1
|
Corda PO, Silva JV, Almeida CR, Pierre P, Fardilha M. De Novo Protein Synthesis Occurs Through the Cytoplasmic Translation Machinery in Mammalian Spermatozoa. J Cell Physiol 2025; 240:e70038. [PMID: 40373039 DOI: 10.1002/jcp.70038] [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: 03/23/2025] [Revised: 04/02/2025] [Accepted: 04/10/2025] [Indexed: 05/17/2025]
Abstract
The current hypothesis suggests that translation occurs in capacitated spermatozoa through mitochondrial ribosomes. Mitochondrial translation has several particularities, which rise some questions about how mitochondrial ribosomes can ensure sperm translation activity. Here, we aimed to elucidate if cytoplasmic translation occurs in mammalian spermatozoa. A bioinformatic workflow was performed to identify translation-related proteins in human spermatozoa and their association with cytoplasmic translation. The surface sensing of translation (SUnSET) method was used to measure translation activity in capacitated human and bovine spermatozoa. Two translation inhibitors, cycloheximide (CHX, cytoplasmic) and D-chloramphenicol (D-CP, mitochondrial) were used to identify which ribosomes were active in sperm. To spot newly synthesized proteins, puromycin-peptides were immunoprecipitated and analysed by mass spectrometry. A second approach was performed using translation inhibitors and analysing the sperm proteome by mass spectrometry. Bioinformatic analysis revealed that human spermatozoa possess 510 translation proteins, which were enriched for cytoplasmic mRNA translation. CHX decreased translation activity in mammalian sperm, whereas no effect was observed after D-CP treatment. Nine proteins were immunoprecipitated and identified as newly synthesized in capacitated bovine spermatozoa. CHX and D-CP decreased the level of 22 proteins that were replaced, or de novo translated during capacitation. New proteins were associated with relevant processes for sperm physiology. Both translation inhibitors decreased sperm rapid progressive motility and increased sperm immotility. Our results proved sperm translation occurs through cytoplasmic translation machinery in capacitated bovine and human spermatozoa. These results also support that sperm translation is required during capacitation to produce relevant proteins for sperm functions.
Collapse
Affiliation(s)
- Pedro O Corda
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Aveiro, Portugal
| | - Joana Vieira Silva
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Aveiro, Portugal
| | - Catarina R Almeida
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Aveiro, Portugal
| | - Philippe Pierre
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Aveiro, Portugal
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, Bouches-du-Rhône, France
| | - Margarida Fardilha
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Aveiro, Portugal
| |
Collapse
|
2
|
Zhuang F, Huang S, Liu L. MALSU1-mediated regulation of mitochondrial function governs proliferation and doxorubicin resistance in triple-negative breast cancer cells. Mol Cell Biochem 2025; 480:1197-1207. [PMID: 38896203 DOI: 10.1007/s11010-024-05053-6] [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: 01/21/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
Triple-negative breast cancer (TNBC) poses a formidable challenge in oncology due to its aggressive nature and limited treatment options. Although doxorubicin, a widely used chemotherapeutic agent, shows efficacy in TNBC treatment, acquired resistance remains a significant obstacle. Our study explores the role of MALSU1, a regulator of mitochondrial translation, in TNBC and its impact on cell proliferation and doxorubicin resistance. We observed increased MALSU1 expression in TNBC, correlating with poor patient prognosis. MALSU1 knockdown in TNBC cells significantly reduced proliferation, indicating its pivotal role in sustaining cell growth. Mechanistically, MALSU1 depletion resulted in decreased activities of mitochondrial respiratory chain complexes, cellular ATP levels, and mitochondrial respiration. Notably, exogenous addition of normal mitochondria restored proliferation and mitochondrial respiration in MALSU1-depleted TNBC cells. Importantly, MALSU1 knockdown enhanced the sensitivity of doxorubicin-resistant TNBC cells to doxorubicin treatment. Furthermore, pharmacological inhibition of mitochondrial translation using tigecycline and chloramphenicol mimicked the effects of MALSU1 knockdown, suggesting mitochondrial translation as a potential therapeutic target. Taken together, our findings not only elucidate the intricate role of MALSU1 in TNBC biology and doxorubicin resistance but also lay the groundwork for future investigations targeting MALSU1 and/or mitochondrial translation as a promising avenue for developing innovative therapeutic strategies against TNBC.
