunta Road, Tirupati 517503, Andhra Pradesh, India
| | - Mohammed Rafi Shaik
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
- Correspondence: (J.S.); (M.R.S.); Tel.: +966-11-4670439 (M.R.S.)
| | - Mujeeb Khan
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Merajuddin Khan
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Baji Shaik
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
156
|
Melatonin-Assisted Cisplatin Suppresses Urinary Bladder Cancer Cell Proliferation and Growth through Inhibiting PrP C-Regulated Cell Stress and Cell Proliferation Signaling. Int J Mol Sci 2023; 24:ijms24043353. [PMID: 36834767 PMCID: PMC9959909 DOI: 10.3390/ijms24043353] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
This study investigated whether melatonin (Mel) would promote cisplatin to suppress the proliferation and growth of bladder cancer (BC) cells by inhibiting cellular prion protein (PrPC)-mediated cell stress and cell proliferation signaling. An immunohistochemical staining of tissue arrays from BC patients demonstrated that the PrPC expression was significantly upregulated from stage I to III BC (p < 0.0001). The BC cellline of T24 was categorized into G1 (T24), G2 (T24 + Mel/100 μM), G3 (T24+cisplatin/6 μM), G4 (PrPC overexpression in T24 (i.e., PrPC-OE-T24)), G5 (PrPC-OE-T24+Mel), and G6 (PrPC-OE-T24+cisplatin). When compared with a human uroepithelial cell line (SV-HUC-1), the cellular viability/wound healing ability/migration rate were significantly increased in T24 cells (G1) and further significantly increased in PrPC-OE-T24 cells (G4); and they were suppressed in Mel (G2/G5) or cisplatin (G3/G6) treatment (all p < 0.0001). Additionally, the protein expressions of cell proliferation (PI3K/p-Akt/p-m-TOR/MMP-9/PrPC), cell cycle/mitochondrial functional integrity (cyclin-D1/clyclin-E1/ckd2/ckd4/mitochondrial-cytochrome-C/PINK1), and cell stress (RAS/c-RAF/p-MEK1/2, p-ERK1/2) markers showed a similar pattern of cell viability among the groups (all p < 0.001). After the BC cell line of UMUC3 was implanted into nude mouse backs, by day 28 mthe BC weight/volume and the cellular levels of PrPC/MMP-2/MMP-9 were significantly, gradually reduced from groups one to four (all p < 0.0001). The protein expressions of cell proliferation (PI3K/p-Akt/p-m-TOR/MMP-9/PrPC), cell cycle/mitophagy (cyclin-D1/clyclin-E1/ckd2/ckd4/PINK1), and cell stress (RAS/c-RAF/p-MEK1,2/p-ERK1,2) signaling were significantly, progressively reduced from groups one to four, whereas the protein expressions of apoptotic (Mit-Bax/cleaved-caspase-3/cleaved-PARP) and oxidative stress/mitochondrial damaged (NOX-1/NOX-2/cytosolic-cytochrome-C/p-DRP1) markers expressed an opposite pattern of cell proliferation signaling among the groups (all p < 0.0001). Mel-cisplatin suppressed BC cell growth/proliferation via inhibiting the PrPC in upregulating the cell proliferation/cell stress/cell cycle signaling.
Collapse
|
157
|
Type I Diabetes Pathoetiology and Pathophysiology: Roles of the Gut Microbiome, Pancreatic Cellular Interactions, and the 'Bystander' Activation of Memory CD8 + T Cells. Int J Mol Sci 2023; 24:ijms24043300. [PMID: 36834709 PMCID: PMC9964837 DOI: 10.3390/ijms24043300] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/10/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) arises from the failure of pancreatic β-cells to produce adequate insulin, usually as a consequence of extensive pancreatic β-cell destruction. T1DM is classed as an immune-mediated condition. However, the processes that drive pancreatic β-cell apoptosis remain to be determined, resulting in a failure to prevent ongoing cellular destruction. Alteration in mitochondrial function is clearly the major pathophysiological process underpinning pancreatic β-cell loss in T1DM. As with many medical conditions, there is a growing interest in T1DM as to the role of the gut microbiome, including the interactions of gut bacteria with Candida albicans fungal infection. Gut dysbiosis and gut permeability are intimately associated with raised levels of circulating lipopolysaccharide and suppressed butyrate levels, which can act to dysregulate immune responses and systemic mitochondrial function. This manuscript reviews broad bodies of data on T1DM pathophysiology, highlighting the importance of alterations in the mitochondrial melatonergic pathway of pancreatic β-cells in driving mitochondrial dysfunction. The suppression of mitochondrial melatonin makes pancreatic β-cells susceptible to oxidative stress and dysfunctional mitophagy, partly mediated by the loss of melatonin's induction of PTEN-induced kinase 1 (PINK1), thereby suppressing mitophagy and increasing autoimmune associated major histocompatibility complex (MHC)-1. The immediate precursor to melatonin, N-acetylserotonin (NAS), is a brain-derived neurotrophic factor (BDNF) mimic, via the activation of the BDNF receptor, TrkB. As both the full-length and truncated TrkB play powerful roles in pancreatic β-cell function and survival, NAS is another important aspect of the melatonergic pathway relevant to pancreatic β-cell destruction in T1DM. The incorporation of the mitochondrial melatonergic pathway in T1DM pathophysiology integrates wide bodies of previously disparate data on pancreatic intercellular processes. The suppression of Akkermansia muciniphila, Lactobacillus johnsonii, butyrate, and the shikimate pathway-including by bacteriophages-contributes to not only pancreatic β-cell apoptosis, but also to the bystander activation of CD8+ T cells, which increases their effector function and prevents their deselection in the thymus. The gut microbiome is therefore a significant determinant of the mitochondrial dysfunction driving pancreatic β-cell loss as well as 'autoimmune' effects derived from cytotoxic CD8+ T cells. This has significant future research and treatment implications.
Collapse
|
158
|
González A, Tapia JA. Special issue: 2022 pancreatic cancer and the role of the stroma in the pathophysiology of the gland. J Physiol Biochem 2023; 79:175-177. [PMID: 36604403 DOI: 10.1007/s13105-022-00942-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/21/2022] [Indexed: 01/07/2023]
Affiliation(s)
- Antonio González
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, Avenida de las Ciencias s/n, E-10003, Cáceres, Spain.
| | - José Antonio Tapia
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, Avenida de las Ciencias s/n, E-10003, Cáceres, Spain
| |
Collapse
|
159
|
Estaras M, Ortiz-Placin C, Castillejo-Rufo A, Fernandez-Bermejo M, Blanco G, Mateos JM, Vara D, Gonzalez-Cordero PL, Chamizo S, Lopez D, Rojas A, Jaen I, de Armas N, Salido GM, Iovanna JL, Santofimia-Castaño P, Gonzalez A. Melatonin controls cell proliferation and modulates mitochondrial physiology in pancreatic stellate cells. J Physiol Biochem 2023; 79:235-249. [PMID: 36334253 PMCID: PMC9905253 DOI: 10.1007/s13105-022-00930-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
We have investigated the effects of melatonin on major pathways related with cellular proliferation and energetic metabolism in pancreatic stellate cells. In the presence of melatonin (1 mM, 100 µM, 10 µM, or 1 µM), decreases in the phosphorylation of c-Jun N-terminal kinase and of p44/42 and an increase in the phosphorylation of p38 were observed. Cell viability dropped in the presence of melatonin. A rise in the phosphorylation of AMP-activated protein kinase was detected in the presence of 1 mM and 100 µM melatonin. Treatment with 1 mM melatonin decreased the phosphorylation of protein kinase B, whereas 100 µM and 10 µM melatonin increased its phosphorylation. An increase in the generation of mitochondrial reactive oxygen species and a decrease of mitochondrial membrane potential were noted following melatonin treatment. Basal and maximal respiration, ATP production by oxidative phosphorylation, spare capacity, and proton leak dropped in the presence of melatonin. The expression of complex I of the mitochondrial respiratory chain was augmented in the presence of melatonin. Conversely, in the presence of 1 mM melatonin, decreases in the expression of mitofusins 1 and 2 were detected. The glycolysis and the glycolytic capacity were diminished in cells treated with 1 mM or 100 µM melatonin. Increases in the expression of phosphofructokinase-1 and lactate dehydrogenase were noted in cells incubated with 100 µM, 10 µM, or 1 µM melatonin. The expression of glucose transporter 1 was increased in cells incubated with 10 µM or 1 µM melatonin. Conversely, 1 mM melatonin decreased the expression of all three proteins. Our results suggest that melatonin, at pharmacological concentrations, might modulate mitochondrial physiology and energy metabolism in addition to major pathways involved in pancreatic stellate cell proliferation.
Collapse
Affiliation(s)
- Matias Estaras
- Departamento de Fisiología, Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Avenida de Las Ciencias S/N, 10003, Cáceres, Spain
| | - Candido Ortiz-Placin
- Departamento de Fisiología, Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Avenida de Las Ciencias S/N, 10003, Cáceres, Spain
| | - Alba Castillejo-Rufo
- Departamento de Fisiología, Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Avenida de Las Ciencias S/N, 10003, Cáceres, Spain
| | | | - Gerardo Blanco
- Unidad de Cirugía Hepatobiliopancreática Y Transplante Hepático, Hospital Universitario, Badajoz, Spain
| | - Jose M Mateos
- Departamento de Gastroenterología, Hospital Universitario, Cáceres, Spain
| | - Daniel Vara
- Departamento de Gastroenterología, Hospital Universitario, Cáceres, Spain
| | | | - Sandra Chamizo
- Departamento de Gastroenterología, Hospital Universitario, Cáceres, Spain
| | - Diego Lopez
- Unidad de Cirugía Hepatobiliopancreática Y Transplante Hepático, Hospital Universitario, Badajoz, Spain
| | - Adela Rojas
- Unidad de Cirugía Hepatobiliopancreática Y Transplante Hepático, Hospital Universitario, Badajoz, Spain
| | - Isabel Jaen
- Unidad de Cirugía Hepatobiliopancreática Y Transplante Hepático, Hospital Universitario, Badajoz, Spain
| | - Noelia de Armas
- Unidad de Cirugía Hepatobiliopancreática Y Transplante Hepático, Hospital Universitario, Badajoz, Spain
| | - Gines M Salido
- Departamento de Fisiología, Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Avenida de Las Ciencias S/N, 10003, Cáceres, Spain
| | - Juan L Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique Et Technologique de Luminy, Marseille, France
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique Et Technologique de Luminy, Marseille, France
| | - Antonio Gonzalez
- Departamento de Fisiología, Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Avenida de Las Ciencias S/N, 10003, Cáceres, Spain.
| |
Collapse
|
160
|
Olivares-Caro L, Nova-Baza D, Radojkovic C, Bustamante L, Duran D, Mennickent D, Melin V, Contreras D, Perez AJ, Mardones C. Berberis microphylla G. Forst Intake Reduces the Cardiovascular Disease Plasmatic Markers Associated with a High-Fat Diet in a Mice Model. Antioxidants (Basel) 2023; 12:antiox12020304. [PMID: 36829862 PMCID: PMC9952125 DOI: 10.3390/antiox12020304] [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: 01/05/2023] [Revised: 01/13/2023] [Accepted: 01/21/2023] [Indexed: 01/31/2023] Open
Abstract
Polyphenols are bioactive substances that participate in the prevention of chronic illnesses. High content has been described in Berberis microphylla G. Forst (calafate), a wild berry extensively distributed in Chilean-Argentine Patagonia. We evaluated its beneficial effect through the study of mouse plasma metabolome changes after chronic consumption of this fruit. Characterized calafate extract was administered in water, for four months, to a group of mice fed with a high-fat diet and compared with a control diet. Metabolome changes were studied using UHPLC-DAD-QTOF-based untargeted metabolomics. The study was complemented by the analysis of protein biomarkers determined using Luminex technology, and quantification of OH radicals by electron paramagnetic resonance spectroscopy. Thirteen features were identified with a maximum annotation level-A, revealing an increase in succinic acid, activation of tricarboxylic acid and reduction of carnitine accumulation. Changes in plasma biomarkers were related to inflammation and cardiovascular disease, with changes in thrombomodulin (-24%), adiponectin (+68%), sE-selectin (-34%), sICAM-1 (-24%) and proMMP-9 (-31%) levels. The production of OH radicals in plasma was reduced after calafate intake (-17%), especially for the group fed with a high-fat diet. These changes could be associated with protection against atherosclerosis due to calafate consumption, which is discussed from a holistic and integrative point of view.
