1
|
Wang X, Padawer-Curry JA, Bice AR, Kim B, Rosenthal ZP, Lee JM, Goyal MS, Macauley SL, Bauer AQ. Spatiotemporal relationships between neuronal, metabolic, and hemodynamic signals in the awake and anesthetized mouse brain. Cell Rep 2024; 43:114723. [PMID: 39277861 PMCID: PMC11523563 DOI: 10.1016/j.celrep.2024.114723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 07/08/2024] [Accepted: 08/21/2024] [Indexed: 09/17/2024] Open
Abstract
Neurovascular coupling (NVC) and neurometabolic coupling (NMC) provide the basis for functional magnetic resonance imaging and positron emission tomography to map brain neurophysiology. While increases in neuronal activity are often accompanied by increases in blood oxygen delivery and oxidative metabolism, these observations are not the rule. This decoupling is important when interpreting brain network organization (e.g., resting-state functional connectivity [RSFC]) because it is unclear whether changes in NMC/NVC affect RSFC measures. We leverage wide-field optical imaging in Thy1-jRGECO1a mice to map cortical calcium activity in pyramidal neurons, flavoprotein autofluorescence (representing oxidative metabolism), and hemodynamic activity during wake and ketamine/xylazine anesthesia. Spontaneous dynamics of all contrasts exhibit patterns consistent with RSFC. NMC/NVC relative to excitatory activity varies over the cortex. Ketamine/xylazine profoundly alters NVC but not NMC. Compared to awake RSFC, ketamine/xylazine affects metabolic-based connectomes moreso than hemodynamic-based measures of RSFC. Anesthesia-related differences in NMC/NVC timing do not appreciably alter RSFC structure.
Collapse
Affiliation(s)
- Xiaodan Wang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO 63130, USA
| | - Jonah A Padawer-Curry
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Imaging Sciences Program, Washington University in Saint Louis, St. Louis, MO 63130, USA
| | - Annie R Bice
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Byungchan Kim
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Zachary P Rosenthal
- Department of Psychiatry, University of Pennsylvania Health System Penn Medicine, Philadelphia, PA 19104, USA
| | - Jin-Moo Lee
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO 63130, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Manu S Goyal
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shannon L Macauley
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA
| | - Adam Q Bauer
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO 63130, USA; Imaging Sciences Program, Washington University in Saint Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
2
|
Hsing CH, Hung YP, Lin MC, Chen CL, Wang YT, Tseng PC, Satria RD, Lin CF. Overdose with the anesthetic propofol causes hematological cytotoxicity and immune cell alteration in an experimental ex vivo whole blood culture model. Toxicol In Vitro 2024; 94:105729. [PMID: 37935310 DOI: 10.1016/j.tiv.2023.105729] [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: 08/09/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Propofol, an anesthetic characterized by its benefits of rapid induction, maintenance, and recovery times, may cause cytotoxic effects, resulting in propofol infusion syndrome (PRIS). In addition to causing dyslipidemia in PRIS, our previous works showed that propofol overdose induced phagocyte apoptosis. This study, using an experimental ex vivo model of propofol treatment, investigated the possible cytopathology in the blood. A complete blood count examination showed the deregulating effects of propofol overdose 24 h postinoculation, characterized by mononuclear cell increase (lymphocyte and monocyte subsets) and granulocyte decrease. Advanced marker-based flow cytometric analysis confirmed these findings, although there was no change in CD14+ monocytes. Blood smear staining showed the deregulating effects of propofol overdose 24 h postinoculation, characterized by cytosolic vacuolization and cytotoxicity, particularly in neutrophils. Immune cell profiling of caspase-3 activation demonstrated the induction of cell apoptosis following propofol overdose treatment, particularly in granulocytes. Using multiparameter flow cytometry, this study further analyzed the changes in the profile of immune cells, showing a notable increase in CD4 + HLA-DR-CD62L- helper T cells. These studies explored an ex vivo model of cytopathogenic propofol overdose and its special immune-deregulating effects on peripheral blood cells.
Collapse
Affiliation(s)
- Chung-Hsi Hsing
- Department of Anesthesiology, Chi-Mei Medical Center, Tainan 710, Taiwan; Department of Medical Research, Chi-Mei Medical Center, Tainan 710, Taiwan; Department of Anesthesiology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Ping Hung
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Ming-Chung Lin
- Department of Anesthesiology, Chi-Mei Medical Center, Tainan 710, Taiwan; Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan 717, Taiwan
| | - Chia-Ling Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yung-Ting Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Po-Chun Tseng
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei 110, Taiwan
| | - Rahmat Dani Satria
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Department of Clinical Pathology and Laboratory Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia; Clinical Laboratory Installation, Dr. Sardjito Central General Hospital, Yogyakarta 55281, Indonesia
| | - Chiou-Feng Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
3
|
Zhou Z, Dai W, Liu T, Shi M, Wei Y, Chen L, Xie Y. Transfer of massive mitochondria from astrocytes reduce propofol neurotoxicity. Neurosci Lett 2024; 818:137542. [PMID: 37926293 DOI: 10.1016/j.neulet.2023.137542] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023]
Abstract
Studies have shown that propofol-induced neurotoxicity is mediated by disruption of mitochondrial fission and fusion, leading to an imbalance in energy supply for developing neurons. Healthy mitochondria released from astrocytes migrate to compromised neurons to mitigate propofol-induced neurotoxicity, yet the precise mechanisms involved require further clarification. In our investigation, primary neurons were incubated with propofol, which decreased ATP synthesis and mitochondrial membrane potential, increased ROS generation and neuronal apoptosis. Notably, astrocytes did not respond to the deleterious effects of propofol. The culture medium of neurons or astrocytes incubated with propofol was collected. It was found that mitochondrial ratio was decreased and mitochondrial function was impaired. Non-contact co-culture of neuro-astrocytes facilitated transcellular mitochondrial transfer in both physiological and propofol interventions, but failed to reverse propofol-induced neurotoxicity. The more pronounced damage to neuronal mitochondria induced by propofol compared to that in astrocytes alludes to secondary injury. Damaged neurons incubated with large, functional extracellular mitochondria derived from astrocytes demonstrates transfer of mitochondria to neurons, effectively reversing propofol-induced neurotoxicity. This discovery presents a novel mitochondrial transfer of neuro-astrocytes crosstalk that contributes to neuroprotection and neurological recovery in neurotoxicity.
Collapse
Affiliation(s)
- Zhan Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Weixin Dai
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Tianxiao Liu
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Min Shi
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Yi Wei
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Lifei Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Yubo Xie
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
4
|
Slingerland-Boot R, Kummerow M, Arbous SM, van Zanten ARH. Association between first-week propofol administration and long-term outcomes of critically ill mechanically ventilated patients: A retrospective cohort study. Clin Nutr 2024; 43:42-51. [PMID: 38000194 DOI: 10.1016/j.clnu.2023.10.029] [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: 06/20/2023] [Revised: 09/21/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND & AIM Propofol is commonly used in ICUs, but its long-term effects have not been thoroughly studied. In vitro studies suggest it may harm mitochondrial function, potentially affecting clinical outcomes. This study aimed to investigate the association between substantial propofol sedation and clinical outcomes in critically ill patients. METHODS We conducted a single-centre cohort study of critically ill, mechanically ventilated (≥7 days) adults to compare patients who received a substantial dose of propofol (cumulative >500 mg) during the first week of ICU admission with those who did not. The primary outcome was the association between substantial propofol administration and 6-month mortality, adjusted for relevant covariates. Subanalyses were performed for administration in the early (day 1-3) and late (day 4-7) acute phases of critical illness due to the metabolic changes in this period. Secondary outcomes included tracheostomy need and duration, length of ICU and hospital stay (LOS), discharge destinations, ICU, hospital, and 3-month mortality. RESULTS A total of 839 patients were enrolled, with 73.7 % receiving substantial propofol administration (substantial propofol dose group). Six-month all-cause mortality was 32.4 %. After adjusting for relevant variables, we found no statistically significant difference in 6-month mortality between both groups. There were also no significant differences in secondary outcomes. CONCLUSION Our study suggests that substantial propofol administration during the first week of ICU stay in the least sick critically ill, mechanically ventilated adult patients is safe, with no significant associations found with 6-month mortality, ICU or hospital LOS, differences in discharge destinations or need for tracheostomy.
Collapse
Affiliation(s)
- Rianne Slingerland-Boot
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Ede, the Netherlands; Wageningen University & Research, Division of Human Nutrition and Health, Wageningen, the Netherlands
| | - Maren Kummerow
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Ede, the Netherlands
| | - Sesmu M Arbous
- Department of Intensive Care Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Arthur R H van Zanten
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Ede, the Netherlands; Wageningen University & Research, Division of Human Nutrition and Health, Wageningen, the Netherlands.
| |
Collapse
|
5
|
Van S, Lam V, Patel K, Humphries A, Siddiqi J. Propofol-Related Infusion Syndrome: A Bibliometric Analysis of the 100 Most-Cited Articles. Cureus 2023; 15:e46497. [PMID: 37927719 PMCID: PMC10624560 DOI: 10.7759/cureus.46497] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023] Open
Abstract
Propofol-related infusion syndrome (PRIS) is a rare, yet life-threatening sequelae to prolonged administration of the anesthetic propofol in mechanically intubated patients. The condition is characterized by progressive multi-system organ failure and eventual mortality; of note, the predominant characteristics of PRIS involve but are not limited to cardiovascular impairment and collapse, metabolic and lactic acidosis, rhabdomyolysis, hyperkalemia, and acute renal failure. While potent or extended doses of propofol have been found to be the primary precipitating factor of this condition, others such as age, critical illness, steroid therapy, and hyperlipidemia have been discovered to play a role as well. This bibliometric analysis was done to reflect the current relevance and understanding of PRIS in recent literature. The SCOPUS database was utilized to conduct a search for articles with keywords "propofol infusion syndrome" and "propofol syndrome" from February 24, 2001, until April 16, 2023, with parameters for article title, citation number, citation per year, author, institution, publishing journal, and country of origin. PRIS was first defined in 1990, just a year after its approval by the Food and Drug Administration for use as a sedative-hypnotic. Since then, interest in PRIS slowly rose up to 13 publications per year in 2013. Seven papers on the topic were published in Critical Care Medicine, six in Neurocritical Care, and four in Anesthesia. The most common institutions were Mayo Clinic, Northeastern University, and Tufts Medical Center. To our knowledge, this is the first bibliometric analysis to evaluate the most influential publications about PRIS. A majority of the research is case-based, possibly owing to the rarity of the condition. Our research suggests that confounding factors outside the precipitating dosage of propofol may be implicated in the onset and progression of PRIS. This study could therefore bring renewed interest to the topic and lead to additional research focused on fully understanding the pathophysiology of PRIS in order to promote the development of novel diagnostics and treatment.