Collapse
Affiliation(s)
- Feifei Zhuang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong Province, China
| | - Shaoyan Huang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong Province, China
| | - Lei Liu
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong Province, China.
| |
Collapse
|
3
|
Karamanolis NN, Kounatidis D, Vallianou NG, Dimitriou K, Tsaroucha E, Tsioulos G, Anastasiou IA, Mavrothalassitis E, Karampela I, Dalamaga M. Unraveling the Anti-Cancer Mechanisms of Antibiotics: Current Insights, Controversies, and Future Perspectives. Antibiotics (Basel) 2024; 14:9. [PMID: 39858295 PMCID: PMC11762948 DOI: 10.3390/antibiotics14010009] [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: 12/01/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Cancer persists as a significant global health challenge, claiming millions of lives annually despite remarkable strides in therapeutic innovation. Challenges such as drug resistance, toxicity, and suboptimal efficacy underscore the need for novel treatment paradigms. In this context, the repurposing of antibiotics as anti-cancer agents has emerged as an attractive prospect for investigation. Diverse classes of antibiotics have exhibited promising anti-cancer properties in both in vitro and in vivo studies. These mechanisms include the induction of apoptosis and cell cycle arrest, generation of reactive oxygen species, and inhibition of key regulators of cell proliferation and migration. Additional effects involve the disruption of angiogenesis and modulation of pivotal processes such as inflammation, immune response, mitochondrial dynamics, ferroptosis, and autophagy. Furthermore, antibiotics have demonstrated the potential to enhance the efficacy of conventional modalities like chemotherapy and radiotherapy, while alleviating treatment-induced toxicities. Nevertheless, the integration of antibiotics into oncological applications remains contentious, with concerns centered on their disruption of gut microbiota, interference with immunotherapeutic strategies, contribution to microbial resistance, and potential association with tumorigenesis. This narrative review explores the mechanisms of antibiotics' anti-cancer activity, addresses controversies about their dual role in cancer biology, and envisions future perspectives that include the development of novel derivatives and innovative frameworks for their incorporation into cancer treatment paradigms.
Collapse
Affiliation(s)
- Nikolaos Nektarios Karamanolis
- Second Department of Internal Medicine, Hippokratio General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.N.K.); (K.D.)
| | - Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (I.A.A.)
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (E.T.); (E.M.)
| | - Krystalia Dimitriou
- Second Department of Internal Medicine, Hippokratio General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.N.K.); (K.D.)
| | - Eleni Tsaroucha
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (E.T.); (E.M.)
| | - Georgios Tsioulos
- Fourth Department of Internal Medicine, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Ioanna A. Anastasiou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (I.A.A.)
| | - Evangelos Mavrothalassitis
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (E.T.); (E.M.)
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12461 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
4
|
Gabrielli AP, Novikova L, Ranjan A, Wang X, Ernst NJ, Abeykoon D, Roberts A, Kopp A, Mansel C, Qiao L, Lysaker CR, Wiedling IW, Wilkins HM, Swerdlow RH. Inhibiting mtDNA transcript translation alters Alzheimer's disease-associated biology. Alzheimers Dement 2024; 20:8429-8443. [PMID: 39441557 PMCID: PMC11667520 DOI: 10.1002/alz.14275] [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: 06/29/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) features changes in mitochondrial structure and function. Investigators debate where to position mitochondrial pathology within the chronology and context of other AD features. METHODS To address whether mitochondrial dysfunction alters AD-implicated genes and proteins, we treated SH-SY5Y cells and induced pluripotent stem cell (iPSC)-derived neurons with chloramphenicol, an antibiotic that inhibits mtDNA-generated transcript translation. We characterized adaptive, AD-associated gene, and AD-associated protein responses. RESULTS SH-SY5Y cells and iPSC neurons responded to mtDNA transcript translation inhibition by increasing mtDNA copy number and transcription. Nuclear-expressed respiratory chain mRNA and protein levels also changed. There were AD-consistent concordant and model-specific changes in amyloid precursor protein, beta amyloid, apolipoprotein E, tau, and α-synuclein biology. DISCUSSION Primary mitochondrial dysfunction induces compensatory organelle responses, changes nuclear gene expression, and alters the biology of AD-associated genes and proteins in ways that may recapitulate brain aging and AD molecular phenomena. HIGHLIGHTS In AD, mitochondrial dysfunction could represent a disease cause or consequence. We inhibited mitochondrial translation in human neuronal cells and neurons. Mitochondrial and nuclear gene expression shifted in adaptive-consistent patterns. APP, Aβ, APOE, tau, and α-synuclein biology changed in AD-consistent patterns. Mitochondrial stress creates an environment that promotes AD pathology.