Collapse
Affiliation(s)
- Lia Olivares-Caro
- Departamento de Análisis Instrumental, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile
| | - Daniela Nova-Baza
- Departamento de Análisis Instrumental, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile
| | - Claudia Radojkovic
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile
| | - Luis Bustamante
- Departamento de Análisis Instrumental, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile
| | - Daniel Duran
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile
| | - Daniela Mennickent
- Departamento de Análisis Instrumental, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile
| | - Victoria Melin
- Departamento de Ingeniería Mecánica, Facultad de Ingeniería, Universidad de Tarapacá, Avda. General Velásquez 1775, Arica 1000007, Chile
| | - David Contreras
- Departamento de Química Analítica e Inorgánica, Facultad de Ciencias Químicas, Universidad de Concepción, Concepción 4070386, Chile
| | - Andy J. Perez
- Departamento de Análisis Instrumental, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile
| | - Claudia Mardones
- Departamento de Análisis Instrumental, Facultad de Farmacia, Universidad de Concepción, Concepción 4070386, Chile
- Unidad de Desarrollo Tecnológico, Universidad de Concepción, Coronel 4191996, Chile
- Correspondence: ; Tel.: +56-983616340
| |
Collapse
|
161
|
Ishii T, Warabi E, Mann GE. Stress Activated MAP Kinases and Cyclin-Dependent Kinase 5 Mediate Nuclear Translocation of Nrf2 via Hsp90α-Pin1-Dynein Motor Transport Machinery. Antioxidants (Basel) 2023; 12:antiox12020274. [PMID: 36829834 PMCID: PMC9952688 DOI: 10.3390/antiox12020274] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/19/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
Non-lethal low levels of oxidative stress leads to rapid activation of the transcription factor nuclear factor-E2-related factor 2 (Nrf2), which upregulates the expression of genes important for detoxification, glutathione synthesis, and defense against oxidative damage. Stress-activated MAP kinases p38, ERK, and JNK cooperate in the efficient nuclear accumulation of Nrf2 in a cell-type-dependent manner. Activation of p38 induces membrane trafficking of a glutathione sensor neutral sphingomyelinase 2, which generates ceramide upon depletion of cellular glutathione. We previously proposed that caveolin-1 in lipid rafts provides a signaling hub for the phosphorylation of Nrf2 by ceramide-activated PKCζ and casein kinase 2 to stabilize Nrf2 and mask a nuclear export signal. We further propose a mechanism of facilitated Nrf2 nuclear translocation by ERK and JNK. ERK and JNK phosphorylation of Nrf2 induces the association of prolyl cis/trans isomerase Pin1, which specifically recognizes phosphorylated serine or threonine immediately preceding a proline residue. Pin1-induced structural changes allow importin-α5 to associate with Nrf2. Pin1 is a co-chaperone of Hsp90α and mediates the association of the Nrf2-Pin1-Hsp90α complex with the dynein motor complex, which is involved in transporting the signaling complex to the nucleus along microtubules. In addition to ERK and JNK, cyclin-dependent kinase 5 could phosphorylate Nrf2 and mediate the transport of Nrf2 to the nucleus via the Pin1-Hsp90α system. Some other ERK target proteins, such as pyruvate kinase M2 and hypoxia-inducible transcription factor-1, are also transported to the nucleus via the Pin1-Hsp90α system to modulate gene expression and energy metabolism. Notably, as malignant tumors often express enhanced Pin1-Hsp90α signaling pathways, this provides a potential therapeutic target for tumors.
Collapse
Affiliation(s)
- Tetsuro Ishii
- School of Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
- Correspondence:
| | - Eiji Warabi
- School of Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Giovanni E. Mann
- King’s British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, 150 Stamford Street, London SE1 9NH, UK
| |
Collapse
|
162
|
Cuellar-Santoyo AO, Ruiz-Rodríguez VM, Mares-Barbosa TB, Patrón-Soberano A, Howe AG, Portales-Pérez DP, Miquelajáuregui Graf A, Estrada-Sánchez AM. Revealing the contribution of astrocytes to glutamatergic neuronal transmission. Front Cell Neurosci 2023; 16:1037641. [PMID: 36744061 PMCID: PMC9893894 DOI: 10.3389/fncel.2022.1037641] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Research on glutamatergic neurotransmission has focused mainly on the function of presynaptic and postsynaptic neurons, leaving astrocytes with a secondary role only to ensure successful neurotransmission. However, recent evidence indicates that astrocytes contribute actively and even regulate neuronal transmission at different levels. This review establishes a framework by comparing glutamatergic components between neurons and astrocytes to examine how astrocytes modulate or otherwise influence neuronal transmission. We have included the most recent findings about the role of astrocytes in neurotransmission, allowing us to understand the complex network of neuron-astrocyte interactions. However, despite the knowledge of synaptic modulation by astrocytes, their contribution to specific physiological and pathological conditions remains to be elucidated. A full understanding of the astrocyte's role in neuronal processing could open fruitful new frontiers in the development of therapeutic applications.
Collapse
Affiliation(s)
- Ares Orlando Cuellar-Santoyo
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Victor Manuel Ruiz-Rodríguez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Teresa Belem Mares-Barbosa
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | - Araceli Patrón-Soberano
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Andrew G. Howe
- Intelligent Systems Laboratory, HRL Laboratories, LLC, Malibu, CA, United States
| | - Diana Patricia Portales-Pérez
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | | | - Ana María Estrada-Sánchez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| |
Collapse
|
163
|
ARMS-NF-κB signaling regulates intracellular ROS to induce autophagy-associated cell death upon oxidative stress. iScience 2023; 26:106005. [PMID: 36798436 PMCID: PMC9926119 DOI: 10.1016/j.isci.2023.106005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/23/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Ankyrin repeat-rich membrane spanning (ARMS) plays roles in neural development, neuropathies, and tumor formation. Such pleiotropic function of ARMS is often attributed to diverse ARMS-interacting molecules in different cell context. However, it might be achieved by ARMS' effect on global biological mediator like reactive oxygen species (ROS). We established ARMS-knockdown in melanoma cells (siARMS) and in Drosophila eyes (GMR>dARMS RNAi ) and challenged them with H2O2. Decreased ARMS in both systems compromises nuclear translocation of NF-κB and induces ROS, which in turn augments autophagy flux and confers susceptibility to H2O2-triggered autophagic cell death. Resuming NF-κB activity or reducing ROS by antioxidants in siARMS cells and GMR>dARMS RNAi fly decreases intracellular peroxides level concurrent with reduced autophagy and attenuated cell death. Conversely, blocking NF-κB activity in wild-type flies/melanoma enhances ROS and induces autophagy with cell death. We thus uncover intracellular ROS modulated by ARMS-NFκB signaling primes autophagy for autophagic cell death upon oxidative stress.
Collapse
|
164
|
Fan Y, Huang H, Shao J, Huang W. MicroRNA-mediated regulation of reactive astrocytes in central nervous system diseases. Front Mol Neurosci 2023; 15:1061343. [PMID: 36710937 PMCID: PMC9877358 DOI: 10.3389/fnmol.2022.1061343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
Astrocytes (AST) are abundant glial cells in the human brain, accounting for approximately 20-50% percent of mammalian central nervous system (CNS) cells. They display essential functions necessary to sustain the physiological processes of the CNS, including maintaining neuronal structure, forming the blood-brain barrier, coordinating neuronal metabolism, maintaining the extracellular environment, regulating cerebral blood flow, stabilizing intercellular communication, participating in neurotransmitter synthesis, and defending against oxidative stress et al. During the pathological development of brain tumors, stroke, spinal cord injury (SCI), neurodegenerative diseases, and other neurological disorders, astrocytes undergo a series of highly heterogeneous changes, which are called reactive astrocytes, and mediate the corresponding pathophysiological process. However, the pathophysiological mechanisms of reactive astrocytes and their therapeutic relevance remain unclear. The microRNAs (miRNAs) are essential for cell differentiation, proliferation, and survival, which play a crucial role in the pathophysiological development of CNS diseases. In this review, we summarize the regulatory mechanism of miRNAs on reactive astrocytes in CNS diseases, which might provide a theoretical basis for the diagnosis and treatment of CNS diseases.
Collapse
|
165
|
Astrain-Redin N, Talavera I, Moreno E, Ramírez MJ, Martínez-Sáez N, Encío I, Sharma AK, Sanmartín C, Plano D. Seleno-Analogs of Scaffolds Resembling Natural Products a Novel Warhead toward Dual Compounds. Antioxidants (Basel) 2023; 12:139. [PMID: 36671001 PMCID: PMC9854712 DOI: 10.3390/antiox12010139] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Nowadays, oxidative cell damage is one of the common features of cancer and Alzheimer's disease (AD), and Se-containing molecules, such as ebselen, which has demonstrated strong antioxidant activity, have demonstrated well-established preventive effects against both diseases. In this study, a total of 39 Se-derivatives were synthesized, purified, and spectroscopically characterized by NMR. Antioxidant ability was tested using the DPPH assay, while antiproliferative activity was screened in breast, lung, prostate, and colorectal cancer cell lines. In addition, as a first approach to evaluate their potential anti-Alzheimer activity, the in vitro acetylcholinesterase inhibition (AChEI) was tested. Regarding antioxidant properties, compound 13a showed concentration- and time-dependent radical scavenging activity. Additionally, compounds 14a and 17a showed high activity in the melanoma and ovarian cancer cell lines, with LD50 values below 9.2 µM. Interestingly, in the AChEI test, compound 14a showed almost identical inhibitory activity to galantamine along with a 3-fold higher in vitro BBB permeation (Pe = 36.92 × 10-6 cm/s). Molecular dynamics simulations of the aspirin derivatives (14a and 14b) confirm the importance of the allylic group instead of the propargyl one. Altogether, it is concluded that some of these newly synthesized Se-derivatives, such as 14a, might become very promising candidates to treat both cancer and AD.
Collapse
Affiliation(s)
- Nora Astrain-Redin
- Departamento de Tecnología y Química Farmacéuticas, Facultad de Farmacia y Nutrición, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | - Irene Talavera
- Departamento de Tecnología y Química Farmacéuticas, Facultad de Farmacia y Nutrición, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | - Esther Moreno
- Departamento de Tecnología y Química Farmacéuticas, Facultad de Farmacia y Nutrición, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | - María J. Ramírez
- Departamento de Farmacología y Toxicología, Facultad de Farmacia y Nutrición, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | - Nuria Martínez-Sáez
- Departamento de Tecnología y Química Farmacéuticas, Facultad de Farmacia y Nutrición, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | - Ignacio Encío
- Departamento de Ciencias de la Salud, Universidad Pública de Navarra, Avda. Barañain s/n, E-31008 Pamplona, Spain
| | - Arun K. Sharma
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Carmen Sanmartín
- Departamento de Tecnología y Química Farmacéuticas, Facultad de Farmacia y Nutrición, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | - Daniel Plano
- Departamento de Tecnología y Química Farmacéuticas, Facultad de Farmacia y Nutrición, Universidad de Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| |
Collapse
|
166
|
Schubert R, Gaynullina D, Shvetsova A, Tarasova OS. Myography of isolated blood vessels: Considerations for experimental design and combination with supplementary techniques. Front Physiol 2023; 14:1176748. [PMID: 37168231 PMCID: PMC10165122 DOI: 10.3389/fphys.2023.1176748] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
The study of the mechanisms of regulation of vascular tone is an urgent task of modern science, since diseases of the cardiovascular system remain the main cause of reduction in the quality of life and mortality of the population. Myography (isometric and isobaric) of isolated blood vessels is one of the most physiologically relevant approaches to study the function of cells in the vessel wall. On the one hand, cell-cell interactions as well as mechanical stretch of the vessel wall remain preserved in myography studies, in contrast to studies on isolated cells, e.g., cell culture. On the other hand, in vitro studies in isolated vessels allow control of numerous parameters that are difficult to control in vivo. The aim of this review was to 1) discuss the specifics of experimental design and interpretation of data obtained by myography and 2) highlight the importance of the combined use of myography with various complementary techniques necessary for a deep understanding of vascular physiology.