Collapse
Affiliation(s)
- Sophie Van
- Anesthesiology, California University of Science and Medicine, Colton, USA
| | - Vicky Lam
- Anesthesiology, California University of Science and Medicine, Colton, USA
| | - Kisan Patel
- Physical Medicine and Rehabilitation, California University of Science and Medicine, Colton, USA
| | - Andrew Humphries
- Anesthesiology, California University of Science and Medicine, Colton, USA
| | - Javed Siddiqi
- Neurological Surgery, Riverside University Health System Medical Center, Moreno Valley, USA
| |
Collapse
|
6
|
Zhou R, Konishi Y, Zhang A, Nishiwaki K. Propofol elicits apoptosis and attenuates cell growth in esophageal cancer cell lines. NAGOYA JOURNAL OF MEDICAL SCIENCE 2023; 85:579-591. [PMID: 37829490 PMCID: PMC10565583 DOI: 10.18999/nagjms.85.3.579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/24/2022] [Indexed: 10/14/2023]
Abstract
Propofol is a pharmaceutical agent commonly used as an intravenous anesthetic in surgical treatments and a sedative in intensive care. However, it is largely unknown how exposure to propofol affects the proliferation, invasion, and apoptosis of neoplastic cells in esophageal cancer. In this study, we sought to elucidate the impact of propofol exposure on the growth properties of human esophageal cancer cell lines in vitro. We treated two human esophageal cancer cell lines, KYSE30 and KYSE960, with up to 10 µg/mL of propofol for 12-36 h. The treated cells were then analyzed by cell proliferation assay, Matrigel invasion assay, quantification of caspase-3/7 and -9 activities, and cell staining with Annexin V and 7-aminoactinomycin D to detect early apoptosis and cell death, respectively, via flow cytometry. We found that 3-5 µg/mL propofol reduced the growth and Matrigel invasion of both cell lines in a dose-dependent manner. Executioner caspase-3/7, but not caspase-9 involved in intrinsic apoptosis pathway, was activated by cell exposure to 3-5 µg/mL propofol. In addition, 3-5 µg/mL propofol augmented early apoptosis in both cell lines and increased cell death in the KYSE30 cell line. In summary, exposure to propofol, at concentrations up to 5 µg/mL, led to the reduction of cell growth and Matrigel invasion, as well as the augmentation of apoptosis in esophageal cancer cell lines. These data will help define a methodology to safely utilize propofol, a common general anesthetic and sedative, with esophageal cancer patients.
Collapse
Affiliation(s)
- Rui Zhou
- Department of Anesthesiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuko Konishi
- Endowed Division of Perioperative Management, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ailing Zhang
- Department of Anesthesiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kimitoshi Nishiwaki
- Department of Anesthesiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
7
|
Guan S, Sun L, Wang X, Huang X, Luo T. Propofol inhibits neuroinflammation and metabolic reprogramming in microglia in vitro and in vivo. Front Pharmacol 2023; 14:1161810. [PMID: 37383725 PMCID: PMC10293632 DOI: 10.3389/fphar.2023.1161810] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/12/2023] [Indexed: 06/30/2023] Open
Abstract
Microglial activation-induced neuroinflammation is closely related to the development of sepsis-associated encephalopathy. Accumulating evidence suggests that changes in the metabolic profile of microglia is crucial for their response to inflammation. Propofol is widely used for sedation in mechanically ventilated patients with sepsis. Here, we investigate the effect of propofol on lipopolysaccharide-induced neuroinflammation, neuronal injuries, microglia metabolic reprogramming as well as the underlying molecular mechanisms. The neuroprotective effects of propofol (80 mg/kg) in vivo were measured in the lipopolysaccharide (2 mg/kg)-induced sepsis in mice through behavioral tests, Western blot analysis and immunofluorescent staining. The anti-inflammatory effects of propofol (50 μM) in microglial cell cultures under lipopolysaccharide (10 ng/ml) challenge were examined with Seahorse XF Glycolysis Stress test, ROS assay, Western blot, and immunofluorescent staining. We showed that propofol treatment reduced microglia activation and neuroinflammation, inhibited neuronal apoptosis and improved lipopolysaccharide-induced cognitive dysfunction. Propofol also attenuated lipopolysaccharide-stimulated increases of inducible nitric oxide synthase, nitric oxide, tumor necrosis factor-α, interlukin-1β and COX-2 in cultured BV-2 cells. Propofol-treated microglia showed a remarkable suppression of lipopolysaccharide-induced HIF-1α, PFKFB3, HK2 expression and along with downregulation of the ROS/PI3K/Akt/mTOR signaling pathway. Moreover, propofol attenuated the enhancement of mitochondrial respiration and glycolysis induced by lipopolysaccharide. Together, our data suggest that propofol attenuated inflammatory response by inhibiting metabolic reprogramming, at least in part, through downregulation of the ROS/PI3K/Akt/mTOR/HIF-1α signaling pathway.
Collapse
|
8
|
Yoshida T, Okumura T, Matsuo Y, Okuyama T, Michiura T, Kaibori M, Umezaki N, Bono H, Hirota K, Sekimoto M. Activation of transcription factor HIF inhibits IL-1β-induced NO production in primary cultured rat hepatocytes. Nitric Oxide 2022; 124:1-14. [PMID: 35460897 DOI: 10.1016/j.niox.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
Roxadustat and other hypoxia-inducible factor-prolyl hydroxylase inhibitors (HIF-PHIs) have recently been approved for the treatment of chronic renal anemia. In macrophages and monocytes, the activation of HIF-1 by pro-inflammatory cytokines induces iNOS expression and activity through the NF-κB pathway to produce nitric oxide (NO), which causes liver injury when excessively produced. Few studies have reported a relationship between HIF activity and iNOS induction in hepatocytes. We investigated the effect of drug- and hypoxia-induced HIF activations on NO production in primary cultured rat hepatocytes. Roxadustat treatment and hypoxic conditions activated HIF. Contrary to expectations, HIF-PHI treatment and hypoxia inhibited IL-1β-induced NO production. RNA-Seq analysis of mRNA expression in rat hepatocytes showed that roxadustat treatment decreased the expression of genes related to inflammation, and genes in the NF-κB signaling pathway were induced by IL-1β. Moreover, roxadustat suppressed IL-1β-activated signaling pathways in an HIF-dependent manner. GalN/LPS-treated rats were used as in vivo models of hepatic injury, and roxadustat treatment showed a tendency to suppress the death of rats. Therefore, exogenous HIF-1 activation, including HIF-PHI and hypoxia exposures, suppressed IL-1β-induced iNOS mRNA expression and subsequent NO production in hepatocytes, by suppressing the NF-κB signaling pathway. Roxadustat treatment suppresses the expression of pro-inflammatory genes by activating HIF, and thus may exhibit hepatoprotective effects.
Collapse
Affiliation(s)
- Terufumi Yoshida
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.
| | - Tadayoshi Okumura
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan; Research Organization of Science and Technology, Ritsumeikan University, 1-1 Noji-higashi, Kusatsu, Shiga, 525-8577, Japan.
| | - Yoshiyuki Matsuo
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.
| | - Tetsuya Okuyama
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan; Research Organization of Science and Technology, Ritsumeikan University, 1-1 Noji-higashi, Kusatsu, Shiga, 525-8577, Japan.
| | - Taku Michiura
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.
| | - Masaki Kaibori
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.
| | - Nodoka Umezaki
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.
| | - Hidemasa Bono
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-0046, Japan.
| | - Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.
| | - Mitsugu Sekimoto
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.
| |
Collapse
|
9
|
Takenaga K, Koshikawa N, Nagase H. Intercellular transfer of mitochondrial DNA carrying metastasis-enhancing pathogenic mutations from high- to low-metastatic tumor cells and stromal cells via extracellular vesicles. BMC Mol Cell Biol 2021; 22:52. [PMID: 34615464 PMCID: PMC8496074 DOI: 10.1186/s12860-021-00391-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/20/2021] [Indexed: 01/08/2023] Open
Abstract
Background Mitochondrial DNA (mtDNA) carrying certain pathogenic mutations or single nucleotide variants (SNVs) enhances the invasion and metastasis of tumor cells, and some of these mutations are homoplasmic in tumor cells and even in tumor tissues. On the other hand, intercellular transfer of mitochondria and cellular components via extracellular vesicles (EVs) and tunneling nanotubes (TNTs) has recently attracted intense attention in terms of cell-to-cell communication in the tumor microenvironment. It remains unclear whether metastasis-enhancing pathogenic mutant mtDNA in tumor cells is intercellularly transferred between tumor cells and stromal cells. In this study, we investigated whether mtDNA with the NADH dehydrogenase subunit 6 (ND6) G13997A pathogenic mutation in highly metastatic cells can be horizontally transferred to low-metastatic cells and stromal cells in the tumor microenvironment. Results When MitoTracker Deep Red-labeled high-metastatic Lewis lung carcinoma A11 cells carrying the ND6 G13997A mtDNA mutation were cocultured with CellLight mitochondria-GFP-labeled low-metastatic P29 cells harboring wild-type mtDNA, bidirectional transfer of red- and green-colored vesicles, probably mitochondria-related EVs, was observed in a time-dependent manner. Similarly, intercellular transfer of mitochondria-related EVs occurred between A11 cells and α-smooth muscle actin (α-SMA)-positive cancer-associated fibroblasts (CAFs, WA-mFib), macrophages (RAW264.7) and cytotoxic T cells (CTLL-2). Intercellular transfer was suppressed by inhibitors of EV release. The large and small EV fractions (L-EV and S-EV, respectively) prepared from the conditioned medium by differential ultracentrifugation both were found to contain mtDNA, although only S-EVs were efficiently incorporated into the cells. Several subpopulations had evidence of LC3-II and contained degenerated mitochondrial components in the S-EV fraction, signaling to the existence of autophagy-related S-EVs. Interestingly, the S-EV fraction contained a MitoTracker-positive subpopulation, which was inhibited by the respiration inhibitor antimycin A, indicating the presence of mitochondria with membrane potential. It was also demonstrated that mtDNA was transferred into mtDNA-less ρ0 cells after coculture with the S-EV fraction. In syngeneic mouse subcutaneous tumors formed by a mixture of A11 and P29 cells, the mitochondria-related EVs released from A11 cells reached distantly positioned P29 cells and CAFs. Conclusions These results suggest that metastasis-enhancing pathogenic mtDNA derived from metastatic tumor cells is transferred to low-metastatic tumor cells and stromal cells via S-EVs in vitro and in the tumor microenvironment, inferring a novel mechanism of enhancement of metastatic potential during tumor progression. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00391-5.
Collapse
Affiliation(s)
- Keizo Takenaga
- Division of Cancer Genetics, Chiba Cancer Center Research Institute, Nitona, Chuoh-ku, Chiba, Japan.
| | - Nobuko Koshikawa
- Division of Cancer Genetics, Chiba Cancer Center Research Institute, Nitona, Chuoh-ku, Chiba, Japan
| | - Hiroki Nagase
- Division of Cancer Genetics, Chiba Cancer Center Research Institute, Nitona, Chuoh-ku, Chiba, Japan
| |
Collapse
|
10
|
Cao Y, Fan L, Li L, Zhou J. Propofol suppresses cell proliferation in gastric cancer cells through NRF2-mediated polyol pathway. Clin Exp Pharmacol Physiol 2021; 49:264-274. [PMID: 34570396 PMCID: PMC9299175 DOI: 10.1111/1440-1681.13595] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/15/2021] [Accepted: 09/21/2021] [Indexed: 12/28/2022]
Abstract
Propofol, a widely used short‐acting intravenous sedative agent, has gradually gained attention due to the tumour‐suppressing role and non‐anaesthetic effect. Dysfunction of metabolic reprogramming has been recognised as a well‐documented factor for tumour progression. The aim of this study is to explore the effect of propofol on the polyol pathway in gastric cancer cells. In this study, we found that propofol treatment led to a significant downregulation of cell proliferation in BGC823 and GES‐1 cells, which was attributed to the decreased AR‐mediated polyol pathway. Both aldo‐keto reductase family 1, member B1 (AKR1B1) and AKR1B10 were significantly reduced in BGC823 and GES‐1 cells in response to propofol stimulation, leading to decreased AR activity and sorbitol level. Addition of sorbitol could reverse the inhibitory effect of propofol on cell proliferation. Mechanically, propofol treatment drastically inhibited phosphorylation and nuclear translocation of nuclear factor (erythroid‐derived 2)‐like 2 (NRF2), subsequently decreased the binding of NRF2 to AR promoter. Overexpression of NRF2 resulted in the recovery of AR expression in gastric cancer cell with propofol treatment. Taken together, these finding showed that propofol suppressed cell proliferation in BGC823 and GES‐1 cell through NRF2‐mediated polyol pathway, which would aid the selection of sedation for patients with gastric cancer.