Collapse
Affiliation(s)
- Alexander P. Gabrielli
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Cell Biology and PhysiologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Lesya Novikova
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
| | - Amol Ranjan
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
| | - Xiaowan Wang
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
| | - Nicholas J. Ernst
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
| | - Dhanushki Abeykoon
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
| | - Anysja Roberts
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
| | - Annie Kopp
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
| | - Clayton Mansel
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
| | - Linlan Qiao
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
| | - Colton R. Lysaker
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Biochemistry and Molecular Biology, University of Kansas Medical CenterKansas CityKansasUSA
- Neurologythe University of Kansas Medical CenterKansas CityKansasUSA
| | - Ian W. Wiedling
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Neurologythe University of Kansas Medical CenterKansas CityKansasUSA
| | - Heather M. Wilkins
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Biochemistry and Molecular Biology, University of Kansas Medical CenterKansas CityKansasUSA
- Neurologythe University of Kansas Medical CenterKansas CityKansasUSA
| | - Russell H. Swerdlow
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Cell Biology and PhysiologyUniversity of Kansas Medical CenterKansas CityKansasUSA
- Biochemistry and Molecular Biology, University of Kansas Medical CenterKansas CityKansasUSA
- Neurologythe University of Kansas Medical CenterKansas CityKansasUSA
| |
Collapse
|
5
|
Matei E, Ionescu AC, Enciu M, Popovici V, Mitroi AF, Aschie M, Deacu M, Băltățescu GI, Nicolau AA, Roșu MC, Cristian M, Dobrin N, Ștefanov C, Pundiche Butcaru M, Cozaru GC. Cell death and DNA damage via ROS mechanisms after applied antibiotics and antioxidants doses in prostate hyperplasia primary cell cultures. Medicine (Baltimore) 2024; 103:e39450. [PMID: 39287312 PMCID: PMC11404886 DOI: 10.1097/md.0000000000039450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/08/2024] [Accepted: 08/05/2024] [Indexed: 09/19/2024] Open
Abstract
Tumor heterogeneity results in aggressive cancer phenotypes with acquired resistance. However, combining chemical treatment with adjuvant therapies that cause cellular structure and function perturbations may diminish the ability of cancer cells to resist at chemical treatment and lead to a less aggressive cancer phenotype. Applied treatments on prostate hyperplasia primary cell cultures exerted their antitumor activities through mechanisms including cell cycle blockage, oxidative stress, and cell death induction by flow cytometry methods. A 5.37 mM Chloramphenicol dose acts on prostate hyperplasia cells by increasing the pro-oxidant status, inducing apoptosis, autophagy, and DNA damage, but without ROS changes. Adding 6.30 mM vitamin C or 622 µM vitamin E as a supplement to 859.33 µM Chloramphenicol dose in prostate hyperplasia cells determines a significant increase of ROS level for a part of cells. However, other cells remain refractory to initial ROS, with significant changes in apoptosis, autophagy, and cell cycle arrest in G0/G1 or G2/M. When the dose of Chloramphenicol was increased to 5.37 mM for 6.30 mM of vitamin C, prostate hyperplasia cells reacted by ROS level drastically decreased, cell cycle arrest in G2/M, active apoptosis, and autophagy. The pro-oxidant action of 1.51 mM Erythromycin dose in prostate hyperplasia cell cultures induces changes in the apoptosis mechanisms and cell cycle arrest in G0/G1. Addition of 6.30 mM vitamin C to 1.51 mM Erythromycin dose in hyperplasia cell cultures, the pro-oxidant status determines diminished caspase 3/7 mechanism activation, but ROS level presents similar changes as Chloramphenicol dose and cell cycle arrest in G2/M. Flow cytometric analysis of cell death, oxidative stress, and cell cycle are recommended as laboratory techniques in therapeutic and diagnostic fields.