Collapse
Affiliation(s)
- Rudolf Schubert
- Physiology, Institute of Theoretical Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- *Correspondence: Rudolf Schubert,
| | - Dina Gaynullina
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | | | - Olga S. Tarasova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
- State Research Center of the Russian Federation, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
167
|
Smith PJ, McKeown SR, Patterson LH. Targeting DNA topoisomerase IIα (TOP2A) in the hypoxic tumour microenvironment using unidirectional hypoxia-activated prodrugs (uHAPs). IUBMB Life 2023; 75:40-54. [PMID: 35499745 PMCID: PMC10084299 DOI: 10.1002/iub.2619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/24/2022] [Accepted: 04/03/2022] [Indexed: 12/29/2022]
Abstract
The hypoxic tumour microenvironment (hTME), arising from inadequate and chaotic vascularity, can present a major obstacle for the treatment of solid tumours. Hypoxic tumour cells compromise responses to treatment since they can generate resistance to radiotherapy, chemotherapy and immunotherapy. The hTME impairs the delivery of a range of anti-cancer drugs, creates routes for metastasis and exerts selection pressures for aggressive phenotypes; these changes potentially occur within an immunosuppressed environment. Therapeutic strategies aimed at the hTME include targeting the molecular changes associated with hypoxia. An alternative approach is to exploit the prevailing lack of oxygen as a principle for the selective activation of prodrugs to target cellular components within the hTME. This review focuses on the design concepts and rationale for the use of unidirectional Hypoxia-Activated Prodrugs (uHAPs) to target the hTME as exemplified by the uHAPs AQ4N and OCT1002. These agents undergo irreversible reduction in a hypoxic environment to active forms that target DNA topoisomerase IIα (TOP2A). This nuclear enzyme is essential for cell division and is a recognised chemotherapeutic target. An activated uHAP interacts with the enzyme-DNA complex to induce DNA damage, cell cycle arrest and tumour cell death. uHAPs are designed to overcome the shortcomings of conventional HAPs and offer unique pharmacodynamic properties for effective targeting of TOP2A in the hTME. uHAP therapy in combination with standard of care treatments has the potential to enhance outcomes by co-addressing the therapeutic challenge presented by the hTME.
Collapse
Affiliation(s)
- Paul J Smith
- Cancer and Genetics Division, School of Medicine, Cardiff University, Cardiff, UK
| | | | - Laurence H Patterson
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
168
|
Li F, Wu X, Liang Y, Wu W. Potential implications of oxidative modification on dietary protein nutritional value: A review. Compr Rev Food Sci Food Saf 2023; 22:714-751. [PMID: 36527316 DOI: 10.1111/1541-4337.13090] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/06/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Abstract
During food processing and storage, proteins are sensitive to oxidative modification, changing the structural characteristics and functional properties. Recently, the impact of dietary protein oxidation on body health has drawn increasing attention. However, few reviews summarized and highlighted the impact of oxidative modification on the nutritional value of dietary proteins and related mechanisms. Therefore, this review seeks to give an updated discussion of the effects of oxidative modification on the structural characteristics and nutritional value of dietary proteins, and elucidate the interaction with gut microbiota, intestinal tissues, and organs. Additionally, the specific mechanisms related to pathological conditions are also characterized. Dietary protein oxidation during food processing and storage change protein structure, which further influences the in vitro digestion properties of proteins. In vivo research demonstrates that oxidized dietary proteins threaten body health via complicated pathways and affect the intestinal microenvironment via gut microbiota, metabolites, and intestinal morphology. This review highlights the influence of oxidative modification on the nutritional value of dietary proteins based on organs and the intestinal tract, and illustrates the necessity of appropriate experimental design for comprehensively exploring the health consequences of oxidized dietary proteins.
Collapse
Affiliation(s)
- Fang Li
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P. R. China.,National Engineering Research Center of Rice and Byproduct Deep Processing, Changsha, Hunan, P. R. China
| | - Xiaojuan Wu
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P. R. China.,National Engineering Research Center of Rice and Byproduct Deep Processing, Changsha, Hunan, P. R. China
| | - Ying Liang
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P. R. China.,National Engineering Research Center of Rice and Byproduct Deep Processing, Changsha, Hunan, P. R. China
| | - Wei Wu
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P. R. China.,National Engineering Research Center of Rice and Byproduct Deep Processing, Changsha, Hunan, P. R. China
| |
Collapse
|
169
|
Lu C, Gao S, Zhang L, Shi X, Chen Y, Wei S, Zuo L, Zhang L. Nuclear Protein 1 Expression Is Associated with PPARG in Bladder Transitional Cell Carcinoma. PPAR Res 2023; 2023:6797694. [PMID: 37197716 PMCID: PMC10185424 DOI: 10.1155/2023/6797694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/20/2023] [Accepted: 04/20/2023] [Indexed: 05/19/2023] Open
Abstract
Background The Nuclear protein 1 gene was first discovered in acute pancreatitis and functions as an oncogene in cancer progression and drug resistance. However, the role of Nuclear protein 1 in bladder transitional cell carcinoma (BTCC) is still unclear. Methods The Cancer Genome Atlas database and immunohistochemical analysis were adopted to evaluate Nuclear protein 1 expression in BTCC. We applied lentivirus-mediated small-interfering RNA to down-regulate the expression of Nuclear protein 1 in BTCC cell lines. We further performed an Affymetrix microarray and Gene Set Enrichment Analysis (GSEA) to assess the genes and signaling pathways related to Nuclear protein 1. Results We found that Nuclear protein 1 expression was up-regulated in BTCC and positively related to the degree of BTCC malignancy. Compared with Caucasian patients with BTCC, Nuclear protein 1 expression was attenuated in Asian patients. The Affymetrix microarray showed that lipopolysaccharide was the upstream regulatory factor of Nuclear protein 1 in BTCC. The GSEA indicated that Nuclear protein 1 expression was associated with signaling pathways in cancer, peroxisome proliferator-activated receptor (PPAR) pathways, and RNA degradation. The expression of Nuclear protein 1 was negatively correlated with PPARG (R = -0.290, P < 0.001), but not with PPARA (R = 0.047, P = 0.344) and PPARD (R = -0.055, P = 0.260). Conclusions The study findings indicate that Nuclear protein 1 is positively associated with the malignancy degree of BTCC and that Nuclear protein 1 expression is negatively correlated with PPARG.
Collapse
Affiliation(s)
- Chao Lu
- Department of Urology, Changzhou Second People's Hospital, 29 Xinglong Road, Changzhou 213003, China
| | - Shenglin Gao
- Department of Urology, Changzhou Second People's Hospital, 29 Xinglong Road, Changzhou 213003, China
| | - Li Zhang
- Department of Urology, Changzhou Second People's Hospital, 29 Xinglong Road, Changzhou 213003, China
| | - Xiaokai Shi
- Department of Urology, Changzhou Second People's Hospital, 29 Xinglong Road, Changzhou 213003, China
| | - Yin Chen
- Department of Urology, Changzhou Second People's Hospital, 29 Xinglong Road, Changzhou 213003, China
| | - Shuzhang Wei
- Department of Urology, Changzhou Second People's Hospital, 29 Xinglong Road, Changzhou 213003, China
| | - Li Zuo
- Department of Urology, Changzhou Second People's Hospital, 29 Xinglong Road, Changzhou 213003, China
| | - Lifeng Zhang
- Department of Urology, Changzhou Second People's Hospital, 29 Xinglong Road, Changzhou 213003, China
| |
Collapse
|
170
|
Li B, Yang Y, Ding Y, Ge Y, Xu Y, Xie Y, Shi Y, Le G. Dityrosine in food: A review of its occurrence, health effects, detection methods, and mitigation strategies. Compr Rev Food Sci Food Saf 2023; 22:355-379. [PMID: 36382862 DOI: 10.1111/1541-4337.13071] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/07/2022] [Accepted: 10/15/2022] [Indexed: 11/17/2022]
Abstract
Protein and amino acid oxidation in food products produce many new compounds, of which the reactive and toxic compound dityrosine, derived from oxidized tyrosine, is the most widely studied. The high reactivity of dityrosine enables this compound to induce oxidative stress and disrupt thyroid hormone function, contributing to the pathological processes of several diseases, such as obesity, diabetes, cognitive dysfunction, aging, and age-related diseases. From the perspective of food safety and human health, protein-oxidation products in food are the main concern of consumers, health management departments, and the food industry. This review highlights the latest research on the formation pathways, toxicity, detection methods, occurrence in food, and mitigation strategies for dityrosine. Furthermore, the control of dityrosine in family cooking and food-processing industry has been discussed. Food-derived dityrosine primarily originates from high-protein foods, such as meat and dairy products. Considering its toxicity, combining rapid high sensitivity dityrosine detection techniques with feasible control methods could be an effective strategy to ensure food safety and maintain human health. However, the current dityrosine detection and mitigation strategies exhibit some inherent characteristics and limitations. Therefore, developing technologies for rapid and effective dityrosine detection and control at the industrial level is necessary.
Collapse
Affiliation(s)
- Bowen Li
- National Engineering Laboratory/Key Laboratory of Henan Province, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, Henan Province, 450001, China.,State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, 214122, China
| | - Yuhui Yang
- National Engineering Laboratory/Key Laboratory of Henan Province, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, Henan Province, 450001, China
| | - Yinyi Ding
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang Province, 310018, China
| | - Yueting Ge
- College of Life Science, Xinyang Normal University, Xinyang, Henan Province, 464000, China
| | - Yuncong Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yanli Xie
- National Engineering Laboratory/Key Laboratory of Henan Province, College of Food Science and Engineering, Henan University of Technology, Zhengzhou, Henan Province, 450001, China
| | - Yonghui Shi
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, 214122, China
| | - Guowei Le
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, 214122, China
| |
Collapse
|
171
|
Díaz-Velasco S, Delgado J, Peña FJ, Estévez M. Ellagic Acid Triggers the Necrosis of Differentiated Human Enterocytes Exposed to 3-Nitro-Tyrosine: An MS-Based Proteomic Study. Antioxidants (Basel) 2022; 11:antiox11122485. [PMID: 36552693 PMCID: PMC9774974 DOI: 10.3390/antiox11122485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
To study the molecular basis of the toxicological effect of a dietary nitrosated amino acid, namely, 3-nitrotyrosine (3-NT), differentiated human enterocytes were exposed to dietary concentrations of this species (200 μM) and analyzed for flow cytometry, protein oxidation markers and MS-based proteomics. The possible protective role of a dietary phytochemical, ellagic acid (EA) (200 μM), was also tested. The results revealed that cell viability was significantly affected by exposure to 3-NT, with a concomitant significant increase in necrosis (p < 0.05). 3-NT affected several biological processes, such as histocompatibility complex class II (MHC class II), and pathways related to type 3 metabotropic glutamate receptors binding. Addition of EA to 3-NT-treated cells stimulated the toxicological effects of the latter by reducing the abundance of proteins involved in mitochondrial conformation. These results emphasize the impact of dietary nitrosated amino acids in intestinal cell physiology and warn about the potential negative effects of ellagic acid when combined with noxious metabolites.
Collapse
Affiliation(s)
- Silvia Díaz-Velasco
- Food Technology and Quality (TECAL), Institute of Meat and Meat Products (IPROCAR), Universidad de Extremadura, 10003 Cáceres, Spain
| | - Josué Delgado
- Food Hygiene and Safety (HISEALI), Institute of Meat and Meat Products (IPROCAR), Universidad de Extremadura, 10003 Cáceres, Spain
| | - Fernando J. Peña
- Spermatology Laboratory, Universidad de Extremadura, 10003 Cáceres, Spain
| | - Mario Estévez
- Food Technology and Quality (TECAL), Institute of Meat and Meat Products (IPROCAR), Universidad de Extremadura, 10003 Cáceres, Spain
- Correspondence:
| |
Collapse
|
172
|
Torres FF, Bernardo VS, de Paula CP, da Silva JPMDO, de Almeida EA, da Cunha AF, da Silva DGH. Influence of Melatonin Treatment on Cellular Mechanisms of Redox Adaptation in K562 Erythroleukemic Cells. Genes (Basel) 2022; 13:genes13122337. [PMID: 36553603 PMCID: PMC9778059 DOI: 10.3390/genes13122337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Melatonin (MEL) presents well-documented pleiotropic actions against oxidative stress (OS), acting indirectly through activation of transcription factors, e.g., FoxO3 and Nrf2. Thus, this study aimed to investigate the possible modulating effects of MEL on the redox signaling pathways PI3K/AKT/FoxO3 and Keap1/Nrf2/ARE in K562 erythroleukemic cells subjected to OS induction. For this, the viability, and transcript levels of genes involved in redox adaptation were evaluated in K562 cells in different periods of erythroid differentiation: under OS induction by hydrogen peroxide (100 µM H2O2); treated with 1 nM (C1) and 1 mM (C2) MEL; and associated or not with stress induction. We observed a restoration of physiological levels of Nrf2 in both MEL concentrations under OS. The C1 was related to enhanced expression of antioxidant and proteasome genes through the Nrf2-ARE pathway, while C2 to the induction of FOXO3 expression, suggesting an involvement with apoptotic pathway, according to BIM transcript levels. The effects of MEL administration in these cells showed a period and dose-dependent pattern against induced-OS, with direct and indirect actions through different pathways of cellular adaptation, reinforcing the importance of this indolamine in the regulation of cellular homeostasis, being a promising therapeutic alternative for diseases that present an exacerbated OS.