Collapse
Affiliation(s)
- Yajun Cao
- Department of Anesthesia, Zhuhai Center for Maternal and Child Health Care, Zhuhai, China
| | - Long Fan
- Department of Pharmacy, Zhuhai Center for Maternal and Child Health Care, Zhuhai, China
| | - Linkai Li
- Department of Pharmacy, Zhuhai Center for Maternal and Child Health Care, Zhuhai, China
| | - Jiexian Zhou
- Department of Anesthesia, Zhuhai Center for Maternal and Child Health Care, Zhuhai, China
| |
Collapse
|
11
|
Venu VKP, Saifeddine M, Mihara K, Faiza M, Gorobets E, Flewelling AJ, Derksen DJ, Hirota SA, Marei I, Al-Majid D, Motahhary M, Ding H, Triggle CR, Hollenberg MD. Metformin Prevents Hyperglycemia-Associated, Oxidative Stress-Induced Vascular Endothelial Dysfunction: Essential Role for the Orphan Nuclear Receptor Human Nuclear Receptor 4A1 (Nur77). Mol Pharmacol 2021; 100:428-455. [PMID: 34452975 DOI: 10.1124/molpharm.120.000148] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 08/17/2021] [Indexed: 01/22/2023] Open
Abstract
Vascular pathology is increased in diabetes because of reactive-oxygen-species (ROS)-induced endothelial cell damage. We found that in vitro and in a streptozotocin diabetes model in vivo, metformin at diabetes-therapeutic concentrations (1-50 µM) protects tissue-intact and cultured vascular endothelial cells from hyperglycemia/ROS-induced dysfunction typified by reduced agonist-stimulated endothelium-dependent, nitric oxide-mediated vasorelaxation in response to muscarinic or proteinase-activated-receptor 2 agonists. Metformin not only attenuated hyperglycemia-induced ROS production in aorta-derived endothelial cell cultures but also prevented hyperglycemia-induced endothelial mitochondrial dysfunction (reduced oxygen consumption rate). These endothelium-protective effects of metformin were absent in orphan-nuclear-receptor Nr4a1-null murine aorta tissues in accord with our observing a direct metformin-Nr4a1 interaction. Using in silico modeling of metformin-NR4A1 interactions, Nr4a1-mutagenesis, and a transfected human embryonic kidney 293T cell functional assay for metformin-activated Nr4a1, we identified two Nr4a1 prolines, P505/P549 (mouse sequences corresponding to human P501/P546), as key residues for enabling metformin to affect mitochondrial function. Our data indicate a critical role for Nr4a1 in metformin's endothelial-protective effects observed at micromolar concentrations, which activate AMPKinase but do not affect mitochondrial complex-I or complex-III oxygen consumption rates, as does 0.5 mM metformin. Thus, therapeutic metformin concentrations requiring the expression of Nr4a1 protect the vasculature from hyperglycemia-induced dysfunction in addition to metformin's action to enhance insulin action in patients with diabetes. SIGNIFICANCE STATEMENT: Metformin improves diabetic vasodilator function, having cardioprotective effects beyond glycemic control, but its mechanism to do so is unknown. We found that metformin at therapeutic concentrations (1-50µM) prevents hyperglycemia-induced endothelial dysfunction by attenuating reactive oxygen species-induced damage, whereas high metformin (>250 µM) impairs vascular function. However, metformin's action requires the expression of the orphan nuclear receptor NR4A1/Nur77. Our data reveal a novel mechanism whereby metformin preserves diabetic vascular endothelial function, with implications for developing new metformin-related therapeutic agents.
Collapse
Affiliation(s)
- Vivek Krishna Pulakazhi Venu
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Mahmoud Saifeddine
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Koichiro Mihara
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Muniba Faiza
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Evgueni Gorobets
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Andrew J Flewelling
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Darren J Derksen
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Simon A Hirota
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Isra Marei
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Dana Al-Majid
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Majid Motahhary
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Hong Ding
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Chris R Triggle
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Morley D Hollenberg
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| |
Collapse
|
12
|
Hu C, Iwasaki M, Liu Z, Wang B, Li X, Lin H, Li J, Li JV, Lian Q, Ma D. Lung but not brain cancer cell malignancy inhibited by commonly used anesthetic propofol during surgery: Implication of reducing cancer recurrence risk. J Adv Res 2021; 31:1-12. [PMID: 34194828 PMCID: PMC8240101 DOI: 10.1016/j.jare.2020.12.007] [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: 08/26/2020] [Revised: 10/27/2020] [Accepted: 12/12/2020] [Indexed: 12/27/2022] Open
Abstract
Introduction Intravenous anesthesia with propofol was reported to improve cancer surgical outcomes when compared with inhalational anesthesia. However, the underlying molecular mechanisms largely remain unknown. Objectives The anti-tumor effects of propofol and the possible underlying mechanism including altered metabolic and signaling pathways were studied in the current study. Methods The cell viability, proliferation, migration, and invasion of cancer cells were analyzed with CCK-8, Ki-67 staining, wound healing, and Transwell assay, respectively. The protein changes were analyzed with Western blot and immunofluorescent staining. The metabolomics alteration was studied with 1H-NMR spectroscopy. The gene expression regulations were analyzed with PCR gene array and qRT-PCR experiments. Results In this study, we found that propofol reduced cell viability and inhibited cell proliferation, migration and invasion of lung cancer cells, but not neuroglioma cells. In lung cancer cells, propofol downregulated glucose transporter 1 (GLUT1), mitochondrial pyruvate carrier 1 (MPC1), p-Akt, p-Erk1/2, and hypoxia- inducible factor 1 alpha (HIF-1 α ) expressions and upregulated pigment epithelium-derived factor (PEDF) expression. Propofol increased intracellular glutamate and glycine but decreased acetate and formate whilst increased glucose, lactate, glutamine, succinate, pyruvate, arginine, valine, isoleucine, and leucine and glycerol, and decreased acetate, ethanol, isopropanol in the culture media of lung cancer cells. Furthermore, VEGFA, CTBP1, CST7, CTSK, CXCL12, and CXCR4 gene expressions were downregulated, while NR4A3, RB1, NME1, MTSS1, NME4, SYK, APC, and FAT1 were upregulated following the propofol treatment. Consistent with the phenotypical changes, these molecular and metabolic changes were not found in the neuroglioma cells. Conclusion Our findings indicated anti-tumor effects of propofol on the lung cancer but not brain cancer, through the regulation of tumor metastasis-related genes, multi-cellular signaling and cellular metabolism.
Collapse
Affiliation(s)
- Cong Hu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, United Kingdom
| | - Masae Iwasaki
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, United Kingdom
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan
| | - Zhigang Liu
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| | - Bincheng Wang
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, United Kingdom
| | - Xiaomeng Li
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, United Kingdom
| | - Han Lin
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jun Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jia V. Li
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| | - Qingquan Lian
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, United Kingdom
| |
Collapse
|
13
|
Abstract
OBJECTIVE The aim of this study was to define the influence of trauma on cardiac glucose and fatty acid transport. The effects were investigated in vivo in a porcine mono- and polytrauma model and in vitro in human cardiomyocytes, which were treated simultaneously with different inflammatory substances, mimicking posttraumatic inflammatory conditions. METHODS AND RESULTS In the porcine fracture- and polytrauma model, blood glucose concentrations were measured by blood gas analysis during an observation period of 72 h. The expression of cardiac glucose and fatty acid transporters in the left ventricle was determined by RT-qPCR and immunofluorescence. Cardiac and hepatic glycogen storage was examined. Furthermore, human cardiomyocytes were exposed to a defined trauma-cocktail and the expression levels of glucose- and fatty acid transporters were determined. Early after polytrauma, hyperglycemia was observed. After 48 and 72 h, pigs with fracture- and polytrauma developed hypoglycemia. The propofol demand significantly increased posttrauma. The hepatic glycogen concentration was reduced 72 h after trauma. Cardiac glucose and fatty acid transporters changed in both trauma models in vivo as well as in vitro in human cardiomyocytes in presence of proinflammatory mediators. CONCLUSIONS Monotrauma as well as polytrauma changed the cardiac energy transport by altering the expression of glucose and fatty acid transporters. In vitro data suggest that human cardiomyocytes shift to a state alike myocardial hibernation preferring glucose as primary energy source to maintain cardiac function.
Collapse
|
14
|
Hirota K. Hypoxia-dependent signaling in perioperative and critical care medicine. J Anesth 2021; 35:741-756. [PMID: 34003375 PMCID: PMC8128984 DOI: 10.1007/s00540-021-02940-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 04/24/2021] [Indexed: 12/14/2022]
Abstract
A critical goal of patient management for anesthesiologists and intensivists is to maintain oxygen homeostasis in patients admitted to operation theaters and intensive care units. For this purpose, it is imperative to understand the strategies of the body against oxygen imbalance—especially oxygen deficiency (hypoxia). Adaptation to hypoxia and maintenance of oxygen homeostasis involve a wide range of responses that occur at different organizational levels in the body. These responses are greatly influenced by perioperative patient management including factors such as perioperative drugs. Herein, the influence of perioperative patient management on the body's response to oxygen imbalance was reviewed with a special emphasis on hypoxia-inducible factors (HIFs), transcription factors whose activity are regulated by the perturbation of oxygen metabolism. The 2019 Nobel Prize in Physiology or Medicine was awarded to three researchers who made outstanding achievements in this field. While previous studies have reported the effect of perioperatively used drugs on hypoxia-induced gene expression mediated by HIFs, this review focused on effects of subacute or chronic hypoxia changes in gene expression that are mediated by the transcriptional regulator HIFs. The clinical implications and perspectives of these findings also will be discussed. Understanding the basic biology of the transcription factor HIF can be informative for us since anesthesiologists manage patients during the perioperative period facing the imbalances the oxygen metabolism in organ and tissue. The clinical implications of hypoxia-dependent signaling in critical illness, including Coronavirus disease (COVID-19), in which disturbances in oxygen metabolism play a major role in its pathogenesis will also be discussed.
Collapse
Affiliation(s)
- Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan.
| |
Collapse
|
15
|
Mack N, Mazzio E, Badisa R, Soliman KFA. Metabolic Response to the Mitochondrial Toxin 1-Methyl-4-phenylpyridinium (MPP+) in LDH-A/B Double-knockout LS174T Colon Cancer Cells. Cancer Genomics Proteomics 2021; 18:385-405. [PMID: 33994363 DOI: 10.21873/cgp.20267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/15/2021] [Accepted: 04/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND/AIM Rapid glycolytic substrate-level phosphorylation (SLP) and accumulation of lactic acid are characteristics of diverse cancers. Recent advances in drug discovery have included the use of glycolytic inhibitors with mitochondrial targeting drugs to attempt to invoke an energy crisis in aggressive metabolically active chemo-resistant cancers. In this work, we examine the consequences of inhibiting mitochondrial oxidative phosphorylation (OXPHOS) with 1-methyl-4-phenylpyridinium (MPP+) in LS14T colon cancer cells containing a genetic double knock out (DKO) of lactic acid dehydrogenase (LDHA and LDHB). MATERIALS AND METHODS Several metabolic parameters were evaluated concomitant to whole transcriptomic (WT) mRNA, microRNA, and long intergenic non-coding RNAs using Affymetrix 2.1 human ST arrays. RESULTS MPP+ effectively blocked OXPHOS where a compensatory shift toward anaerobic SLP was only observed in the control vector (CV), and not observed in the LDH-A/B DKOs (lacking the ability to produce lactic acid). Despite this, there was an unexpected resilience to MPP+ in the latter in terms of energy, which displayed significantly higher resting baseline respiratory OXPHOS capacity relative to controls. At the transcriptome level, MPP+ invoked 1738 differential expressed genes (DEGs) out of 48,226; LDH-A/B DKO resulted in 855 DEGs while 349 DEGs were found to be overlapping in both groups versus respective controls, including loss of mitochondrial complex I (subunits 3 and 6), cell cycle transcripts and fluctuations in epigenetic chromatin remodeling systems. In terms of energy, the effects of MPP+ in the CV transcripts reflect the funneling of carbon intermediates toward glycolysis. The LDH-A/B DKO transcripts reflect a flow of carbons away from glycolysis toward the production of acetyl-CoA. CONCLUSION The findings from this study suggest a metabolic resilience to MPP+ in cancer cells devoid of LDH-A/B, explainable in-part by higher baseline OXPHOS respiratory ATP production, necessitating more toxin to suppress the electron transport chain.