Collapse
Affiliation(s)
- Elena Matei
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, “Ovidius” University of Constanta, Constanta, Romania
| | - Anita Cristina Ionescu
- Institute of Oncology “Prof. Dr. Alexandru Trestioreanu”, Bucharest, Romania
- Medicine Faculty, “Ovidius” University of Constanta, Constanta, Romania
| | - Manuela Enciu
- Medicine Faculty, “Ovidius” University of Constanta, Constanta, Romania
- Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, Constanta, Romania
| | - Violeta Popovici
- Laboratory of Bacteriology, Microbiology and Pharmacology, Center for Mountain Economics (INCE-CE-MONT), National Institute of Economic Research “Costin C. Kiritescu”, Suceava County, Romania
| | - Anca Florentina Mitroi
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, “Ovidius” University of Constanta, Constanta, Romania
- Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, Constanta, Romania
| | - Mariana Aschie
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, “Ovidius” University of Constanta, Constanta, Romania
- Medicine Faculty, “Ovidius” University of Constanta, Constanta, Romania
- Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, Constanta, Romania
- Romanian Academy of Scientists, Bucharest, Romania
| | - Mariana Deacu
- Medicine Faculty, “Ovidius” University of Constanta, Constanta, Romania
- Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, Constanta, Romania
| | - Gabriela Isabela Băltățescu
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, “Ovidius” University of Constanta, Constanta, Romania
- Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, Constanta, Romania
| | - Antonela-Anca Nicolau
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, “Ovidius” University of Constanta, Constanta, Romania
- Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, Constanta, Romania
| | - Mihai Cătălin Roșu
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, “Ovidius” University of Constanta, Constanta, Romania
| | - Miruna Cristian
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, “Ovidius” University of Constanta, Constanta, Romania
| | - Nicolae Dobrin
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, “Ovidius” University of Constanta, Constanta, Romania
| | - Constanța Ștefanov
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, “Ovidius” University of Constanta, Constanta, Romania
| | | | - Georgeta Camelia Cozaru
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, “Ovidius” University of Constanta, Constanta, Romania
- Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, Constanta, Romania
| |
Collapse
|
6
|
Huang S, Jiang H, Huang Z, Li Y, Hu H. Mitochondrial RNA modification: A novel therapeutic target to combat metastasis. Cell Biol Int 2024; 48:233-236. [PMID: 38225665 DOI: 10.1002/cbin.12124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/04/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024]
Affiliation(s)
- Shifang Huang
- Department of Pharmacology, Yongzhou Vocational Technical College, Yongzhou, China
| | - Honglu Jiang
- Department of Pharmacology, Yongzhou Radio and TV University, Yongzhou, China
| | - Zhen Huang
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuting Li
- Department of Pharmacology, Yongzhou Vocational Technical College, Yongzhou, China
| | - Haoliang Hu
- Zoology Key Laboratory of Hunan Higher Education, Changde Research Centre for Artificial Intelligence and Biomedicine, College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, China
| |
Collapse
|
7
|
Kiy RT, Khoo SH, Chadwick AE. Assessing the mitochondrial safety profile of the molnupiravir active metabolite, β-d-N4-hydroxycytidine (NHC), in the physiologically relevant HepaRG model. Toxicol Res (Camb) 2024; 13:tfae012. [PMID: 38328743 PMCID: PMC10848230 DOI: 10.1093/toxres/tfae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 02/09/2024] Open
Abstract
Background β-d-N4-Hydroxycytidine (NHC) is the active metabolite of molnupiravir, a broad-spectrum antiviral approved by the MHRA for COVID-19 treatment. NHC induces lethal mutagenesis of the SARS-CoV-2 virus, undergoing incorporation into the viral genome and arresting viral replication. It has previously been reported that several nucleoside analogues elicit off-target inhibition of mitochondrial DNA (mtDNA) or RNA replication. Although NHC does not exert these effects in HepG2 cells, HepaRG are proven to be advantageous over HepG2 for modelling nucleoside analogue-induced mitochondrial dysfunction. Therefore, the objective of this work was to assess the mitotoxic potential of NHC in HepaRG cells, a model more closely resembling physiological human liver. Methods Differentiated HepaRG cells were exposed to 1-60 μM NHC for 3-14 days to investigate effects of sub-, supra-, and clinically-relevant exposures (in the UK, molnupiravir for COVID-19 is indicated for 5 days and reported Cmax is 16 μM). Following drug incubation, cell viability, mtDNA copy number, mitochondrial protein expression, and mitochondrial respiration were assessed. Results NHC induced minor decreases in cell viability at clinically relevant exposures, but did not decrease mitochondrial protein expression. The effects on mtDNA were variable, but typically copy number was increased. At supra-clinical concentrations (60 μM), NHC reduced mitochondrial respiration, but did not appear to induce direct electron transport chain dysfunction. Conclusions Overall, NHC does not cause direct mitochondrial toxicity in HepaRG cells at clinically relevant concentrations, but may induce minor cellular perturbations. As HepaRG cells have increased physiological relevance, these findings provide additional assurance of the mitochondrial safety profile of NHC.