Collapse
Affiliation(s)
- Flaviene Felix Torres
- Department of Biology, Universidade Estadual Paulista (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Victoria Simões Bernardo
- Department of Biology, Universidade Estadual Paulista (UNESP), São José do Rio Preto 15054-000, SP, Brazil
| | - Carla Peres de Paula
- Department of Genetics and Evolution, Universidade Federal de São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil
| | | | - Eduardo Alves de Almeida
- Department of Natural Sciences, Fundação Universidade Regional de Blumenau (FURB), Blumenau 89030-000, SC, Brazil
| | - Anderson Ferreira da Cunha
- Department of Genetics and Evolution, Universidade Federal de São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil
| | - Danilo Grünig Humberto da Silva
- Campus de Três Lagoas, Universidade Federal de Mato Grosso do Sul (CPTL/UFMS), Três Lagoas 79613-000, MS, Brazil
- Correspondence:
| |
Collapse
|
173
|
Vujić T, Schvartz D, Furlani IL, Meister I, González-Ruiz V, Rudaz S, Sanchez JC. Oxidative Stress and Extracellular Matrix Remodeling Are Signature Pathways of Extracellular Vesicles Released upon Morphine Exposure on Human Brain Microvascular Endothelial Cells. Cells 2022; 11:cells11233926. [PMID: 36497184 PMCID: PMC9741159 DOI: 10.3390/cells11233926] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/25/2022] [Accepted: 11/01/2022] [Indexed: 12/09/2022] Open
Abstract
Morphine, a commonly used antinociceptive drug in hospitals, is known to cross the blood-brain barrier (BBB) by first passing through brain endothelial cells. Despite its pain-relieving effect, morphine also has detrimental effects, such as the potential induction of redox imbalance in the brain. However, there is still insufficient evidence of these effects on the brain, particularly on the brain endothelial cells and the extracellular vesicles that they naturally release. Indeed, extracellular vesicles (EVs) are nanosized bioparticles produced by almost all cell types and are currently thought to reflect the physiological state of their parent cells. These vesicles have emerged as a promising source of biomarkers by indicating the functional or dysfunctional state of their parent cells and, thus, allowing a better understanding of the biological processes involved in an adverse state. However, there is very little information on the morphine effect on human brain microvascular endothelial cells (HBMECs), and even less on their released EVs. Therefore, the current study aimed at unraveling the detrimental mechanisms of morphine exposure (at 1, 10, 25, 50 and 100 µM) for 24 h on human brain microvascular endothelial cells as well as on their associated EVs. Isolation of EVs was carried out using an affinity-based method. Several orthogonal techniques (NTA, western blotting and proteomics analysis) were used to validate the EVs enrichment, quality and concentration. Data-independent mass spectrometry (DIA-MS)-based proteomics was applied in order to analyze the proteome modulations induced by morphine on HBMECs and EVs. We were able to quantify almost 5500 proteins in HBMECs and 1500 proteins in EVs, of which 256 and 148, respectively, were found to be differentially expressed in at least one condition. Pathway enrichment analysis revealed that the "cell adhesion and extracellular matrix remodeling" process and the "HIF1 pathway", a pathway related to oxidative stress responses, were significantly modulated upon morphine exposure in HBMECs and EVs. Altogether, the combination of proteomics and bioinformatics findings highlighted shared pathways between HBMECs exposed to morphine and their released EVs. These results put forward molecular signatures of morphine-induced toxicity in HBMECs that were also carried by EVs. Therefore, EVs could potentially be regarded as a useful tool to investigate brain endothelial cells dysfunction, and to a different extent, the BBB dysfunction in patient circulation using these "signature pathways".
Collapse
Affiliation(s)
- Tatjana Vujić
- Department of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | | | - Izadora Liranço Furlani
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Department of Chemistry, Federal University of São Carlos, São Carlos 13565-904, Brazil
| | - Isabel Meister
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Swiss Centre for Applied Human Toxicology, 4055 Basel, Switzerland
| | - Víctor González-Ruiz
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Swiss Centre for Applied Human Toxicology, 4055 Basel, Switzerland
| | - Serge Rudaz
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Swiss Centre for Applied Human Toxicology, 4055 Basel, Switzerland
| | - Jean-Charles Sanchez
- Department of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Correspondence: ; Tel.: +41-22-379-54-86
| |
Collapse
|
174
|
Zhang W, Chen X, Du Z, Mao X, Gao R, Chen Z, Wang H, Zhang G, Zhang N, Li H, Song Y, Chang L, Wu Y. Knockdown of astrocytic Grin2a exacerbated sleep deprivation-induced cognitive impairments and elevation of amyloid-beta. Sleep Med 2022; 100:280-290. [PMID: 36148760 DOI: 10.1016/j.sleep.2022.08.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/19/2022] [Accepted: 08/25/2022] [Indexed: 01/12/2023]
Abstract
Sleep disorders are associated with cognitive impairments, greater amyloid-β (Aβ) burden and increased risk of developing Alzheimer's disease, while the underlying mechanism is unclear. N-methyl-d-aspartate receptors (NMDARs), as vital modulators of cognition, are sensitive to sleep disturbance. Sleep deprivation (SD) could induce the alterations of neuronal NMDAR subunits expression, however the alterations of astrocytic NMDARs in SD have not been reported. Our previous study has demonstrated knockdown of astrocytic Grin2a (gene encoding NMDAR subunit GluN2A) could aggravate Aβ-induced cognitive impairments, but what role astrocytic GluN2A may play in SD is unknown. Here we focused on the changes and roles of hippocampal astrocytic GluN2A in SD. Our results showed SD increased the expression of astrocytic GluN2A. Specific knockdown of hippocampal astrocytic Grin2a aggravated SD-induced cognitive decline, elevated Aβ, and attenuated the SD-induced increase in autophagy flux. Our finding, for the first time, revealed a novel neuroprotective role for astrocytic GluN2A in SD, which may be helpful for developing new preventive and therapeutic targets to sleep disorders.
Collapse
Affiliation(s)
- Wanning Zhang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xinyue Chen
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Zunshu Du
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xin Mao
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ruiqi Gao
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ziyan Chen
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Hongqi Wang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Guitao Zhang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ning Zhang
- Department of Neuropsychiatry and Behavioral Neurology and Clinical Psychology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hui Li
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yizhi Song
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Lirong Chang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| | - Yan Wu
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
175
|
Rodríguez-Giraldo M, González-Reyes RE, Ramírez-Guerrero S, Bonilla-Trilleras CE, Guardo-Maya S, Nava-Mesa MO. Astrocytes as a Therapeutic Target in Alzheimer's Disease-Comprehensive Review and Recent Developments. Int J Mol Sci 2022; 23:13630. [PMID: 36362415 PMCID: PMC9654484 DOI: 10.3390/ijms232113630] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 09/20/2023] Open
Abstract
Alzheimer's disease (AD) is a frequent and disabling neurodegenerative disorder, in which astrocytes participate in several pathophysiological processes including neuroinflammation, excitotoxicity, oxidative stress and lipid metabolism (along with a critical role in apolipoprotein E function). Current evidence shows that astrocytes have both neuroprotective and neurotoxic effects depending on the disease stage and microenvironmental factors. Furthermore, astrocytes appear to be affected by the presence of amyloid-beta (Aβ), with alterations in calcium levels, gliotransmission and proinflammatory activity via RAGE-NF-κB pathway. In addition, astrocytes play an important role in the metabolism of tau and clearance of Aβ through the glymphatic system. In this review, we will discuss novel pharmacological and non-pharmacological treatments focused on astrocytes as therapeutic targets for AD. These interventions include effects on anti-inflammatory/antioxidant systems, glutamate activity, lipid metabolism, neurovascular coupling and glymphatic system, calcium dysregulation, and in the release of peptides which affects glial and neuronal function. According to the AD stage, these therapies may be of benefit in either preventing or delaying the progression of the disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Mauricio O. Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia
| |
Collapse
|
176
|
Wang L, Shi Y, Jiang J, Li C, Zhang H, Zhang X, Jiang T, Wang L, Wang Y, Feng L. Micro-Nanocarriers Based Drug Delivery Technology for Blood-Brain Barrier Crossing and Brain Tumor Targeting Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203678. [PMID: 36103614 DOI: 10.1002/smll.202203678] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/04/2022] [Indexed: 06/15/2023]
Abstract
The greatest obstacle to using drugs to treat brain tumors is the blood-brain barrier (BBB), making it difficult for conventional drug molecules to enter the brain. Therefore, how to safely and effectively penetrate the BBB to achieve targeted drug delivery to brain tumors has been a challenging research problem. With the intensive research in micro- and nanotechnology in recent years, nano drug-targeted delivery technologies have shown great potential to overcome this challenge, such as inorganic nanocarriers, organic polymer-carriers, liposomes, and biobased carriers, which can be designed in different sizes, shapes, and surface functional groups to enhance their ability to penetrate the BBB and targeted drug delivery for brain tumors. In this review, the composition and overcoming patterns of the BBB are detailed, and then the hot research topics of drug delivery carriers for brain tumors in recent years are summarized, and their mechanisms of action on the BBB and the factors affecting drug delivery are described in detail, and the effectiveness of targeted therapy for brain tumors is evaluated. Finally, the challenges and dilemmas in developing brain tumor drug delivery systems are discussed, which will be promising in the future for targeted drug delivery to brain tumors based on micro-nanocarriers technology.
Collapse
Affiliation(s)
- Luyao Wang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Youyuan Shi
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Jingzhen Jiang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Chan Li
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Hengrui Zhang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Xinhui Zhang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Tao Jiang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yinyan Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Lin Feng
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| |
Collapse
|
177
|
Zheng X, Zhao J, Wang S, Hu L. Research Progress of Antioxidant Nanomaterials for Acute Pancreatitis. Molecules 2022; 27:7238. [PMID: 36364064 PMCID: PMC9658789 DOI: 10.3390/molecules27217238] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/16/2022] [Accepted: 10/21/2022] [Indexed: 08/30/2023] Open
Abstract
Acute pancreatitis (AP) is a complex inflammatory disease caused by multiple etiologies, the pathogenesis of which has not been fully elucidated. Oxidative stress is important for the regulation of inflammation-related signaling pathways, the recruitment of inflammatory cells, the release of inflammatory factors, and other processes, and plays a key role in the occurrence and development of AP. In recent years, antioxidant therapy that suppresses oxidative stress by scavenging reactive oxygen species has become a research highlight of AP. However, traditional antioxidant drugs have problems such as poor drug stability and low delivery efficiency, which limit their clinical translation and applications. Nanomaterials bring a brand-new opportunity for the antioxidant treatment of AP. This review focuses on the multiple advantages of nanomaterials, including small size, good stability, high permeability, and long retention effect, which can be used not only as effective carriers of traditional antioxidant drugs but also directly as antioxidants. In this review, after first discussing the association between oxidative stress and AP, we focused on summarizing the literature related to antioxidant nanomaterials for the treatment of AP and highlighting the effects of these nanomaterials on the indicators related to oxidative stress in pathological states, aiming to provide references for follow-up research and promote clinical application.