Collapse
Affiliation(s)
- Nzinga Mack
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Elizabeth Mazzio
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Ramesh Badisa
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Karam F A Soliman
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A.
| |
Collapse
|
16
|
Li J, Wu G, Song W, Liu Y, Han Z, Shen Z, Li Y. Prophylactic Melatonin Treatment Ameliorated Propofol-Induced Cognitive Dysfunction in Aged Rats. Neurotox Res 2021; 39:227-239. [PMID: 33159663 DOI: 10.1007/s12640-020-00307-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/19/2022]
Abstract
Considering the fact that melatonin acts as protective agent in various cognitive impairment, we decided to explore the precise effect of pretreatment with melatonin on cognitive function, mitochondrial activity, apoptosis and synaptic integrity in aged rats anesthetized by propofol. We first randomly allocated the thirty Sprague Dawley rats into three groups: Control vehicle-treated group (Con), Propofol-treated group (Pro) and Melatonin + Propofol group (Mel + Pro). The Barnes maze, open field and contextual fear conditioning test were employed to evaluate spatial memory, exploratory behavior and general locomotor activity, and hippocampus-dependent learning and memory ability, respectively. Moreover, mitochondrial function (including reactive oxygen species, mitochondrial membrane potential and ATP levels) and apoptosis were detected in the regions of hippocampus (HIP) and prefrontal cortex (PFC). The results of behavioral tests suggested that melatonin improved propofol-induced memory impairment in aged rats. Melatonin mitigated mitochondrial dysfunction and decreased the apoptotic cell counts in the regions of HIP and PFC. Furthermore, prophylactic melatonin treatment also reversed the propofol-induced inactivation of PKA/CREB/BDNF signaling and synaptic dysfunction. On the whole, our results indicated that melatonin ameliorated the propofol-induced cognitive disorders via attenuating mitochondrial dysfunction, apoptosis, inactivation of PKA/CREB/BDNF signaling and synaptic dysfunction.
Collapse
Affiliation(s)
- Junhua Li
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Guiyun Wu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Wen Song
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yafang Liu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhixiao Han
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhiwen Shen
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yujuan Li
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
17
|
Zeng W, Xing Z, Tan M, Wu Y, Zhang C. Propofol regulates activated macrophages metabolism through inhibition of ROS-mediated GLUT1 expression. Inflamm Res 2021; 70:473-481. [PMID: 33751130 DOI: 10.1007/s00011-021-01449-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/02/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE Activated macrophages undergo a metabolic shift from oxidative phosphorylation (OXPHOS) to aerobic glycolysis, which plays a critical role in inflammation. Increasing evidence suggests the important role of propofol in the regulation of inflammatory response and metabolism, but the effect of propofol on the metabolic shift in macrophage, and the mechanisms involved remain unclear. METHODS The effect of propofol on the metabolic switch was analyzed by extracellular acidification rate and oxygen consumption rate assays. The effect of propofol on glycolysis was analyzed by lactate and glucose uptake assay. The mRNA, protein, cell surface levels of glucose transporter 1 (GLUT1) and the silencing of GLUT1 were employed to understand the effects of GLUT1-mediated metabolism by propofol. Finally, to understand the antioxidation of propofol on the regulation of metabolism, the reactive oxygen species (ROS) production and NADPH activity were performed. RESULTS We show that propofol can change the metabolic pathway switch from aerobic glycolysis to OXPHOS in LPS-activated macrophages. Moreover, propofol suppresses aerobic glycolysis via inhibited GLUT1-mediated glucose uptake. Furthermore, we show that propofol reduces ROS overproduction, which in turn inhibits GLUT1 expression. Finally, we find that propofol reduces ROS production via inhibits NADPH activity. CONCLUSION These findings shed light on the function and mechanism of propofol in the metabolic switch and highlight the importance of targeting metabolism by propofol in the clinical medication of inflammatory diseases.
Collapse
Affiliation(s)
- Wei Zeng
- Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Guangdong, 528400, China
| | - Zeting Xing
- Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Guangdong, 528400, China
| | - Meiyun Tan
- Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Guangdong, 528400, China
| | - Yanwen Wu
- Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Guangdong, 528400, China
| | - Chunyuan Zhang
- Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Guangdong, 528400, China.
| |
Collapse
|
18
|
A synthetic peptide rescues rat cortical neurons from anesthetic-induced cell death, perturbation of growth and synaptic assembly. Sci Rep 2021; 11:4567. [PMID: 33633281 PMCID: PMC7907385 DOI: 10.1038/s41598-021-84168-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Anesthetics are deemed necessary for all major surgical procedures. However, they have also been found to exert neurotoxic effects when tested on various experimental models, but the underlying mechanisms remain unknown. Earlier studies have implicated mitochondrial fragmentation as a potential target of anesthetic-induced toxicity, although clinical strategies to protect their structure and function remain sparse. Here, we sought to determine if preserving mitochondrial networks with a non-toxic, short-life synthetic peptide—P110, would protect cortical neurons against both inhalational and intravenous anesthetic-induced neurotoxicity. This study provides the first direct and comparative account of three key anesthetics (desflurane, propofol, and ketamine) when used under identical conditions, and demonstrates their impact on neonatal, rat cortical neuronal viability, neurite outgrowth and synaptic assembly. Furthermore, we discovered that inhibiting Fis1-mediated mitochondrial fission reverses anesthetic-induced aberrations in an agent-specific manner. This study underscores the importance of designing mitigation strategies invoking mitochondria-mediated protection from anesthetic-induced toxicity in both animals and humans.
Collapse
|
19
|
Mehta SD, Leavitt WL, Alex G, Saynhalath R, Kiss E. Endotracheal Intubation Using Alfentanil in a Pediatric Patient with a Mitochondrial Myopathy and Gastroparesis. J Pediatr Genet 2021; 10:53-56. [PMID: 33552640 DOI: 10.1055/s-0040-1702153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/15/2020] [Indexed: 10/25/2022]
Abstract
Children with mitochondrial disorders represent a subset of patients who require unique anesthetic considerations. Routinely administered medications for general anesthesia, such as propofol, have been shown to increase the risk of developing metabolic acidosis. In addition, both depolarizing and nondepolarizing neuromuscular blockers are contraindicated due to the risk of hyperkalemic cardiac arrest and worsening of preexisting muscle weakness, respectively. These limitations pose challenges while choosing appropriate medications for induction of general anesthesia, especially when the risk of aspiration is high. We present a novel case of using inhaled sevoflurane and intravenous alfentanil to facilitate intubation in a 4-year-old girl with a complex 1 mitochondrial disorder suffering from severe gastroparesis and esophageal dysmotility.
Collapse
Affiliation(s)
- Sonia D Mehta
- Division of Pediatric Anesthesiology, Department of Anesthesiology, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Wendy L Leavitt
- Division of Pediatric Anesthesiology, Department of Anesthesiology, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Gijo Alex
- Division of Pediatric Anesthesiology, Department of Anesthesiology, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Rita Saynhalath
- Division of Pediatric Anesthesiology, Department of Anesthesiology, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Edgar Kiss
- Division of Pediatric Anesthesiology, Department of Anesthesiology, UT Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
20
|
McCann MR, George De la Rosa MV, Rosania GR, Stringer KA. L-Carnitine and Acylcarnitines: Mitochondrial Biomarkers for Precision Medicine. Metabolites 2021; 11:51. [PMID: 33466750 PMCID: PMC7829830 DOI: 10.3390/metabo11010051] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
Biomarker discovery and implementation are at the forefront of the precision medicine movement. Modern advances in the field of metabolomics afford the opportunity to readily identify new metabolite biomarkers across a wide array of disciplines. Many of the metabolites are derived from or directly reflective of mitochondrial metabolism. L-carnitine and acylcarnitines are established mitochondrial biomarkers used to screen neonates for a series of genetic disorders affecting fatty acid oxidation, known as the inborn errors of metabolism. However, L-carnitine and acylcarnitines are not routinely measured beyond this screening, despite the growing evidence that shows their clinical utility outside of these disorders. Measurements of the carnitine pool have been used to identify the disease and prognosticate mortality among disorders such as diabetes, sepsis, cancer, and heart failure, as well as identify subjects experiencing adverse drug reactions from various medications like valproic acid, clofazimine, zidovudine, cisplatin, propofol, and cyclosporine. The aim of this review is to collect and interpret the literature evidence supporting the clinical biomarker application of L-carnitine and acylcarnitines. Further study of these metabolites could ultimately provide mechanistic insights that guide therapeutic decisions and elucidate new pharmacologic targets.
Collapse
Affiliation(s)
- Marc R. McCann
- The NMR Metabolomics Laboratory, Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Mery Vet George De la Rosa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, USA; (M.V.G.); (G.R.R.)
| | - Gus R. Rosania
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, USA; (M.V.G.); (G.R.R.)
| | - Kathleen A. Stringer
- The NMR Metabolomics Laboratory, Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA;
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
21
|
Zhang Q, Cai S, Guo L, Zhao G. Propofol induces mitochondrial-associated protein LRPPRC and protects mitochondria against hypoxia in cardiac cells. PLoS One 2020; 15:e0238857. [PMID: 32898195 PMCID: PMC7478836 DOI: 10.1371/journal.pone.0238857] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/25/2020] [Indexed: 12/26/2022] Open
Abstract
Background Hypoxia-induced oxidative stress is one of the main mechanisms of myocardial injury, which frequently results in cardiomyocyte death and precipitates life-threatening heart failure. Propofol (2,6-diisopropylphenol), which is used to sedate patients during surgery, was shown to strongly affect the regulation of physiological processes, including hypoxia-induced oxidative stress. However, the exact mechanism is still unclear. Methods Expression of LRPPRC, SLIRP, and Bcl-2 after propofol treatment was measured by RT-qPCR and western blot analyses. The effects of propofol under hypoxia were determine by assessing mitochondrial homeostasis and mitochondrial function, including the ATP level and mitochondrial mass. Autophagy/mitophagy was measured by detecting the presence of LC3B, and autophagosomes were observed by transmission microscopy Results Propofol treatment inhibited cleaved caspase-9 and caspase-3, indicating its inhibitory roles in mitochondrial-related apoptosis. Propofol treatment also transcriptionally activated LRPPRC, a mitochondrial-associated protein that exerts multiple functions by maintaining mitochondrial homeostasis, in a manner dependent on the presence of hypoxia-induced factor (HIF)-1α transcriptional activity in H9C2 and primary rat cardiomyocytes. LRPPRC induced by propofol maintained the mitochondrial membrane potential (MMP) and promoted mitochondrial function, including ATP synthesis and transcriptional activity. Furthermore, LRPPRC induced by propofol contributes, at least partially, to the inhibition of apoptotic cell death induced by hypoxia. Conclusion Taken together, our results indicate that LRPPRC may have a protective antioxidant effect by maintaining mitochondrial homoeostasis induced by propofol and provide new insight into the protective mechanism of propofol against oxidative stress.