Collapse
Affiliation(s)
- Robyn T Kiy
- Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, United Kingdom
| | - Saye H Khoo
- Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, United Kingdom
- Tropical Infectious Diseases Unit, Royal Liverpool University Hospital, Prescot Street, Liverpool, L7 8XP, United Kingdom
| | - Amy E Chadwick
- Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, United Kingdom
| |
Collapse
|
8
|
Miki K, Yagi M, Noguchi N, Do Y, Otsuji R, Kuga D, Kang D, Yoshimoto K, Uchiumi T. Induction of glioblastoma cell ferroptosis using combined treatment with chloramphenicol and 2-deoxy-D-glucose. Sci Rep 2023; 13:10497. [PMID: 37380755 DOI: 10.1038/s41598-023-37483-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023] Open
Abstract
Glioblastoma, a malignant tumor, has no curative treatment. Recently, mitochondria have been considered a potential target for treating glioblastoma. Previously, we reported that agents initiating mitochondrial dysfunction were effective under glucose-starved conditions. Therefore, this study aimed to develop a mitochondria-targeted treatment to achieve normal glucose conditions. This study used U87MG (U87), U373, and patient-derived stem-like cells as well as chloramphenicol (CAP) and 2-deoxy-D-glucose (2-DG). We investigated whether CAP and 2-DG inhibited the growth of cells under normal and high glucose concentrations. In U87 cells, 2-DG and long-term CAP administration were more effective under normal glucose than high-glucose conditions. In addition, combined CAP and 2-DG treatment was significantly effective under normal glucose concentration in both normal oxygen and hypoxic conditions; this was validated in U373 and patient-derived stem-like cells. 2-DG and CAP acted by influencing iron dynamics; however, deferoxamine inhibited the efficacy of these agents. Thus, ferroptosis could be the underlying mechanism through which 2-DG and CAP act. In conclusion, combined treatment of CAP and 2-DG drastically inhibits cell growth of glioblastoma cell lines even under normal glucose conditions; therefore, this treatment could be effective for glioblastoma patients.
Collapse
Affiliation(s)
- Kenji Miki
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Mikako Yagi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Naoki Noguchi
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yura Do
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Ryosuke Otsuji
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Daisuke Kuga
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Koji Yoshimoto
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan.
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
9
|
Fujii S. Antimicrobial Stewardship Initiatives and Studies by the Hospital Pharmacist: Study of Linezolid-associated Thrombocytopenia. YAKUGAKU ZASSHI 2022; 142:1307-1312. [DOI: 10.1248/yakushi.22-00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Affiliation(s)
- Satoshi Fujii
- Department of Hospital Pharmacy, Sapporo Medical University Hospital
| |
Collapse
|
10
|
Richardson BC, Shek R, Van Voorhis WC, French JB. Structure of Klebsiella pneumoniae adenosine monophosphate nucleosidase. PLoS One 2022; 17:e0275023. [PMID: 36264993 PMCID: PMC9584410 DOI: 10.1371/journal.pone.0275023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/09/2022] [Indexed: 11/05/2022] Open
Abstract
Klebsiella pneumoniae is a bacterial pathogen that is increasingly responsible for hospital-acquired pneumonia and sepsis. Progressive development of antibiotic resistance has led to higher mortality rates and creates a need for novel treatments. Because of the essential role that nucleotides play in many bacterial processes, enzymes involved in purine and pyrimidine metabolism and transport are ideal targets for the development of novel antibiotics. Herein we describe the structure of K. pneumoniae adenosine monophosphate nucleosidase (KpAmn), a purine salvage enzyme unique to bacteria, as determined by cryoelectron microscopy. The data detail a well conserved fold with a hexameric overall structure and clear density for the putative active site residues. Comparison to the crystal structures of homologous prokaryotic proteins confirms the presence of many of the conserved structural features of this protein yet reveals differences in distal loops in the absence of crystal contacts. This first cryo-EM structure of an Amn enzyme provides a basis for future structure-guided drug development and extends the accuracy of structural characterization of this family of proteins beyond this clinically relevant organism.