Collapse
Affiliation(s)
- Xiaoyi Zheng
- Ningxia Medical University, Postgraduate Training Base in Shanghai Gongli Hospital, Pudong New Area, No. 219 Miao Pu Road, Shanghai 200135, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, China
| | - Jiulong Zhao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, China
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, China
| | - Lianghao Hu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
178
|
Zhan Y, Zhang Z, Liu Y, Fang Y, Xie Y, Zheng Y, Li G, Liang L, Ding Y. NUPR1 contributes to radiation resistance by maintaining ROS homeostasis via AhR/CYP signal axis in hepatocellular carcinoma. BMC Med 2022; 20:365. [PMID: 36258210 PMCID: PMC9580158 DOI: 10.1186/s12916-022-02554-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Radiotherapy (RT) is one of the major therapeutic approaches to hepatocellular carcinoma (HCC). Ionizing radiation (IR) inducing the generation of reactive oxygen species (ROS) leads to a promising antitumor effect. However, the dysregulation of the redox system often causes radioresistance and impairs the efficacy of RT. Increasing evidence indicates that nuclear protein 1 (NUPR1) plays a critical role in redox reactions. In this study, we aim to explore the role of NUPR1 in maintaining ROS homeostasis and radioresistance in HCC. METHODS The radioresistant role of NUPR1 was determined by colony formation assay, comet assay in vitro, and xenograft tumor models in vivo. Probes for ROS, apoptosis assay, and lipid peroxidation assay were used to investigate the functional effect of NUPR1 on ROS homeostasis and oxidative stress. RNA sequencing and co-immunoprecipitation assay were performed to clarify the mechanism of NUPR1 inhibiting the AhR/CYP signal axis. Finally, we analyzed clinical specimens to assess the predictive value of NUPR1 and AhR in the radiotherapeutic efficacy of HCC. RESULTS We demonstrated that NUPR1 was upregulated in HCC tissues and verified that NUPR1 increased the radioresistance of HCC in vitro and in vivo. NUPR1 alleviated the generation of ROS and suppressed oxidative stress, including apoptosis and lipid peroxidation by downregulating cytochrome P450 (CYP) upon IR. ROS scavenger N-acetyl-L-cysteine (NAC) and CYP inhibitor alizarin restored the viability of NUPR1-knockdown cells during IR. Mechanistically, the interaction between NUPR1 and aryl hydrocarbon receptor (AhR) promoted the degradation and decreased nuclear translation of AhR via the autophagy-lysosome pathway, followed by being incapable of CYP's transcription. Furthermore, genetically and pharmacologically activating AhR abrogated the radioresistant role of NUPR1. Clinical data suggested that NUPR1 and AhR could serve as novel biomarkers for predicting the radiation response of HCC. CONCLUSIONS Our findings revealed the role of NUPR1 in regulating ROS homeostasis and oxidative stress via the AhR/CYP signal axis upon IR. Strategies targeting the NUPR1/AhR/CYP pathway may have important clinical applications for improving the radiotherapeutic efficacy of HCC.
Collapse
Affiliation(s)
- Yizhi Zhan
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhanqiao Zhang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yuechen Liu
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yuan Fang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yuwen Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yilin Zheng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guoxin Li
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China.
| | - Yi Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
179
|
Loan JJM, Al-Shahi Salman R, McColl BW, Hardingham GE. Activation of Nrf2 to Optimise Immune Responses to Intracerebral Haemorrhage. Biomolecules 2022; 12:1438. [PMID: 36291647 PMCID: PMC9599325 DOI: 10.3390/biom12101438] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
Haemorrhage into the brain parenchyma can be devastating. This manifests as spontaneous intracerebral haemorrhage (ICH) after head trauma, and in the context of vascular dementia. Randomised controlled trials have not reliably shown that haemostatic treatments aimed at limiting ICH haematoma expansion and surgical approaches to reducing haematoma volume are effective. Consequently, treatments to modulate the pathophysiological responses to ICH, which may cause secondary brain injury, are appealing. Following ICH, microglia and monocyte derived cells are recruited to the peri-haematomal environment where they phagocytose haematoma breakdown products and secrete inflammatory cytokines, which may trigger both protective and harmful responses. The transcription factor Nrf2, is activated by oxidative stress, is highly expressed by central nervous system microglia and macroglia. When active, Nrf2 induces a transcriptional programme characterised by increased expression of antioxidant, haem and heavy metal detoxification and proteostasis genes, as well as suppression of proinflammatory factors. Therefore, Nrf2 activation may facilitate adaptive-protective immune cell responses to ICH by boosting resistance to oxidative stress and heavy metal toxicity, whilst limiting harmful inflammatory signalling, which can contribute to further blood brain barrier dysfunction and cerebral oedema. In this review, we consider the responses of immune cells to ICH and how these might be modulated by Nrf2 activation. Finally, we propose potential therapeutic strategies to harness Nrf2 to improve the outcomes of patients with ICH.
Collapse
Affiliation(s)
- James J. M. Loan
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
- UK Dementia Research Institute at Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | | | - Barry W. McColl
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
- UK Dementia Research Institute at Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Giles E. Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
- UK Dementia Research Institute at Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, UK
| |
Collapse
|
180
|
Sabry MM, Ahmed MM, Maksoud OMA, Rashed L, Morcos MA, El-Maaty AA, Maher Galal A, Sharawy N. Carnitine, apelin and resveratrol regulate mitochondrial quality control (QC) related proteins and ameliorate acute kidney injury: role of hydrogen peroxide. Arch Physiol Biochem 2022; 128:1391-1400. [PMID: 32538173 DOI: 10.1080/13813455.2020.1773504] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondrial impairment is recognised as a prominent feature in kidney diseases. Therefore, we investigated whether the effects of resveratrol, L-carnitine, and apelin in the acute kidney injury model were associated with modulation of mitochondrial quality control (QC) related proteins, intra-renal renin-angiotensin (RAS) activity, adenosine triphosphate (ATP) and Na+-K+ ATPase gene expression. Rats were randomly assigned to 7 groups: Distilled water injected control group, DMSO injected control group, distilled water injected lipopolysaccharide (LPS) group, DMSO injected LPS group, resveratrol injected LPS group, L-carnitine injected LPS group and apelin 13 injected LPS group. We observed that resveratrol, L-carnitine, and apelin treatments altered mitochondrial (QC) related protein levels (Pink1, Parkin, BNIP-3, Drp1, and PGC1α), decreased intra-renal RAS parameters, increased ATP level and upregulated Na+-K+ ATPase gene expression in renal tissue. Our results provide new insight into the role of mitochondrial quality control and how different antioxidants exert beneficial effects on acute kidney injury.
Collapse
Affiliation(s)
- Maha Mohamed Sabry
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mona Mohamed Ahmed
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Laila Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mary Attia Morcos
- Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Amal Abo El-Maaty
- Department of Animal Reproduction and Artificial Insemination, Veterinary Division, National Research Centre, Cairo, Egypt
| | - Amr Maher Galal
- Department of Pharmacology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nivin Sharawy
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of physiology, Cairo University Hospitals, Cairo, Egypt
| |
Collapse
|
181
|
Liu D, Zhao L, Jiang Y, Li L, Guo M, Mu Y, Zhu H. Integrated analysis of plasma and urine reveals unique metabolomic profiles in idiopathic inflammatory myopathies subtypes. J Cachexia Sarcopenia Muscle 2022; 13:2456-2472. [PMID: 35860906 PMCID: PMC9530549 DOI: 10.1002/jcsm.13045] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES Idiopathic inflammatory myopathies (IIM) are a class of autoimmune diseases with high heterogeneity that can be divided into different subtypes based on clinical manifestations and myositis-specific autoantibodies (MSAs). However, even in each IIM subtype, the clinical symptoms and prognoses of patients are very different. Thus, the identification of more potential biomarkers associated with IIM classification, clinical symptoms, and prognosis is urgently needed. METHODS Plasma and urine samples from 79 newly diagnosed IIM patients (mean disease duration 4 months) and 52 normal control (NC) samples were analysed by high-performance liquid chromatography of quadrupole time-of-flight mass spectrometry (HPLC-Q-TOF-MS)/MS-based untargeted metabolomics. Orthogonal partial least-squares discriminate analysis (OPLS-DA) were performed to measure the significance of metabolites. Pathway enrichment analysis was conducted based on the KEGG human metabolic pathways. Ten machine learning (ML) algorithms [linear support vector machine (SVM), radial basis function SVM, random forest, nearest neighbour, Gaussian processes, decision trees, neural networks, adaptive boosting (AdaBoost), Gaussian naive Bayes and quadratic discriminant analysis] were used to classify each IIM subtype and select the most important metabolites as potential biomarkers. RESULTS OPLS-DA showed a clear separation between NC and IIM subtypes in plasma and urine metabolic profiles. KEGG pathway enrichment analysis revealed multiple unique and shared disturbed metabolic pathways in IIM main [dermatomyositis (DM), anti-synthetase syndrome (ASS), and immune-mediated necrotizing myopathy (IMNM)] and MSA-defined subtypes (anti-Mi2+, anti-MDA5+, anti-TIF1γ+, anti-Jo1+, anti-PL7+, anti-PL12+, anti-EJ+, and anti-SRP+), such that fatty acid biosynthesis was significantly altered in both plasma and urine in all main IIM subtypes (enrichment ratio > 1). Random forest and AdaBoost performed best in classifying each IIM subtype among the 10 ML models. Using the feature selection methods in ML models, we identified 9 plasma and 10 urine metabolites that contributed most to separate IIM main subtypes and MSA-defined subtypes, such as plasma creatine (fold change = 3.344, P = 0.024) in IMNM subtype and urine tiglylcarnitine (fold change = 0.351, P = 0.037) in anti-EJ+ ASS subtype. Sixteen common metabolites were found in both the plasma and urine samples of IIM subtypes. Among them, some were correlated with clinical features, such as plasma hypogeic acid (r = -0.416, P = 0.005) and urine malonyl carnitine (r = -0.374, P = 0.042), which were negatively correlated with the prevalence of interstitial lung disease. CONCLUSIONS In both plasma and urine samples, IIM main and MSA-defined subtypes have specific metabolic signatures and pathways. This study provides useful clues for understanding the molecular mechanisms, searching potential diagnosis biomarkers and therapeutic targets for IIM.
Collapse
Affiliation(s)
- Di Liu
- Department of Rheumatology and Immunology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Department of Rheumatology and Clinical Immunology, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Lijuan Zhao
- Department of Rheumatology and Immunology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yu Jiang
- Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Institute of Emergency MedicineHunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal UniversityChangshaHunanChina
| | - Liya Li
- Department of Rheumatology and Immunology, The Third Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Muyao Guo
- Department of Rheumatology and Immunology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yibing Mu
- Department of NutritionHunan Provincial Maternal and Child Health Care HospitalChangshaHunanChina
| | - Honglin Zhu
- Department of Rheumatology and Immunology, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
182
|
Liu YJ, Li YL, Fang ZH, Liao HL, Zhang YY, Lin J, Liu F, Shen JF. NMDARs mediate peripheral and central sensitization contributing to chronic orofacial pain. Front Cell Neurosci 2022; 16:999509. [PMID: 36238833 PMCID: PMC9553029 DOI: 10.3389/fncel.2022.999509] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022] Open
Abstract
Peripheral and central sensitizations of the trigeminal nervous system are the main mechanisms to promote the development and maintenance of chronic orofacial pain characterized by allodynia, hyperalgesia, and ectopic pain after trigeminal nerve injury or inflammation. Although the pathomechanisms of chronic orofacial pain are complex and not well known, sufficient clinical and preclinical evidence supports the contribution of the N-methyl-D-aspartate receptors (NMDARs, a subclass of ionotropic glutamate receptors) to the trigeminal nociceptive signal processing pathway under various pathological conditions. NMDARs not only have been implicated as a potential mediator of pain-related neuroplasticity in the peripheral nervous system (PNS) but also mediate excitatory synaptic transmission and synaptic plasticity in the central nervous system (CNS). In this review, we focus on the pivotal roles and mechanisms of NMDARs in the trigeminal nervous system under orofacial neuropathic and inflammatory pain. In particular, we summarize the types, components, and distribution of NMDARs in the trigeminal nervous system. Besides, we discuss the regulatory roles of neuron-nonneuronal cell/neuron-neuron communication mediated by NMDARs in the peripheral mechanisms of chronic orofacial pain following neuropathic injury and inflammation. Furthermore, we review the functional roles and mechanisms of NMDARs in the ascending and descending circuits under orofacial neuropathic and inflammatory pain conditions, which contribute to the central sensitization. These findings are not only relevant to understanding the underlying mechanisms, but also shed new light on the targeted therapy of chronic orofacial pain.