Collapse
Affiliation(s)
- Qianlu Zhang
- Department of Anesthesiology, The First Affiliated Hospital of the University of South China, Hengyang, Hunan Province, P.R. China
| | - Shiwei Cai
- Cardiovascular Surgery, The First Affiliated Hospital of the University of South China, Hengyang, Hunan Province, P.R. China
| | - Liping Guo
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People’s Hospital, Qingyuan, Guangdong Province, P.R. China
| | - Guojun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People’s Hospital, Qingyuan, Guangdong Province, P.R. China
- * E-mail:
| |
Collapse
|
22
|
Effects of Acute Subdural Hematoma-Induced Brain Injury On Energy Metabolism in Peripheral Blood Mononuclear Cells. Shock 2020; 55:407-417. [PMID: 32826816 DOI: 10.1097/shk.0000000000001642] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT In activated immune cells, differentiation and function are determined by cell type-specific modifications of metabolic patterns. After traumatic brain injury both immune cell activation and suppression were reported. Therefore, we sought to explore immune cell energy metabolism in a long-term, resuscitated porcine model of acute subdural hematoma (ASDH)-induced acute brain injury devoid of impaired systemic hemodynamics and oxygen transport.Before and up to 50 h after induction of ASDH, peripheral blood mononuclear cells (PBMCs) were separated by density gradient centrifugation, and cell metabolism was analyzed using high-resolution respirometry for mitochondrial respiration and electron spin resonance for reactive oxygen species production. After incubation with stable isotope-labeled 1,2-13C2-glucose or 13C5-glutamine, distinct labeling patterns of intermediates of glycolysis or tricarboxylic acid (TCA) cycle and 13CO2 production were measured by gas chromatography-mass spectroscopy. Principal component analysis was followed by a varimax rotation on the covariance across all measured variables and all measured time points.After ASDH induction, average PBMC metabolic activity remained unaffected, possibly because strict adherence to intensive care unit guidelines limited trauma to ASDH induction without any change in parameters of systemic hemodynamics, oxygen transport, and whole-body metabolism. Despite decreased glycolytic activity fueling the TCA cycle, the principal component analysis indicated a cell type-specific activation pattern with biosynthetic and proliferative characteristics.
Collapse
|
23
|
Guntani A, Yoshiga R, Mii S. A case of suspected propofol infusion syndrome after abdominal aortic aneurysm surgery. Surg Case Rep 2020; 6:188. [PMID: 32737619 PMCID: PMC7393332 DOI: 10.1186/s40792-020-00946-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022] Open
Abstract
Background Propofol infusion syndrome (PRIS) is a rare but potentially lethal side effect during propofol administration. Case presentation The patient was scheduled for abdominal aortic aneurysm resection and reconstruction. Propofol used during sedation for ventilation after the surgery-induced rhabdomyolysis, heart failure, and renal failure. Discontinuation of propofol administration led to a dramatic improvement in the fatal symptoms, resulting in a diagnosis of PRIS. Conclusions We herein report a rare case of a PRIS during sedation in the intensive care unit after abdominal aortic aneurysm surgery. Physicians using propofol should therefore be aware of the potential risk of PRIS.
Collapse
Affiliation(s)
- Atsushi Guntani
- Department of Vascular Surgery, Saiseikai Yahata General Hospital, 5-9-27 Haruno-machi, Yahatahigashi-ku, Kitakyushu, 805-8527, Japan.
| | | | - Shinsuke Mii
- Department of Vascular Surgery, Saiseikai Yahata General Hospital, 5-9-27 Haruno-machi, Yahatahigashi-ku, Kitakyushu, 805-8527, Japan
| |
Collapse
|
24
|
Propofol induces the elevation of intracellular calcium via morphological changes in intracellular organelles, including the endoplasmic reticulum and mitochondria. Eur J Pharmacol 2020; 884:173303. [PMID: 32681942 DOI: 10.1016/j.ejphar.2020.173303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/20/2020] [Accepted: 06/19/2020] [Indexed: 12/27/2022]
Abstract
Propofol, most frequently used as a general anesthetic due to its versatility and short-acting characteristics, is thought to exert its anesthetic actions via GABAA receptors; however, the precise mechanisms of its adverse action including angialgia remain unclear. We examined the propofol-induced elevation of intracellular calcium and morphological changes in intracellular organelles using SHSY-5Y neuroblastoma cells, COS-7 cells, HEK293 cells, and HUVECs loaded with fluorescent dyes for live imaging. Although propofol (>50 μM) increased intracellular calcium in a dose-dependent manner in these cells, it was not influenced by the elimination of extracellular calcium. The calcium elevation was abolished when intracellular or intraendoplasmic reticulum (ER) calcium was depleted by BAPTA-AM or thapsigargin, respectively, suggesting that calcium was mobilized from the ER. Studies using U-73122, xestospongin C, and dantrolene revealed that propofol-induced calcium elevation was not mediated by G-protein coupled receptors, IP3 receptors, or ryanodine receptors. We performed live imaging of the ER, mitochondria and Golgi apparatus during propofol stimulation using fluorescent dyes. Concomitant with the calcium elevation, the structure of the ER and mitochondria was fragmented and aggregated, and these changes were not reversed during the observation period, suggesting that propofol-induced calcium elevation occurs due to calcium leakage from these organelles. Although the concentration of propofol used in this experiment was greater than that used clinically (30 μM), it is possible that the concentration exceeds 30 μM at the site where propofol is injected, leading the idea that these phenomena might relate to the various propofol-induced adverse effects including angialgia.
Collapse
|
25
|
A Metabolic Mechanism for Anaesthetic Suppression of Cortical Synaptic Function in Mouse Brain Slices-A Pilot Investigation. Int J Mol Sci 2020; 21:ijms21134703. [PMID: 32630300 PMCID: PMC7370287 DOI: 10.3390/ijms21134703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Regulation of synaptically located ionotropic receptors is thought to be the main mechanism by which anaesthetics cause unconsciousness. An alternative explanation, which has received much less attention, is that of primary anaesthetic disruption of brain metabolism via suppression of mitochondrial proteins. In this pilot study in mouse cortical slices, we investigated the effect of disrupting cellular metabolism on tissue oxygen handling and cortical population seizure-like event (SLE) activity, using the mitochondrial complex I inhibitor rotenone, and compared this to the effects of the general anaesthetics sevoflurane, propofol and ketamine. Rotenone caused an increase in tissue oxygen (98 mmHg to 157 mmHg (p < 0.01)) before any measurable change in SLE activity. Thereafter, tissue oxygen continued to increase and was accompanied by a significant and prolonged reduction in SLE root mean square (RMS) activity (baseline RMS of 1.7 to 0.7 µV, p < 0.001) and SLE frequency (baseline 4.2 to 0.4 events/min, p = 0.001). This temporal sequence of effects was replicated by all three anaesthetic drugs. In conclusion, anaesthetics with differing synaptic receptor mechanisms all effect changes in tissue oxygen handling and cortical network activity, consistent with a common inhibitory effect on mitochondrial function. The temporal sequence suggests that the observed synaptic depression—as seen in anaesthesia—may be secondary to a reduction in cellular metabolic capacity.
Collapse
|
26
|
Yu C, Du F, Zhang C, Li Y, Liao C, He L, Cheng X, Zhang X. Salmonella enterica serovar Typhimurium sseK3 induces apoptosis and enhances glycolysis in macrophages. BMC Microbiol 2020; 20:151. [PMID: 32517648 PMCID: PMC7282050 DOI: 10.1186/s12866-020-01838-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 06/02/2020] [Indexed: 02/07/2023] Open
Abstract
Background Salmonella enterica serovar Typhimurium (S. Typhimurium) is an important infectious disease pathogen that can survive and replicate in macrophages. Glycolysis is essential for immune responses against S. Typhimurium infection in macrophages, and is also associated with apoptosis. S. Typhimurium secreted effector K3 (SseK3) was recently identified as a novel translated and secreted protein. However, there is no study about the role of sseK3 in the relationship between apoptosis and glycolysis in cells infected with S. Typhimurium. It is unclear whether this protein exerts a significant role in the progress of apoptosis and glycolysis in S. Typhimurium-infected macrophages. Results Macrophages were infected with S. Typhimurium SL1344 wild-type (WT), ΔsseK3 mutant or sseK3-complemented strain, and the effects of sseK3 on apoptosis and glycolysis were determined. The adherence and invasion in the ΔsseK3 mutant group were similar to that in the WT and sseK3-complemented groups, indicating that SseK3 was not essential for the adherence and invasion of S. Typhimurium in macrophages. However, the percentage of apoptosis in the ΔsseK3 mutant group was much lower than that in the WT and sseK3-complemented groups. Caspase-3, caspase-8, and caspase-9 enzyme activity in the ΔsseK3 mutant group were significantly lower than in the WT group and sseK3-complemented groups, indicating that sseK3 could improve the caspase-3, caspase-8, and caspase-9 enzyme activity. We also found that there were no significant differences in pyruvic acid levels between the three groups, but the lactic acid level in the ΔsseK3 mutant group was much lower than that in the WT and sseK3-complemented groups. The ATP levels in the ΔsseK3 mutant group were remarkably higher than those in the WT and sseK3-complemented groups. These indicated that the sseK3 enhanced the level of glycolysis in macrophages infected by S. Typhimurium. Conclusions S. Typhimurium sseK3 is likely involved in promoting macrophage apoptosis and modulating glycolysis in macrophages. Our results could improve our understanding of the relationship between apoptosis and glycolysis in macrophages induced by S. Typhimurium sseK3.
Collapse
Affiliation(s)
- Chuan Yu
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, Henan, China.,Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang, 471023, Henan, China.,Luoyang Polytechnic, 6 Airport Road, Luoyang, 471023, Henan, China
| | - Fuyu Du
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, Henan, China.,Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang, 471023, Henan, China
| | - Chunjie Zhang
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, Henan, China. .,Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang, 471023, Henan, China.
| | - Yinju Li
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, Henan, China.,Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang, 471023, Henan, China
| | - Chengshui Liao
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, Henan, China.,Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang, 471023, Henan, China
| | - Lei He
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, Henan, China.,Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang, 471023, Henan, China
| | - Xiangchao Cheng
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, Henan, China.,Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang, 471023, Henan, China.,Luoyang Polytechnic, 6 Airport Road, Luoyang, 471023, Henan, China
| | - Xiaojie Zhang
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, Henan, China.,Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang, 471023, Henan, China
| |
Collapse
|
27
|
Ji X, Guo Y, Qiu Q, Wang Z, Wang Y, Ji J, Sun Q, Cai Y, Zhou G. [Molecular mechanism underlying the inhibitory effect of propofol on lipopolysaccharide-induced pyroptosis of mouse bone marrow-derived macrophages]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:525-530. [PMID: 32895145 DOI: 10.12122/j.issn.1673-4254.2020.04.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the molecular mechanism underlying the inhibitory effect of propofol on pyroptosis of macrophages. METHODS Macrophages derived from bone marrow were extracted and divided into three groups: control group, LPS+ATP group and propofol+LPS+ATP group. The control group was not given any treatment; LPS+ATP group was given LPS 1 μg/mL stimulation for 4 h, then ATP 4 mM stimulation for 1 h; Propofol+LPS+ATP group was given propofol+LPS 1 μg/mL stimulation for 4 h, then ATP stimulation for 1 h. After treatment, the supernatant and cells of cell culture were collected. the cell activity was detected by CCK8 and flow cytometry. The inflammatory cytokines IL-1βand IL-18 were detected by Elisa. Western blot was used to detect the expression of caspase-1 protein and TLR4 on cell membran Immunohistochemical fluorescence was used to detect apoptosis of cells. RESULTS LPS+ATP significantly decreased the viability of the macrophages and increased the cellular production of IL-1β and IL-18, activation of caspase-1 protein and the expression of TLR-4 on the cell membrane (P < 0.05). Treatment with propofol obviously reversed the changes induced by LPS+ATP. CONCLUSIONS LPS+ATP can induce pyroptosis of mouse bone marrow-derived macrophages, and propofol effectively inhibits such cell death, suggesting that propofol anesthesia is beneficial during operation and helps to regulate the immune function of in patients with sepsis.
Collapse
Affiliation(s)
- Xuexia Ji
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yuanbo Guo
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qianqi Qiu
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| | - Zhipeng Wang
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yan Wang
- Department of Science and Education, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jinquan Ji
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qiang Sun
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yujing Cai
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Guobin Zhou
- Department of Anesthesiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| |
Collapse
|
28
|
Suspected propofol infusion syndrome during normal targeted propofol concentration. J Anesth 2020; 34:619-623. [PMID: 32222909 DOI: 10.1007/s00540-020-02773-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 03/21/2020] [Indexed: 10/24/2022]
Abstract
To this day, the pathophysiology and risk factors of propofol infusion syndrome (PRIS) remain unknown. Moreover, there is no widely accepted definition of PRIS, even though it is a potentially fatal condition. While many suspected cases of PRIS have been reported in both pediatric and adult populations, the actual propofol plasma concentration (Cp) has never been clarified. In this clinical report, we described the first suspected PRIS case in which the propofol Cp was measured 25 min after 226 min of propofol infusion (7.2 µg/mL), which was 12 times higher than the predicted value (0.6 µg/mL). In the presented case, we observed gradually progressive uncontrollable hypercapnia and tachycardia, followed by severe lactic acidosis during surgical anesthesia based on the target-controlled infusion of propofol. Levels of liver enzymes were slightly elevated which suggests little or no liver damage though propofol is mainly metabolized by the liver. Meanwhile, renal impairment, a common secondary feature of PRIS, occurred concomitantly when hypercapnia and metabolic acidosis were manifested. In this case, low or delayed propofol clearance might have been a triggering factor causing severe lactic acidosis.
Collapse
|
29
|
Ohishi T, Abe H, Sakashita C, Saqib U, Baig MS, Ohba SI, Inoue H, Watanabe T, Shibasaki M, Kawada M. Inhibition of mitochondria ATP synthase suppresses prostate cancer growth through reduced insulin-like growth factor-1 secretion by prostate stromal cells. Int J Cancer 2020; 146:3474-3484. [PMID: 32144767 DOI: 10.1002/ijc.32959] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/18/2020] [Accepted: 03/02/2020] [Indexed: 01/07/2023]
Abstract
Modulation of prostate stromal cells (PrSCs) within tumor tissues is gaining attention for the treatment of solid tumors. Using our original in vitro coculture system, we previously reported that leucinostatin (LCS)-A, a peptide mycotoxin, inhibited prostate cancer DU-145 cell growth through reduction of insulin-like growth factor 1 (IGF-I) expression in PrSCs. To further obtain additional bioactive compounds from LCS-A, we designed and synthesized a series of LCS-A derivatives as compounds that target PrSCs. Among the synthesized LCS-A derivatives, LCS-7 reduced IGF-I expression in PrSCs with lower toxicity to PrSCs and mice than LCS-A. As LCS-A has been suggested to interact with mitochondrial adenosine triphosphate (ATP) synthase, a docking study was performed to elucidate the mechanism of reduced IGF-I expression in the PrSCs. As expected, LCS-A and LCS-7 directly interacted with mitochondrial ATP synthase, and like LCS-A and LCS-7, other mitochondrial ATP synthase inhibitors also reduced the expression of IGF-I by PrSCs. Furthermore, LCS-A and LCS-7 significantly decreased the growth of mouse xenograft tumors. Based on these data, we propose that the mitochondrial ATP synthases-IGF-I axis of PrSCs plays a critical role on cancer cell growth and inhibition could be a potential anticancer target for prostate cancer.
Collapse
Affiliation(s)
- Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu-shi, Japan
| | - Hikaru Abe
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Synthetic Organic Chemistry, Microbial Chemistry Research Foundation, Tokyo, Japan
| | - Chiharu Sakashita
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Synthetic Organic Chemistry, Microbial Chemistry Research Foundation, Tokyo, Japan
| | - Uzma Saqib
- Discipline of Chemistry, School of Basic Sciences, Indian Institute of Technology (IIT), Indore, Madhya Pradesh, India
| | - Mirza S Baig
- Centre for Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology (IIT), Indore, Madhya Pradesh, India
| | - Shun-Ichi Ohba
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu-shi, Japan
| | - Hiroyuki Inoue
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu-shi, Japan
| | - Takumi Watanabe
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Synthetic Organic Chemistry, Microbial Chemistry Research Foundation, Tokyo, Japan
| | - Masakatsu Shibasaki
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Synthetic Organic Chemistry, Microbial Chemistry Research Foundation, Tokyo, Japan
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu-shi, Japan.,Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, Tokyo, Japan
| |
Collapse
|
30
|
Subirà González A, Fernández Morales M, Sánchez Royo E, Boliart de San Félix Y, Vila Lolo C. Propofol infusion syndrome; are high doses always required? ACTA ACUST UNITED AC 2020; 67:163-166. [PMID: 32111374 DOI: 10.1016/j.redar.2020.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/17/2019] [Accepted: 01/03/2020] [Indexed: 10/24/2022]
Abstract
Propofol infusion syndrome is a rare condition that mainly affects critically ill patients who receive high doses of this hypnotic for a long time. We describe the case of a patient who presented hepatotoxicity in the immediate postoperative period of two surgeries in which she had received conventional doses of propofol for a short period of time. After studying the patient and monitoring her evolution, we arrived at a differential diagnosis of propofol infusion syndrome due to increased susceptibility. This syndrome should be considered in patients presenting hepatotoxicity in the immediate postoperative period, even when low doses of propofol have been administered.
Collapse
Affiliation(s)
- A Subirà González
- Departamento de Anestesiología, Reanimación y Tratamiento del Dolor, Hospital Universitario Dexeus, Barcelona, España.
| | - M Fernández Morales
- Departamento de Anestesiología, Reanimación y Tratamiento del Dolor, Hospital Universitario Dexeus, Barcelona, España
| | - E Sánchez Royo
- Departamento de Anestesiología, Reanimación y Tratamiento del Dolor, Hospital Universitario Dexeus, Barcelona, España
| | - Y Boliart de San Félix
- Departamento de Anestesiología, Reanimación y Tratamiento del Dolor, Hospital Universitario Dexeus, Barcelona, España
| | - C Vila Lolo
- Departamento de Digestivo, Hospital Universitario Dexeus, Barcelona, España
| |
Collapse
|
31
|
Hirota K. Basic Biology of Hypoxic Responses Mediated by the Transcription Factor HIFs and its Implication for Medicine. Biomedicines 2020; 8:biomedicines8020032. [PMID: 32069878 PMCID: PMC7168341 DOI: 10.3390/biomedicines8020032] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 02/08/2020] [Accepted: 02/12/2020] [Indexed: 12/19/2022] Open
Abstract
Oxygen (O2) is essential for human life. Molecular oxygen is vital for the production of adenosine triphosphate (ATP) in human cells. O2 deficiency leads to a reduction in the energy levels that are required to maintain biological functions. O2 acts as the final acceptor of electrons during oxidative phosphorylation, a series of ATP synthesis reactions that occur in conjunction with the electron transport system in mitochondria. Persistent O2 deficiency may cause death due to malfunctioning biological processes. The above account summarizes the classic view of oxygen. However, this classic view has been reviewed over the last two decades. Although O2 is essential for life, higher organisms such as mammals are unable to biosynthesize molecular O2 in the body. Because the multiple organs of higher organisms are constantly exposed to the risk of “O2 deficiency,” living organisms have evolved elaborate strategies to respond to hypoxia. In this review, I will describe the system that governs oxygen homeostasis in the living body from the point-of-view of the transcription factor hypoxia-inducible factor (HIF).
Collapse
Affiliation(s)
- Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
32
|
He L, Wang X, Zheng S. Inhibition of the electron transport chain in propofol induced neurotoxicity in zebrafish embryos. Neurotoxicol Teratol 2020; 78:106856. [PMID: 31923456 DOI: 10.1016/j.ntt.2020.106856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 01/20/2023]
Abstract
Fetal and neonatal exposure to propofol can lead to neuronal death and long-term neurobehavioral deficiencies in both rodents and nonhuman primates. Zebrafish embryo, which is fertilized ex-utero, has provided us a new model species to study the effects of general anesthetics on developing brain. Inhibited electron transport chain leads to mitochondrial dysfunction and insufficient energy production. The aim of this study was to dissect the role of electron transport chain in propofol-induced neurotoxicity. 6 h post fertilization (hpf) zebrafish embryos were exposed to control or 1, 2 or 4 μg/ml propofol until 48hpf. Acridine orange staining was used to assess cell apoptosis in the brain of zebrafish embryos. The activity of mitochondrial electron transport chain complex was assessed using colorimetric method. Expression of key subunit of cytochrome c oxidase was assessed by western blot and transcription level of cox4i1 was assessed by quantitative real time-PCR. The mitochondrial membrane potential and ATP content were assessed. Exposure to 1, 2 and 4 μg/ml propofol induced significant increases in cell apoptosis in the brain of zebrafish embryos in a dose-dependent manner and led to significant decreases in electron transport chain complex IV activity from (0.161 ± 0.023)μmol/mg/min in blank control-treated group to (0.096 ± 0.015)μmol/mg/min, (0.083 ± 0.013)μmol/mg/min and (0.045 ± 0.014)μmol/mg/min respectively, accompanied by decreased expression of key regulatory subunit of cytochrome c oxidase-subunit IV and decreased transcription level of cox4i1. Propofol exposure also decreased the mitochondrial membrane potential and ATP content. Our findings demonstrate that inhibition of the electron transport chain is involved in the mechanisms by which propofol induces neurotoxicity in the developing brain.
Collapse
Affiliation(s)
- Lin He
- Department of Anesthesiology, Children's Hospital of Fudan University, Shanghai, China
| | - Xuan Wang
- Department of Anesthesiology, Children's Hospital of Fudan University, Shanghai, China
| | - Shan Zheng
- Department of Surgery, Children's Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
33
|
Pharmacological polysulfide suppresses glucose-stimulated insulin secretion in an ATP-sensitive potassium channel-dependent manner. Sci Rep 2019; 9:19377. [PMID: 31852936 PMCID: PMC6920347 DOI: 10.1038/s41598-019-55848-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous gaseous transmitter synthesized in various cell types. It is well established that H2S functions in many physiological processes, including the relaxation of vascular smooth muscle, mediation of neurotransmission, regulation of inflammation, and modulation of insulin signaling. In recent years, it has been revealed that polysulfides, substances with a varying number of sulfur atoms (H2Sn), are generated endogenously from H2S in the presence of oxygen. A series of studies describes that sulfane sulfur has the unique ability to bind reversibly to other sulfur atoms to form hydropersulfides and polysulfides, and that polysulfides activate ion channels and promote calcium influx. Furthermore, polysulfides regulate tumor suppressor activity, promote the activation of transcription factors targeting antioxidant genes and regulate blood pressure by vascular smooth muscle relaxation. Insulin secretion from pancreatic β cells plays a critical role in response to increased blood glucose concentration. H2S has emerged as an important regulator of glycemic control and exhibits characteristic regulation of glucose homeostasis. However, the effects of polysulfides on glucose-stimulated insulin secretion (GSIS) are largely unknown. In this study, we demonstrated that pharmacological polysulfide salts including Na2S2, Na2S3, and Na2S4 considerably inhibit GSIS in mouse and rat pancreatic β-cell-derived MIN6 and INS-1 cell lines, and that the effect is dependent on the activation of ATP-sensitive potassium channels. In addition, we demonstrated that a mixture of Na2S and diethylamine NONOate inhibits GSIS in a similar way to the pharmacological administration of polysulfide salts.