Collapse
Affiliation(s)
- Ya-Jing Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue-Ling Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhong-Han Fang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hong-Lin Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan-Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiu Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie-Fei Shen Fei Liu
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie-Fei Shen Fei Liu
| |
Collapse
|
183
|
Haddow K, Kind PC, Hardingham GE. NMDA Receptor C-Terminal Domain Signalling in Development, Maturity, and Disease. Int J Mol Sci 2022; 23:ijms231911392. [PMID: 36232696 PMCID: PMC9570437 DOI: 10.3390/ijms231911392] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
The NMDA receptor is a Ca2+-permeant glutamate receptor which plays key roles in health and disease. Canonical NMDARs contain two GluN2 subunits, of which 2A and 2B are predominant in the forebrain. Moreover, the relative contribution of 2A vs. 2B is controlled both developmentally and in an activity-dependent manner. The GluN2 subtype influences the biophysical properties of the receptor through difference in their N-terminal extracellular domain and transmembrane regions, but they also have large cytoplasmic Carboxyl (C)-terminal domains (CTDs) which have diverged substantially during evolution. While the CTD identity does not influence NMDAR subunit specific channel properties, it determines the nature of CTD-associated signalling molecules and has been implicated in mediating the control of subunit composition (2A vs. 2B) at the synapse. Historically, much of the research into the differential function of GluN2 CTDs has been conducted in vitro by over-expressing mutant subunits, but more recently, the generation of knock-in (KI) mouse models have allowed CTD function to be probed in vivo and in ex vivo systems without heterologous expression of GluN2 mutants. In some instances, findings involving KI mice have been in disagreement with models that were proposed based on earlier approaches. This review will examine the current research with the aim of addressing these controversies and how methodology may contribute to differences between studies. We will also discuss the outstanding questions regarding the role of GluN2 CTD sequences in regulating NMDAR subunit composition, as well as their relevance to neurodegenerative disease and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Kirsty Haddow
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Peter C. Kind
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Giles E. Hardingham
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
- Correspondence:
| |
Collapse
|
184
|
Yuan H, Liu X, Wang Z, Ren Y, Li Y, Gao C, Jiao T, Cai Y, Yang Y, Zhao S. Alternative splicing signature of alveolar type II epithelial cells of Tibetan pigs under hypoxia-induced. Front Vet Sci 2022; 9:984703. [PMID: 36187824 PMCID: PMC9523697 DOI: 10.3389/fvets.2022.984703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Alternative splicing (AS) allows the generation of multiple transcript variants from a single gene and affects biological processes by generating protein diversity in organisms. In total, 41,642 AS events corresponding to 9,924 genes were identified, and SE is the most abundant alternatively spliced type. The analysis of functional categories demonstrates that alternatively spliced differentially expressed genes (DEGs) were enriched in the MAPK signaling pathway and hypoxia-inducible factor 1 (HIF-1) signaling pathway. Proteoglycans in cancer between the normoxic (21% O2, TN and LN) and hypoxic (2% O2, TL and LL) groups, such as SLC2A1, HK1, HK2, ENO3, and PFKFB3, have the potential to rapidly proliferate alveolar type II epithelial (ATII) cells by increasing the intracellular levels of glucose and quickly divert to anabolic pathways by glycolysis intermediates under hypoxia. ACADL, EHHADH, and CPT1A undergo one or two AS types with different frequencies in ATII cells between TN and TL groups (excluding alternatively spliced DEGs shared between normoxic and hypoxic groups), and a constant supply of lipids might be obtained either from the circulation or de novo synthesis for better growth of ATII cells under hypoxia condition. MCM7 and MCM3 undergo different AS types between LN and LL groups (excluding alternatively spliced DEGs shared between normoxic and hypoxic groups), which may bind to the amino-terminal PER-SIM-ARNT domain and the carboxyl terminus of HIF-1α to maintain their stability. Overall, AS and expression levels of candidate mRNAs between Tibetan pigs and Landrace pigs revealed by RNA-seq suggest their potential involvement in the ATII cells grown under hypoxia conditions.
Collapse
Affiliation(s)
- Haonan Yuan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xuanbo Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Zhengwen Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yue Ren
- Academy of Agriculture and Animal Husbandry Sciences, Institute of Animal Husbandry and Veterinary Medicine, Lhasa, China
| | - Yongqing Li
- Xinjiang Academy of Animal Sciences, Xinjiang, China
| | - Caixia Gao
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ting Jiao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- College of Grassland Science, Gansu Agricultural University, Lanzhou, China
| | - Yuan Cai
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yanan Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- *Correspondence: Yanan Yang
| | - Shengguo Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- Shengguo Zhao
| |
Collapse
|
185
|
Insight into the Effects of High-Altitude Hypoxic Exposure on Learning and Memory. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4163188. [PMID: 36160703 PMCID: PMC9492407 DOI: 10.1155/2022/4163188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/22/2022] [Indexed: 02/05/2023]
Abstract
The earth land area is heterogeneous in terms of elevation; about 45% of its land area belongs to higher elevation with altitude above 500 meters compared to sea level. In most cases, oxygen concentration decreases as altitude increases. Thus, high-altitude hypoxic stress is commonly faced by residents in areas with an average elevation exceeding 2500 meters and those who have just entered the plateau. High-altitude hypoxia significantly affects advanced neurobehaviors including learning and memory (L&M). Hippocampus, the integration center of L&M, could be the most crucial target affected by high-altitude hypoxia exposure. Based on these points, this review thoroughly discussed the relationship between high-altitude hypoxia and L&M impairment, in terms of hippocampal neuron apoptosis and dysfunction, neuronal oxidative stress disorder, neurotransmitters and related receptors, and nerve cell energy metabolism disorder, which is of great significance to find potential targets for medical intervention. Studies illustrate that the mechanism of L&M damaged by high-altitude hypoxia should be further investigated based on the entire review of issues related to this topic.
Collapse
|
186
|
Seillier C, Lesept F, Toutirais O, Potzeha F, Blanc M, Vivien D. Targeting NMDA Receptors at the Neurovascular Unit: Past and Future Treatments for Central Nervous System Diseases. Int J Mol Sci 2022; 23:ijms231810336. [PMID: 36142247 PMCID: PMC9499580 DOI: 10.3390/ijms231810336] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
The excitatory neurotransmission of the central nervous system (CNS) mainly involves glutamate and its receptors, especially N-methyl-D-Aspartate receptors (NMDARs). These receptors have been extensively described on neurons and, more recently, also on other cell types. Nowadays, the study of their differential expression and function is taking a growing place in preclinical and clinical research. The diversity of NMDAR subtypes and their signaling pathways give rise to pleiotropic functions such as brain development, neuronal plasticity, maturation along with excitotoxicity, blood-brain barrier integrity, and inflammation. NMDARs have thus emerged as key targets for the treatment of neurological disorders. By their large extracellular regions and complex intracellular structures, NMDARs are modulated by a variety of endogenous and pharmacological compounds. Here, we will present an overview of NMDAR functions on neurons and other important cell types involved in the pathophysiology of neurodegenerative, neurovascular, mental, autoimmune, and neurodevelopmental diseases. We will then discuss past and future development of NMDAR targeting drugs, including innovative and promising new approaches.
Collapse
Affiliation(s)
- Célia Seillier
- Normandie University, UNICAEN, INSERM, GIP Cyceron, Institute Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), 14000 Caen, France
| | - Flavie Lesept
- Lys Therapeutics, Cyceron, Boulevard Henri Becquerel, 14000 Caen, France
| | - Olivier Toutirais
- Normandie University, UNICAEN, INSERM, GIP Cyceron, Institute Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), 14000 Caen, France
- Department of Immunology and Histocompatibility (HLA), Caen University Hospital, CHU, 14000 Caen, France
| | - Fanny Potzeha
- Lys Therapeutics, Cyceron, Boulevard Henri Becquerel, 14000 Caen, France
| | - Manuel Blanc
- Lys Therapeutics, Cyceron, Boulevard Henri Becquerel, 14000 Caen, France
| | - Denis Vivien
- Normandie University, UNICAEN, INSERM, GIP Cyceron, Institute Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), 14000 Caen, France
- Department of Clinical Research, Caen University Hospital, CHU, 14000 Caen, France
- Correspondence:
| |
Collapse
|
187
|
Erdogan CS, Al Hassadi Y, Aru B, Yilmaz B, Gemici B. Combinatorial effects of melatonin and paclitaxel differ depending on the treatment scheme in colorectal cancer in vitro. Life Sci 2022; 308:120927. [PMID: 36063977 DOI: 10.1016/j.lfs.2022.120927] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/22/2022] [Accepted: 08/30/2022] [Indexed: 10/31/2022]
Abstract
AIMS Colorectal carcinoma (CRC) is the third most prevalent cancer with high mortality. Besides regulating the circadian rhythm, melatonin (MTN) exerts anticancer activities. Paclitaxel (PTX) is successful against different malignancies, however, acquired resistance and variability in patient response restrict its use. mTOR and MAPK pathways are often deregulated in human cancers. We aimed to investigate whether MTN enhances or sensitizes the chemotherapeutic activity of PTX and if so, determine the underlying possible mechanisms in CRC in vitro. MAIN METHODS Antiproliferative and cytotoxic activities of PTX and MTN were assessed alone and in combination, as well as with different treatment regimens (renewal or replacement of the treatment after 24 h), up to 48 h. Apoptosis, viability and autophagy were assessed by flow cytometry. mTOR and MAPK pathway activities were investigated by immunoblotting. KEY FINDINGS Both drugs reduced cell viability in a dose-dependent manner at 24 and 48 h. Only the highest dose of MTN (500 μM) potentiated the cytotoxicity of PTX (50 nM). Replacement of PTX after 24 h with MTN was superior in reducing cell viability than vice versa via apoptosis induction. Renewal of MTN treatment every 24 h reduced autophagy compared to the control group, while other treatments did not alter the autophagic activity. A 24 h MTN treatment followed by 24 h PTX treatment increased S6 phosphorylation in a mTOR-independent manner and increased Erk1/2 phosphorylation. SIGNIFICANCE The present study suggests that sequential treatment with MTN and PTX distinctly affect apoptosis and cytotoxicity via regulating mTOR and MAPK pathways differentially in CRC.
Collapse
Affiliation(s)
- Cihan Suleyman Erdogan
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Yasmine Al Hassadi
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Basak Aru
- Yeditepe University, Faculty of Medicine, Department of Immunology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Bayram Yilmaz
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Burcu Gemici
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey.
| |
Collapse
|
188
|
Lee S, Byun JK, Kim NY, Jin J, Woo H, Choi YK, Park KG. Melatonin inhibits glycolysis in hepatocellular carcinoma cells by downregulating mitochondrial respiration and mTORC1 activity. BMB Rep 2022; 55:459-464. [PMID: 35651333 PMCID: PMC9537022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/19/2022] [Accepted: 03/23/2022] [Indexed: 03/08/2024] Open
Abstract
Various mechanisms have been suggested to explain the chemopreventive and tumor-inhibitory effects of melatonin. Despite the growing evidence supporting melatonin-induced mitochondrial dysfunction, it remains largely unknown how this phenomenon modulates metabolic reprogramming in cancer cells. The aim of our study was to identify the mechanism underlying the anti-proliferative and apoptotic effects of melatonin, which is known to inhibit glycolysis. We analyzed the time-dependent effects of melatonin on mitochondrial respiration and glycolysis in liver cancer cells. The results showed that from a cell bioenergetic point of view, melatonin caused an acute reduction in mitochondrial respiration, however, increased reactive oxygen species production, thereby inhibiting mTORC1 activity from an early stage post-treatment without affecting glycolysis. Nevertheless, administration of melatonin for a longer time reduced expression of c-Myc protein, thereby suppressing glycolysis via downregulation of HK2 and LDHA. The data presented herein suggest that melatonin suppresses mitochondrial respiration and glycolysis simultaneously in HCC cells, leading to anti-cancer effects. Thus, melatonin can be used as an adjuvant agent for therapy of liver cancer. [BMB Reports 2022; 55(9): 459-464].
Collapse
Affiliation(s)
- Seunghyeong Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41566, Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Jun-Kyu Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
| | - Na-Young Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Jonghwa Jin
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Hyein Woo
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Yeon-Kyung Choi
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
| | - Keun-Gyu Park
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41566, Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
189
|
Li Y, Zhu Y, Li S, Dong Y, Wan C, Yu X, Xin G, Wei Z, Li F, Wang Y, Zhang K, Chen Q, Zhang C, Wen E, Niu H, Huang W. Deoxyarbutin attenuates severe acute pancreatitis via the HtrA2/PGC-1α pathway. Free Radic Res 2022; 56:651-665. [PMID: 36592372 DOI: 10.1080/10715762.2022.2163244] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Severe acute pancreatitis (SAP) is an inflammatory disorder of the exocrine pancreas associated with high morbidity and mortality. SAP has been proven to trigger mitochondria dysfunction in the pancreas. We found that Deoxyarbutin (dA) recovered impaired mitochondrial function. High-temperature requirement protein A2 (HtrA2), a mitochondrial serine protease upstream of PGC-1α, is charge of quality control in mitochondrial homeostasis. The molecular docking study indicated that there was a potential interaction between dA and HtrA2. However, whether the protective effect of dA against SAP is regulated by HtrA2/PGC-1α remains unknown. Our study in vitro showed that dA significantly reduced the necrosis of primary acinar cells and reactive oxygen species (ROS) accumulation, recovered mitochondrial membrane potential (ΔΨm) and ATP exhaustion, while UCF-101 (HtrA2 inhibitor), and SR-18292 (PGC-1α inhibitor) eliminated the protective effect of dA. Moreover, HtrA2 siRNA transfection efficiently blocked the protective of dA on HtrA2/PGC-1α pathway in 266-6 acinar cells. Meanwhile, dA also decreased LC3II/I ration, as well as p62, and increased Parkin expression, while UCF-101 and Bafilomycin A1 (autophagy inhibitor) reversed the protective effect of dA. Our study in vivo confirmed that dA effectively alleviated severity of SAP by reducing pancreatic edema, plasma amylase, and lipase levels and improved the HtrA2/PGC-1α pathway. Therefore, this is the first study to identify that dA inhibits pancreatic injury caused by oxidative stress, mitochondrial dysfunction, and impaired autophagy in a HtrA2/PGC-1α dependent manner.
Collapse
Affiliation(s)
- Yangying Li
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yuda Zhu
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Shiyi Li
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yuman Dong
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chengyu Wan
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiuxian Yu
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Guang Xin
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zeliang Wei
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Fan Li
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yilan Wang
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Kun Zhang
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qingqiu Chen
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Cuicui Zhang
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - E Wen
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hai Niu
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Huang
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
190
|
Chen S, Li Y, Fu S, Li Y, Wang C, Sun P, Li H, Tian J, Du GQ. Melatonin alleviates arginine vasopressin-induced cardiomyocyte apoptosis via increasing Mst1-Nrf2 pathway activity to reduce oxidative stress. Biochem Pharmacol 2022; 206:115265. [DOI: 10.1016/j.bcp.2022.115265] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 09/08/2022] [Accepted: 09/23/2022] [Indexed: 11/27/2022]
|
191
|
Petsouki E, Cabrera SNS, Heiss EH. AMPK and NRF2: Interactive players in the same team for cellular homeostasis? Free Radic Biol Med 2022; 190:75-93. [PMID: 35918013 DOI: 10.1016/j.freeradbiomed.2022.07.014] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/11/2022] [Accepted: 07/19/2022] [Indexed: 11/27/2022]
Abstract
NRF2 (Nuclear factor E2 p45-related factor 2) is a stress responsive transcription factor lending cells resilience against oxidative, xenobiotic, and also nutrient or proteotoxic insults. AMPK (AMP-activated kinase), considered as prime regulator of cellular energy homeostasis, not only tunes metabolism to provide the cell at any time with sufficient ATP or building blocks, but also controls redox balance and inflammation. Due to observed overlapping cellular responses upon AMPK or NRF2 activation and common stressors impinging on both AMPK and NRF2 signaling, it is plausible to assume that AMPK and NRF2 signaling may interdepend and cooperate to readjust cellular homeostasis. After a short introduction of the two players this narrative review paints the current picture on how AMPK and NRF2 signaling might interact on the molecular level, and highlights their possible crosstalk in selected examples of pathophysiology or bioactivity of drugs and phytochemicals.
Collapse
Affiliation(s)
- Eleni Petsouki
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Faculty of Life Sciences, Althanstrasse 14, 1090 Vienna, Austria
| | - Shara Natalia Sosa Cabrera
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Faculty of Life Sciences, Althanstrasse 14, 1090 Vienna, Austria; Vienna Doctoral School of Pharmaceutical, Nutritional and Sport Sciences (VDS PhaNuSpo), University of Vienna, Austria
| | - Elke H Heiss
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Faculty of Life Sciences, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
192
|
Engineering microbial cell viability for enhancing chemical production by second codon engineering. Metab Eng 2022; 73:235-246. [PMID: 35987432 DOI: 10.1016/j.ymben.2022.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 06/29/2022] [Accepted: 08/12/2022] [Indexed: 11/22/2022]
Abstract
Microbial cell factories offer a promising strategy for the sustainable production of industrial chemicals from renewable biomass feedstock. However, their performance is often limited by poor microbial cell viability (MCV). Here, MCV was engineered to enhance chemical production by optimizing the regulation of lifespan-specific genes to reduce the accumulation of reactive oxygen species (ROS). In Escherichia coli, MCV was improved by reducing ROS accumulation using second codon engineering to regulate hypoxia-inducible transcription factor (arcA), resulting in lysine production up to 213 g L-1 with its productivity 5.90 g L-1·h-1. In Saccharomyces cerevisiae, MCV was increased by decreasing ROS accumulation using second codon engineering to fine-tune ceramide synthase (lag1), leading to glucaric acid production up to 9.50 g L-1 with its productivity 0.057 g L-1·h-1. These results demonstrate that engineering MCV is a potential strategy to boost the performance of microbial cell factories in industrial processes.
Collapse
|
193
|
Sorafenib combined with STAT3 knockdown triggers ER stress-induced HCC apoptosis and cGAS-STING-mediated anti-tumor immunity. Cancer Lett 2022; 547:215880. [PMID: 35981569 DOI: 10.1016/j.canlet.2022.215880] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/01/2022] [Accepted: 08/11/2022] [Indexed: 01/07/2023]
Abstract
Sorafenib is the first-line treatment for advanced hepatocellular carcinoma (HCC). However, it is difficult to alleviate this disease process using single-agent chemotherapy. Using combination therapies for advanced HCC has become a major trend. Given that STAT3 overexpression is involved in chemotherapy resistance and the immune escape of HCC cells, it has become a potential therapeutic target for HCC in recent years. GEO database analysis showed that STAT3 levels in tumor tissues from non-responders were significantly higher than those in responders to sorafenib. Our studies demonstrated that STAT3 knockdown promoted sorafenib-induced ER stress-induced apoptosis. Importantly, the DNA released by dead HCC cells stimulated the cGAS-STING signaling pathway in CD103+ DCs and promoted type I interferon production, thus, enhancing the anti-tumor function of CD8+ T and NK cells. In conclusion, our results revealed that the combination strategy of sorafenib and STAT3 knockdown might be a potential treatment strategy for HCC, directly and efficiently disturbing the tumor features of HCC cells while improving the tumor microenvironment via the cGAS-STING-Type I IFNs axis of DCs, inducing anti-HCC immune responses.
Collapse
|
194
|
Elucidation of an mTORC2-PKC-NRF2 pathway that sustains the ATF4 stress response and identification of Sirt5 as a key ATF4 effector. Cell Death Dis 2022; 8:357. [PMID: 35963851 PMCID: PMC9376072 DOI: 10.1038/s41420-022-01156-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/08/2022]
Abstract
Proliferating cancer cells are dependent on glutamine metabolism for survival when challenged with oxidative stresses caused by reactive oxygen species, hypoxia, nutrient deprivation and matrix detachment. ATF4, a key stress responsive transcription factor, is essential for cancer cells to sustain glutamine metabolism when challenged with these various types of stress. While it is well documented how the ATF4 transcript is translated into protein as a stress response, an important question concerns how the ATF4 message levels are sustained to enable cancer cells to survive the challenges of nutrient deprivation and damaging reactive oxygen species. Here, we now identify the pathway in triple negative breast cancer cells that provides a sustained ATF4 response and enables their survival when encountering these challenges. This signaling pathway starts with mTORC2, which upon sensing cellular stresses arising from glutamine deprivation or an acute inhibition of glutamine metabolism, initiates a cascade of events that triggers an increase in ATF4 transcription. Surprisingly, this signaling pathway is not dependent on AKT activation, but rather requires the mTORC2 target, PKC, which activates the transcription factor Nrf2 that then induces ATF4 expression. Additionally, we identify a sirtuin family member, the NAD+-dependent de-succinylase Sirt5, as a key transcriptional target for ATF4 that promotes cancer cell survival during metabolic stress. Sirt5 plays fundamental roles in supporting cancer cell metabolism by regulating various enzymatic activities and by protecting an enzyme essential for glutaminolysis, glutaminase C (GAC), from degradation. We demonstrate that ectopic expression of Sirt5 compensates for knockdowns of ATF4 in cells exposed to glutamine deprivation-induced stress. These findings provide important new insights into the signaling cues that lead to sustained ATF4 expression as a general stress-induced regulator of glutamine metabolism, as well as highlight Sirt5 an essential effector of the ATF4 response to metabolic stress.
Collapse
|
195
|
Völkl M, Jérôme V, Weig A, Jasinski J, Meides N, Strohriegl P, Scheibel T, Freitag R. Pristine and artificially-aged polystyrene microplastic particles differ in regard to cellular response. JOURNAL OF HAZARDOUS MATERIALS 2022; 435:128955. [PMID: 35472543 DOI: 10.1016/j.jhazmat.2022.128955] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 06/14/2023]
Abstract
Microplastic particles (MP), arising from the gradual decomposition of plastics in the environment, have been identified as a global problem. Most investigations of MP cytotoxicity use pristine spherical particles available from commercial sources when evaluating their impact on mammalian cells, while only limited data is available for the more relevant "weathered microplastic". In this study, we exposed murine macrophages to polystyrene MP either after up to 130 days of accelerated ageing or in pristine condition. Weathered and pristine MP were physicochemically characterized, and their cytotoxicity was investigated using biological assays, transcriptome analysis, and metabolic pathways prediction. Whereas the response to pristine MP is mainly dominated by a TNF-α release, sharp-edged weathered MP induce broader adverse cellular reactions. This study stresses the importance of including more realistic test particles (e.g., weathered particles) in combination with a broad range of biological assays when evaluating the potential risk of microplastic exposure.
Collapse
Affiliation(s)
- Matthias Völkl
- Process Biotechnology, University of Bayreuth, 95447 Bayreuth, Germany
| | - Valérie Jérôme
- Process Biotechnology, University of Bayreuth, 95447 Bayreuth, Germany
| | - Alfons Weig
- Genomics and Bioinformatics, University of Bayreuth, 95447 Bayreuth, Germany
| | - Julia Jasinski
- Biomaterials, University of Bayreuth, 95447 Bayreuth, Germany
| | - Nora Meides
- Macromolecular Chemistry I, University of Bayreuth, 95447 Bayreuth, Germany
| | - Peter Strohriegl
- Macromolecular Chemistry I, University of Bayreuth, 95447 Bayreuth, Germany
| | - Thomas Scheibel
- Biomaterials, University of Bayreuth, 95447 Bayreuth, Germany
| | - Ruth Freitag
- Process Biotechnology, University of Bayreuth, 95447 Bayreuth, Germany.
| |
Collapse
|
196
|
de Souza Almeida RR, Bobermin LD, Parmeggiani B, Wartchow KM, Souza DO, Gonçalves CA, Wajner M, Leipnitz G, Quincozes-Santos A. Methylmalonic acid induces inflammatory response and redox homeostasis disruption in C6 astroglial cells: potential glioprotective roles of melatonin and resveratrol. Amino Acids 2022; 54:1505-1517. [PMID: 35927507 DOI: 10.1007/s00726-022-03191-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 07/22/2022] [Indexed: 11/24/2022]
Abstract
Methylmalonic acidemia is a neurometabolic disorder biochemically characterized by the accumulation of methylmalonic acid (MMA) in different tissues, including the central nervous system (CNS). In this sense, it has been shown that high levels of this organic acid have a key role in the progressive neurological deterioration in patients. Astroglial cells actively participate in a wide range of CNS functions, such as antioxidant defenses and inflammatory response. Considering the role of these cells to maintain brain homeostasis, in the present study, we investigated the effects of MMA on glial parameters, focusing on redox homeostasis and inflammatory process, as well as putative mediators of these events in C6 astroglial cells. MMA decreased cell viability, glutathione levels, and antioxidant enzyme activities, increased inflammatory response, and changed the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), nuclear factor kappa B (NFκB), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), inducible nitric oxide synthase (iNOS), cyclooxygenase 2 (COX-2), and adenosine receptors, suggesting that these transcriptional factors and proteins may underlie the glial responses induced by MMA. Moreover, we also demonstrated the protective roles of melatonin and resveratrol against MMA-induced inflammation and decrease in glutathione levels. In summary, our findings support the hypothesis that astroglial changes are associated with pathogenesis of methylmalonic acidemia. In addition, we showed that these cells might be potential targets for preventive/therapeutic strategies by using molecules, such as melatonin and resveratrol, which mediated glioprotection in this inborn error of metabolism.