Collapse
|
34
|
Kusunoki M, Hayashi M, Shoji T, Uba T, Tanaka H, Sumi C, Matsuo Y, Hirota K. Propofol inhibits stromatoxin-1-sensitive voltage-dependent K + channels in pancreatic β-cells and enhances insulin secretion. PeerJ 2019; 7:e8157. [PMID: 31824770 PMCID: PMC6894434 DOI: 10.7717/peerj.8157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/04/2019] [Indexed: 12/31/2022] Open
Abstract
Background Proper glycemic control is an important goal of critical care medicine, including perioperative patient care that can influence patients’ prognosis. Insulin secretion from pancreatic β-cells is generally assumed to play a critical role in glycemic control in response to an elevated blood glucose concentration. Many animal and human studies have demonstrated that perioperative drugs, including volatile anesthetics, have an impact on glucose-stimulated insulin secretion (GSIS). However, the effects of the intravenous anesthetic propofol on glucose metabolism and insulin sensitivity are largely unknown at present. Methods The effect of propofol on insulin secretion under low glucose or high glucose was examined in mouse MIN6 cells, rat INS-1 cells, and mouse pancreatic β-cells/islets. Cellular oxygen or energy metabolism was measured by Extracellular Flux Analyzer. Expression of glucose transporter 2 (GLUT2), potassium channels, and insulin mRNA was assessed by qRT-PCR. Protein expression of voltage-dependent potassium channels (Kv2) was also assessed by immunoblot. Propofol’s effects on potassium channels including stromatoxin-1-sensitive Kv channels and cellular oxygen and energy metabolisms were also examined. Results We showed that propofol, at clinically relevant doses, facilitates insulin secretion under low glucose conditions and GSIS in MIN6, INS-1 cells, and pancreatic β-cells/islets. Propofol did not affect intracellular ATP or ADP concentrations and cellular oxygen or energy metabolism. The mRNA expression of GLUT2 and channels including the voltage-dependent calcium channels Cav1.2, Kir6.2, and SUR1 subunit of KATP, and Kv2 were not affected by glucose or propofol. Finally, we demonstrated that propofol specifically blocks Kv currents in β-cells, resulting in insulin secretion in the presence of glucose. Conclusions Our data support the hypothesis that glucose induces membrane depolarization at the distal site, leading to KATP channel closure, and that the closure of Kv channels by propofol depolarization in β-cells enhances Ca2+ entry, leading to insulin secretion. Because its activity is dependent on GSIS, propofol and its derivatives are potential compounds that enhance and initiate β-cell electrical activity.
Collapse
Affiliation(s)
- Munenori Kusunoki
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Mikio Hayashi
- Department of Cell Physiology, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Tomohiro Shoji
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Takeo Uba
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Hiromasa Tanaka
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Chisato Sumi
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Yoshiyuki Matsuo
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
35
|
Urban T, Waldauf P, Krajčová A, Jiroutková K, Halačová M, Džupa V, Janoušek L, Pokorná E, Duška F. Kinetic characteristics of propofol-induced inhibition of electron-transfer chain and fatty acid oxidation in human and rodent skeletal and cardiac muscles. PLoS One 2019; 14:e0217254. [PMID: 31584947 PMCID: PMC6777831 DOI: 10.1371/journal.pone.0217254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 09/20/2019] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Propofol causes a profound inhibition of fatty acid oxidation and reduces spare electron transfer chain capacity in a range of human and rodent cells and tissues-a feature that might be related to the pathogenesis of Propofol Infusion Syndrome. We aimed to explore the mechanism of propofol-induced alteration of bioenergetic pathways by describing its kinetic characteristics. METHODS We obtained samples of skeletal and cardiac muscle from Wistar rat (n = 3) and human subjects: vastus lateralis from hip surgery patients (n = 11) and myocardium from brain-dead organ donors (n = 10). We assessed mitochondrial functional indices using standard SUIT protocol and high resolution respirometry in fresh tissue homogenates with or without short-term exposure to a range of propofol concentration (2.5-100 μg/ml). After finding concentrations of propofol causing partial inhibition of a particular pathways, we used that concentration to construct kinetic curves by plotting oxygen flux against substrate concentration during its stepwise titration in the presence or absence of propofol. By spectrophotometry we also measured the influence of the same propofol concentrations on the activity of isolated respiratory complexes. RESULTS We found that human muscle and cardiac tissues are more sensitive to propofol-mediated inhibition of bioenergetic pathways than rat's tissue. In human homogenates, palmitoyl carnitine-driven respiration was inhibited at much lower concentrations of propofol than that required for a reduction of electron transfer chain capacity, suggesting FAO inhibition mechanism different from downstream limitation or carnitine-palmitoyl transferase-1 inhibition. Inhibition of Complex I was characterised by more marked reduction of Vmax, in keeping with non-competitive nature of the inhibition and the pattern was similar to the inhibition of Complex II or electron transfer chain capacity. There was neither inhibition of Complex IV nor increased leak through inner mitochondrial membrane with up to 100 μg/ml of propofol. If measured in isolation by spectrophotometry, propofol 10 μg/ml did not affect the activity of any respiratory complexes. CONCLUSION In human skeletal and heart muscle homogenates, propofol in concentrations that are achieved in propofol-anaesthetized patients, causes a direct inhibition of fatty acid oxidation, in addition to inhibiting flux of electrons through inner mitochondrial membrane. The inhibition is more marked in human as compared to rodent tissues.
Collapse
Affiliation(s)
- Tomáš Urban
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Petr Waldauf
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Adéla Krajčová
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Kateřina Jiroutková
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Milada Halačová
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Valér Džupa
- Department of Orthopaedics and Traumatology, Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Libor Janoušek
- Transplantation Surgery Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Pokorná
- Department of Organ Recovery and Transplantation Databases, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - František Duška
- OXYLAB – Mitochondrial Physiology Lab: Charles University, 3 Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| |
Collapse
|
36
|
Murakami Y, Ueki R, Tachikawa T, Hirose M. The Basic Study of the Mechanism of Propofol-Related Infusion Syndrome Using a Murine Skeletal Muscle Injury Model. Anesth Pain Med 2019; 9:e89417. [PMID: 31497518 PMCID: PMC6712282 DOI: 10.5812/aapm.89417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/05/2019] [Accepted: 04/07/2019] [Indexed: 12/12/2022] Open
Abstract
Background The pathophysiological mechanism of propofol-related infusion syndrome (PRIS) is believed to be due to the injury to the mitochondrial electron transport chain and the resultant metabolic disorders that are caused by both propofol agents and the lipid solvent. However, the mechanisms and causative factors of PRIS have not been fully elucidated. Objectives The aim of this study was to evaluate the possibility of a research model using the culture of differentiated C2C12 cells for fundamental research of PRIS. Methods First, differentiated C2C12 cells were cultured accompanied by several concentrations of chemical reagents of 2,6-diisopropylphenol (2,6 DIP) or dimethyl sulfoxide (DMSO) for 60 hours and the cell death rate was examined by trypan blue staining. Second, The cells were incubated with a commercially available propofol reagent or lipid reagent for 48 hours. The supernatant fluid of the cell culture medium was gathered and the numbers of floating cells were measured by cell counter. To investigate the mitochondrial disorder by the propofol preparation, JC-1, an experiment using fluorescent reagent, was performed for the 48 hours with 100 µg/mL propofol incubation. Results The rate of cell death was increased with elevating concentrations both of chemical reagents of 2,6 DIP group and dimethyl sulfoxide group. The rates of cell death were significantly higher in the 2,6 DIP group than DMSO group. The numbers of floating cells were increased with elevating concentrations both commercially available propofol reagent and lipid reagent groups. The decreased red/green fluorescence ratio by JC-1 staining in the propofol 100µg/mL group proved an attenuated mitochondrial membrane potential. Conclusions The dose-dependent cell damage induced by the propofol reagents and a lipid solvent may provide a proposed model as a basic experimental model for further investigations into PRIS.
Collapse
Affiliation(s)
- Yuryo Murakami
- Department of Anesthesiology and Pain Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Ryusuke Ueki
- Department of Anesthesiology and Pain Medicine, Hyogo College of Medicine, Nishinomiya, Japan
- Corresponding Author: MD, Ph.D. Department of Anesthesiology and Pain Medicine, Hyogo College of Medicine, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo 663-8501, Japan. Tel: 81-798456392, Fax: 81-798456393,
| | - Taihei Tachikawa
- Department of Anesthesiology, Meiwa Hospital, Nishinomiya, Japan
| | - Munetaka Hirose
- Department of Anesthesiology and Pain Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
37
|
Qin J, Li Y, Wang K. Propofol induces impairment of mitochondrial biogenesis through inhibiting the expression of peroxisome proliferator-activated receptor-γ coactivator-1α. J Cell Biochem 2019; 120:18288-18297. [PMID: 31190345 DOI: 10.1002/jcb.29138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 12/28/2018] [Accepted: 01/09/2019] [Indexed: 11/07/2022]
Abstract
Propofol is a commonly used general anesthetic in patient care. Recent studies have shown that propofol has neurological side effects especially in young children, which raises a concern regarding the safety of its use. We explored the effects of the molecular mechanism of propofol on neuronal mitochondrial function in SH-SY5Y cells. Our results demonstrate that clinically relevant doses of propofol reduce the expression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) in a dose- and time-dependent manner. At a concentration of 2%, propofol suppresses the mitochondrial regulator nuclear respiratory factor 1 and mitochondrial transcription factor A and impairs neuronal mitochondrial biogenesis. These impairments involve reduction of mitochondrial mass and reduction of the ratio of mitochondrial to nuclear DNA as well as reduction of cytochrome C oxidase activity. Propofol treatment reduces intracellular adenosine triphosphate (ATP) production, the mitochondrial respiratory rate, and increases mitochondrial reactive oxygen species production, implying that it disturbs neuronal mitochondrial function. Overexpression of PGC-1α rescued propofol-induced reduced mitochondrial mass, ATP production, and respiratory rate, indicating that PGC-1α is the mediator of the effect of propofol on mitochondrial function. Finally, we demonstrate that propofol suppresses PGC-1α by inhibiting cAMP-response element binding protein (CREB) activation and promoting PKA RI expression, and the addition of cyclic adenosine monophosphate rescues propofol-mediated reduced PGC-1α. In conclusion, PGC-1α is the central mediator of propofol-induced impairment of mitochondrial biogenesis and neuronal mitochondrial dysfunction. Our study demonstrates the molecular mechanism behind propofol-induced neurotoxicity and provides valuable information regarding its side effects in clinical practice.
Collapse
Affiliation(s)
- Jing Qin
- Department of Anesthesiology, Linyi Central Hospital, Linyi, Shandong, China
| | - Yizhen Li
- Department of Anesthesiology, Weifang People's Hospital, Weifang, Shandong, China
| | - Kai Wang
- Department of Breast Surgery, Weifang People's Hospital, Weifang, Shandong, China
| |
Collapse
|
38
|
Liu B, Bai W, Ou G, Zhang J. Cdh1-Mediated Metabolic Switch from Pentose Phosphate Pathway to Glycolysis Contributes to Sevoflurane-Induced Neuronal Apoptosis in Developing Brain. ACS Chem Neurosci 2019; 10:2332-2344. [PMID: 30741526 DOI: 10.1021/acschemneuro.8b00644] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cdh1 is a regulatory subunit of the anaphase promoting complex/cyclosome (APC/C), known to be involved in regulating neuronal survival. The role of Cdh1 in volatile anesthetics-induced neuronal apoptosis in the developing brain is unknown. In this study, we used postnatal day 7 (P7) and day 21 (P21) mice exposed to 2.3% sevoflurane for 6 h to investigate at which age and duration of exposure sevoflurane affects the expression of Cdh1 and glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) and that of the pentose phosphate pathway (PPP) enzyme, glucose-6-phosphate dehydrogenase (G6PD). Furthermore, we tested whether the cyclin-dependent kinases (cdks) inhibitor roscovatine could counteract the effects caused by exposure to sevoflurane. Finally, we applied the glycolysis inhibitor 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3-PO), G6PD inhibitor dehydroepiandrosterone (DHEA), and exogenous reduced glutathione to examine the contribution of the glycolysis pathway and PPP to sevoflurane-induced neuroapoptosis. We found that prolonged sevoflurane anesthesia significantly reduces the Cdh1 level in P7 mice compared to in the P21 ones; moreover, the decrease in Cdh1 level results in a switch in glucose metabolism from the PPP to neuronal glycolysis. This leads to an imbalance between reactive oxygen species production and reduced glutathione level in the developing brain, which is more susceptible to oxidative stress. As a result, sevoflurane induces neuroapoptosis through Cdh1-mediated glucose metabolism reprogramming. Our study demonstrates a critical role of Cdh1 in sevoflurane-induced neuroapoptosis by shifting PPP to the glycolytic pathway in the developing brain. These findings suggest that Cdh1 may be a novel target for preventing volatile anesthetics-induced neurotoxicity and memory impairment.