Collapse
Affiliation(s)
- Rômulo Rodrigo de Souza Almeida
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Belisa Parmeggiani
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Krista Minéia Wartchow
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Diogo Onofre Souza
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Carlos-Alberto Gonçalves
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Guilhian Leipnitz
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - André Quincozes-Santos
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil. .,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
197
|
Bao ZH, Hou XB, Li HL, Mao YF, Wang WR. The mechanism and progress of ferroptosis in pancreatic cancer. Acta Histochem 2022; 124:151919. [PMID: 35772355 DOI: 10.1016/j.acthis.2022.151919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/08/2022] [Accepted: 06/08/2022] [Indexed: 12/01/2022]
Abstract
Pancreatic cancer is one of the deadliest cancers in the world, causing hundreds of thousands of deaths worldwide annually. Because of late diagnosis, rapid metastasis and drug resistance to chemotherapy, pancreatic cancer has a poor prognosis. Although the treatment of pancreatic cancer has made tremendous progress, the options for effective treatment are still limited, and new treatment methods are in crying needs to improve prognosis in clinic. Ferroptosis is an iron-dependent non-apoptotic cell death mode, which is mediated by lipid peroxidation and iron accumulation. Ferroptosis plays a momentous role in regulating different cancers in recent years, such as breast cancer, hepatocellular carcinoma, lung cancer and pancreatic cancer. In this present review, we elaborate on the regulatory mechanisms and signaling pathways of ferroptosis in pancreatic cancer, with the intention of delivering directions and new ideas for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Zhi-Hang Bao
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui 233030, China; Department of Clinical Medicine, Bengbu Medical College, Anhui 233030, China
| | - Xiang-Bin Hou
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui 233030, China; Department of Clinical Medicine, Bengbu Medical College, Anhui 233030, China
| | - Hao-Ling Li
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui 233030, China; Department of Clinical Medicine, Bengbu Medical College, Anhui 233030, China
| | - Yi-Feng Mao
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui 233030, China; Department of Clinical Medicine, Bengbu Medical College, Anhui 233030, China
| | - Wen-Rui Wang
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui 233030, China; Department of Life Sciences, Bengbu Medical College, Anhui 233030, China.
| |
Collapse
|
198
|
Estaras M, Martinez R, Garcia A, Ortiz-Placin C, Iovanna JL, Santofimia-Castaño P, Gonzalez A. Melatonin modulates metabolic adaptation of pancreatic stellate cells subjected to hypoxia. Biochem Pharmacol 2022; 202:115118. [PMID: 35671789 DOI: 10.1016/j.bcp.2022.115118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022]
Abstract
Pancreatic stellate cells (PSCs), the main cell type responsible for the development of fibrosis in pancreatic cancer, proliferate actively under hypoxia. Melatonin has received attention as a potential antifibrotic agent due to its anti-proliferative actions on PSCs. In this work, we investigated the activation of the PI3K/Akt/mTOR pathway and the metabolic adaptations that PSCs undergo under hypoxic conditions, as well as the probable modulation by melatonin. Incubation of cells under hypoxia induced an increase in cell proliferation, and in the expression of alpha-smooth muscle actin and of collagen type 1. In addition, an increase in the phosphorylation of Akt was observed, whereas a decrease in the phosphorylation of PTEN and GSK-3b was noted. The phosphorylation of mTOR and its substrate p70 S6K was decreased under hypoxia. Treatment of PSCs with melatonin under hypoxia diminished cell proliferation, the levels of alpha-smooth muscle actin and of collagen type 1, the phosphorylation of Akt and increased phosphorylation of mTOR. Mitochondrial activity decreased in PSCs under hypoxia. A glycolytic shift was observed. Melatonin further decreased mitochondrial activity. Under hypoxia, no increase in autophagic flux was noted. However, melatonin treatment induced autophagy activation. Nevertheless, inhibition of this process did not induce detectable changes in the viability of cells treated with melatonin. We conclude that PSCs undergo metabolic adaptation under hypoxia that might help them survive and that pharmacological concentrations of melatonin modulate cell responses to hypoxia. Our results contribute to the knowledge of the mechanisms by which melatonin could modulate fibrosis within the pancreas.
Collapse
Affiliation(s)
- Matias Estaras
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, Cáceres, España
| | - Remigio Martinez
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de Extremadura, Cáceres, España
| | - Alfredo Garcia
- Departamento de Producción Animal, CICYTEX-La Orden, Guadajira, Badajoz, España
| | - Candido Ortiz-Placin
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, Cáceres, España
| | - Juan L Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Antonio Gonzalez
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, Cáceres, España.
| |
Collapse
|
199
|
De Bock M, De Smet MA, Verwaerde S, Tahiri H, Schumacher S, Van Haver V, Witschas K, Steinhäuser C, Rouach N, Vandenbroucke RE, Leybaert L. Targeting gliovascular connexins prevents inflammatory blood-brain barrier leakage and astrogliosis. JCI Insight 2022; 7:135263. [PMID: 35881483 PMCID: PMC9462469 DOI: 10.1172/jci.insight.135263] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
The blood-brain barrier is formed by capillary endothelial cells expressing Cx37, Cx40 and Cx43, and is joined by closely apposed astrocytes expressing Cx43 and Cx30. We investigated whether connexin-targeting peptides could limit barrier leakage triggered by LPS-induced systemic inflammation in mice. Intraperitoneal LPS increased endothelial and astrocytic Cx43 expression, elevated TNFα, IL1β, IFNγ and IL6 in plasma and IL6 in the brain, and induced barrier leakage recorded over 24h. Barrier leakage was largely prevented by global Cx43 knockdown and Cx43/Cx30 double-knockout in astrocytes, slightly diminished by endothelial Cx43 knockout and not protected by global Cx30 knockout. Intravenous administration of Gap27 or Tat-Gap19 just before LPS also prevented barrier leakage, and intravenous BAPTA-AM to chelate intracellular calcium was equally effective. Patch-clamp experiments demonstrated LPS-induced Cx43 hemichannel opening in endothelial cells, which was suppressed by Gap27, Gap19 and BAPTA. LPS additionally triggered astrogliosis that was prevented by intravenous Tat-Gap19 or BAPTA-AM. Cortically applied Tat-Gap19 or BAPTA-AM to primarily target astrocytes, also strongly diminished barrier leakage. In vivo dye uptake and in vitro patch-clamp showed Cx43 hemichannel opening in astrocytes that was induced by IL6 in a calcium-dependent manner. We conclude that targeting endothelial and astrocytic connexins is a powerful approach to limit barrier failure and astrogliosis.
Collapse
Affiliation(s)
- Marijke De Bock
- Department of Basic & Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Maarten Aj De Smet
- Department of Basic & Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Stijn Verwaerde
- Department of Basic & Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Hanane Tahiri
- Department of Basic & Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Steffi Schumacher
- Department of Basic & Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Valérie Van Haver
- Department of Basic & Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Katja Witschas
- Department of Basic & Applied Medical Sciences, Ghent University, Ghent, Belgium
| | | | - Nathalie Rouach
- Center for Interdisiplinary Research in Biology (CIRB), College de France, Paris, France
| | | | - Luc Leybaert
- Department of Basic & Applied Medical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
200
|
Santofimia-Castaño P, Huang C, Liu X, Xia Y, Audebert S, Camoin L, Peng L, Lomberk G, Urrutia R, Soubeyran P, Neira JL, Iovanna J. NUPR1 protects against hyperPARylation-dependent cell death. Commun Biol 2022; 5:732. [PMID: 35869257 PMCID: PMC9307593 DOI: 10.1038/s42003-022-03705-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 07/12/2022] [Indexed: 01/25/2023] Open
Abstract
Proteomic, cellular and biochemical analysis of the stress protein NUPR1 reveals that it binds to PARP1 into the nucleus and inhibits PARP1 activity in vitro. Mutations on residues Ala33 or Thr68 of NUPR1 or treatment with its inhibitor ZZW-115 inhibits this effect. PARylation induced by 5-fluorouracil (5-FU) treatment is strongly enhanced by ZZW-115 and associated with a decrease of NAD+/NADH ratio and rescued by the PARP inhibitor olaparib. Cell death induced by ZZW-115 treatment of pancreas cancer-derived cells is rescued by olaparib and improved with PARG inhibitor PDD00017273. The mitochondrial catastrophe induced by ZZW-115 treatment or by genetic inactivation of NUPR1 is associated to a hyperPARylation of the mitochondria, disorganization of the mitochondrial network, mitochondrial membrane potential decrease, and with increase of superoxide production, intracellular level of reactive oxygen species (ROS) and cytosolic levels of Ca2+. These features are rescued by olaparib or NAD+ precursor nicotinamide mononucleotide in a dose-dependent manner and partially by antioxidants treatments. In conclusion, inactivation of NUPR1 induces a hyperPARylation, which in turn, induces a mitochondrial catastrophe and consequently a cell death through a non-canonical Parthanatos, since apoptosis inducing-factor (AIF) is not translocated out of the mitochondria.
Collapse
Affiliation(s)
- Patricia Santofimia-Castaño
- grid.5399.60000 0001 2176 4817Centre de Recherche en Cancérologie de Marseille (CRCM), Parc Scientifique et Technologique de Luminy, INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Can Huang
- grid.5399.60000 0001 2176 4817Centre de Recherche en Cancérologie de Marseille (CRCM), Parc Scientifique et Technologique de Luminy, INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Xi Liu
- grid.5399.60000 0001 2176 4817Centre de Recherche en Cancérologie de Marseille (CRCM), Parc Scientifique et Technologique de Luminy, INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Yi Xia
- grid.190737.b0000 0001 0154 0904Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, PR China
| | - Stephane Audebert
- grid.5399.60000 0001 2176 4817Centre de Recherche en Cancérologie de Marseille (CRCM), Parc Scientifique et Technologique de Luminy, INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Luc Camoin
- grid.5399.60000 0001 2176 4817Centre de Recherche en Cancérologie de Marseille (CRCM), Parc Scientifique et Technologique de Luminy, INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Ling Peng
- grid.5399.60000 0001 2176 4817Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, «Equipe Labellisée Ligue Contre le Cancer», Parc Scientifique et Technologique de Luminy, Aix-Marseille Université, Marseille, France
| | - Gwen Lomberk
- grid.30760.320000 0001 2111 8460Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI USA
| | - Raul Urrutia
- grid.30760.320000 0001 2111 8460Genomic Science and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, WI USA
| | - Philippe Soubeyran
- grid.5399.60000 0001 2176 4817Centre de Recherche en Cancérologie de Marseille (CRCM), Parc Scientifique et Technologique de Luminy, INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Jose Luis Neira
- grid.26811.3c0000 0001 0586 4893Instituto de Biología Molecular y Celular, Edificio Torregaitán, Universidad Miguel Hernández, Elche, Alicante Spain
| | - Juan Iovanna
- grid.5399.60000 0001 2176 4817Centre de Recherche en Cancérologie de Marseille (CRCM), Parc Scientifique et Technologique de Luminy, INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| |
Collapse
|