Collapse
Affiliation(s)
- Bin Liu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China 200040
| | - Wenjie Bai
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China 200040
| | - Guoyao Ou
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China 200040
| | - Jun Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China 200040
| |
Collapse
|
39
|
Cancerous phenotypes associated with hypoxia-inducible factors are not influenced by the volatile anesthetic isoflurane in renal cell carcinoma. PLoS One 2019; 14:e0215072. [PMID: 30986231 PMCID: PMC6464189 DOI: 10.1371/journal.pone.0215072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/26/2019] [Indexed: 12/22/2022] Open
Abstract
The possibility that anesthesia during cancer surgery may affect cancer recurrence, metastasis, and patient prognosis has become one of the most important topics of interest in cancer treatment. For example, the volatile anesthetic isoflurane was reported in several studies to induce hypoxia-inducible factors, and thereby enhance malignant phenotypes in vitro. Indeed, these transcription factors are considered critical regulators of cancer-related hallmarks, including “sustained proliferative signaling, evasion of growth suppressors, resistance to cell death, replicative immortality, angiogenesis, invasion, and metastasis.” This study aimed to investigate the impact of isoflurane on the growth and migration of derivatives of the renal cell line RCC4. We indicated that isoflurane treatment did not positively influence cancer cell phenotypes, and that hypoxia-inducible factors (HIFs) maintain hallmark cancer cell phenotypes including gene expressions signature, metabolism, cell proliferation and cell motility. The present results indicate that HIF activity is not influenced by the volatile anesthetic isoflurane.
Collapse
|
40
|
Finley J. Cellular stress and AMPK links metformin and diverse compounds with accelerated emergence from anesthesia and potential recovery from disorders of consciousness. Med Hypotheses 2019; 124:42-52. [PMID: 30798915 DOI: 10.1016/j.mehy.2019.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/19/2019] [Indexed: 01/23/2023]
Abstract
The neural correlates of consciousness and the mechanisms by which general anesthesia (GA) modulate such correlates to induce loss of consciousness (LOC) has been described as one of the biggest mysteries of modern medicine. Several cellular targets and neural circuits have been identified that play a critical role in LOC induced by GA, including the GABAA receptor and ascending arousal nuclei located in the basal forebrain, hypothalamus, and brain stem. General anesthetics (GAs) including propofol and inhalational agents induce LOC in part by potentiating chloride influx through the GABAA receptor, leading to neural inhibition and LOC. Interestingly, nearly all GAs used clinically may also induce paradoxical excitation, a phenomenon in which GAs promote neuronal excitation at low doses before inducing unconsciousness. Additionally, emergence from GA, a passive process that occurs after anesthetic removal, is associated with lower anesthetic concentrations in the brain compared to doses associated with induction of GA. AMPK, an evolutionarily conserved kinase activated by cellular stress (e.g. increases in calcium [Ca2+] and/or reactive oxygen species [ROS], etc.) increases lifespan and healthspan in several model organisms. AMPK is located throughout the mammalian brain, including in neurons of the thalamus, hypothalamus, and striatum as well as in pyramidal neurons in the hippocampus and cortex. Increases in ROS and Ca2+ play critical roles in neuronal excitation and glutamate, the primary excitatory neurotransmitter in the human brain, activates AMPK in cortical neurons. Nearly every neurotransmitter released from ascending arousal circuits that promote wakefulness, arousal, and consciousness activates AMPK, including acetylcholine, histamine, orexin-A, dopamine, and norepinephrine. Several GAs that are commonly used to induce LOC in human patients also activate AMPK (e.g. propofol, sevoflurane, isoflurane, dexmedetomidine, ketamine, midazolam). Various compounds that accelerate emergence from anesthesia, thus mitigating problematic effects associated with delayed emergence such as delirium, also activate AMPK (e.g. nicotine, caffeine, forskolin, carbachol). GAs and neurotransmitters also act as preconditioning agents and the GABAA receptor inhibitor bicuculline, which reverses propofol anesthesia, also activates AMPK in cortical neurons. We propose the novel hypothesis that cellular stress-induced AMPK activation links wakefulness, arousal, and consciousness with paradoxical excitation and accelerated emergence from anesthesia. Because AMPK activators including metformin and nicotine promote proliferation and differentiation of neural stem cells located in the subventricular zone and the dentate gyrus, AMPK activation may also enhance brain repair and promote potential recovery from disorders of consciousness (i.e. minimally conscious state, vegetative state, coma).
Collapse
|
41
|
Michel-Macías C, Morales-Barquet DA, Reyes-Palomino AM, Machuca-Vaca JA, Orozco-Guillén A. Single dose of propofol causing propofol infusion syndrome in a newborn. Oxf Med Case Reports 2018; 2018:omy023. [PMID: 29942532 PMCID: PMC6007798 DOI: 10.1093/omcr/omy023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/14/2018] [Accepted: 04/18/2018] [Indexed: 01/06/2023] Open
Abstract
Propofol infusion syndrome (PRIS) is a rare syndrome originally described in critically ill children undergoing long-term (> 48 h) propofol infusion at high doses (> 4 mg/kg/h). Severe metabolic acidosis, rhabdomyolysis, renal failure and fatal cardiac failure are the features. Herein, we present a case of a newborn who developed PRIS after a single bolus dose of propofol at 3.2 mg/kg/do, developing rhabdomyolysis and severe metabolic acidosis, with a successful outcome after medical therapy.
Collapse
Affiliation(s)
- C Michel-Macías
- Neonatal Intensive Care Unit, Instituto Nacional de Perinatología, Mexico City, Mexico
| | - D A Morales-Barquet
- Neonatal Intensive Care Unit, Instituto Nacional de Perinatología, Mexico City, Mexico
| | - A M Reyes-Palomino
- Neonatal Intensive Care Unit, Instituto Nacional de Perinatología, Mexico City, Mexico
| | - J A Machuca-Vaca
- Neonatal Intensive Care Unit, Instituto Nacional de Perinatología, Mexico City, Mexico
| | - A Orozco-Guillén
- Neonatal Intensive Care Unit, Instituto Nacional de Perinatología, Mexico City, Mexico
| |
Collapse
|
42
|
Sumi C, Okamoto A, Tanaka H, Kusunoki M, Shoji T, Uba T, Adachi T, Iwai T, Nishi K, Harada H, Bono H, Matsuo Y, Hirota K. Suppression of mitochondrial oxygen metabolism mediated by the transcription factor HIF-1 alleviates propofol-induced cell toxicity. Sci Rep 2018; 8:8987. [PMID: 29895831 PMCID: PMC5997661 DOI: 10.1038/s41598-018-27220-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/23/2018] [Indexed: 01/22/2023] Open
Abstract
A line of studies strongly suggest that the intravenous anesthetic, propofol, suppresses mitochondrial oxygen metabolism. It is also indicated that propofol induces the cell death in a reactive oxygen species (ROS)-dependent manner. Because hypoxia-inducible factor 1 (HIF-1) is a transcription factor which is involved in cellular metabolic reprogramming by modulating gene expressions of enzymes including glycolysis pathway and oxygen utilization of mitochondria, we examined the functional role of HIF-1 activity in propofol-induced cell death. The role of HIF-1 activity on oxygen and energy metabolisms and propofol-induced cell death and caspase activity was examined in renal cell-derived RCC4 cells: RCC4-EV cells which lack von Hippel-Lindau protein (VHL) protein expression and RCC4-VHL cells, which express exogenous VHL, and in neuronal SH-SY5Y cells. It was demonstrated that HIF-1 is involved in suppressing oxygen consumption and facilitating glycolysis in cells and that the resistance to propofol-induced cell death was established in a HIF-1 activation-dependent manner. It was also demonstrated that HIF-1 activation by treatment with HIFα-hydroxylase inhibitors such as n-propyl gallate and dimethyloxaloylglycine, alleviated the toxic effects of propofol. Thus, the resistance to propofol toxicity was conferred by HIF-1 activation by not only genetic deletion of VHL but also exposure to HIFα-hydroxylase inhibitors.
Collapse
Affiliation(s)
- Chisato Sumi
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Akihisa Okamoto
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Hiromasa Tanaka
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Munenori Kusunoki
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Tomohiro Shoji
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Takeo Uba
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan.,Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Takehiko Adachi
- Department of Anesthesiology, Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Teppei Iwai
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Kenichiro Nishi
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Radiation Biology Center, Kyoto University, Kyoto, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Saitama, Japan
| | - Hidemasa Bono
- Database Center for Life Science (DBCLS), Research Organization of Information and Systems (ROIS), Mishima, Japan
| | - Yoshiyuki Matsuo
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan.
| |
Collapse
|
43
|
Lagarto JL, Phipps JE, Faller L, Ma D, Unger J, Bec J, Griffey S, Sorger J, Farwell DG, Marcu L. Electrocautery effects on fluorescence lifetime measurements: An in vivo study in the oral cavity. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2018; 185:90-99. [PMID: 29883910 DOI: 10.1016/j.jphotobiol.2018.05.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/27/2018] [Accepted: 05/25/2018] [Indexed: 02/06/2023]
Affiliation(s)
- João L Lagarto
- University of California, Davis, Department of Biomedical Engineering, 1 Shields Avenue, Davis, CA 95616, United States
| | - Jennifer E Phipps
- University of California, Davis, Department of Biomedical Engineering, 1 Shields Avenue, Davis, CA 95616, United States
| | - Leta Faller
- University of California, Davis, Department of Otolaryngology-Head and Neck Surgery, 2521 Stockton Boulevard, Suite 7200, Sacramento, California 95817, United States
| | - Dinglong Ma
- University of California, Davis, Department of Biomedical Engineering, 1 Shields Avenue, Davis, CA 95616, United States
| | - Jakob Unger
- University of California, Davis, Department of Biomedical Engineering, 1 Shields Avenue, Davis, CA 95616, United States
| | - Julien Bec
- University of California, Davis, Department of Biomedical Engineering, 1 Shields Avenue, Davis, CA 95616, United States
| | - Stephen Griffey
- University of California, Davis, Comparative Pathology Laboratory, 1 Shields Avenue, Davis, CA 95616, United States
| | - Jonathan Sorger
- Intuitive Surgical, 1020 Kifer Road, Sunnyvale, CA 94086-5304, United States
| | - D Gregory Farwell
- University of California, Davis, Department of Otolaryngology-Head and Neck Surgery, 2521 Stockton Boulevard, Suite 7200, Sacramento, California 95817, United States
| | - Laura Marcu
- University of California, Davis, Department of Biomedical Engineering, 1 Shields Avenue, Davis, CA 95616, United States.
| |
Collapse
|