Collapse
Affiliation(s)
- Brian C. Richardson
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Roger Shek
- Division of Allergy and Infectious Diseases, Department of Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington School of Medicine, Seattle, WA, United States of America
| | - Wesley C. Van Voorhis
- Division of Allergy and Infectious Diseases, Department of Medicine, Center for Emerging and Re-emerging Infectious Diseases, University of Washington School of Medicine, Seattle, WA, United States of America
| | - Jarrod B. French
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| |
Collapse
|
11
|
Miki K, Yagi M, Yoshimoto K, Kang D, Uchiumi T. Mitochondrial dysfunction and impaired growth of glioblastoma cell lines caused by antimicrobial agents inducing ferroptosis under glucose starvation. Oncogenesis 2022; 11:59. [PMID: 36195584 PMCID: PMC9532440 DOI: 10.1038/s41389-022-00437-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/09/2022] Open
Abstract
Glioblastoma is a difficult-to-cure disease owing to its malignancy. Under normal circumstances, cancer is dependent on the glycolytic system for growth, and mitochondrial oxidative phosphorylation (OXPHOS) is not well utilized. Here, we investigated the efficacy of mitochondria-targeted glioblastoma therapy in cell lines including U87MG, LN229, U373, T98G, and two patient-derived stem-like cells. When glioblastoma cells were exposed to a glucose-starved condition (100 mg/l), they rely on mitochondrial OXPHOS for growth, and mitochondrial translation product production is enhanced. Under these circumstances, drugs that inhibit mitochondrial translation, called antimicrobial agents, can cause mitochondrial dysfunction and thus can serve as a therapeutic option for glioblastoma. Antimicrobial agents activated the nuclear factor erythroid 2-related factor 2–Kelch-like ECH-associated protein 1 pathway, resulting in increased expression of heme oxygenase-1. Accumulation of lipid peroxides resulted from the accumulation of divalent iron, and cell death occurred via ferroptosis. In conclusion, mitochondrial OXPHOS is upregulated in glioblastoma upon glucose starvation. Under this condition, antimicrobial agents cause cell death via ferroptosis. The findings hold promise for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Kenji Miki
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Mikako Yagi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Yoshimoto
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan. .,Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
12
|
Syroegin EA, Aleksandrova EV, Polikanov YS. Structural basis for the inability of chloramphenicol to inhibit peptide bond formation in the presence of A-site glycine. Nucleic Acids Res 2022; 50:7669-7679. [PMID: 35766409 PMCID: PMC9303264 DOI: 10.1093/nar/gkac548] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/08/2022] [Accepted: 06/28/2022] [Indexed: 12/11/2022] Open
Abstract
Ribosome serves as a universal molecular machine capable of synthesis of all the proteins in a cell. Small-molecule inhibitors, such as ribosome-targeting antibiotics, can compromise the catalytic versatility of the ribosome in a context-dependent fashion, preventing transpeptidation only between particular combinations of substrates. Classic peptidyl transferase center inhibitor chloramphenicol (CHL) fails to inhibit transpeptidation reaction when the incoming A site acceptor substrate is glycine, and the molecular basis for this phenomenon is unknown. Here, we present a set of high-resolution X-ray crystal structures that explain why CHL is unable to inhibit peptide bond formation between the incoming glycyl-tRNA and a nascent peptide that otherwise is conducive to the drug action. Our structures reveal that fully accommodated glycine residue can co-exist in the A site with the ribosome-bound CHL. Moreover, binding of CHL to a ribosome complex carrying glycyl-tRNA does not affect the positions of the reacting substrates, leaving the peptide bond formation reaction unperturbed. These data exemplify how small-molecule inhibitors can reshape the A-site amino acid binding pocket rendering it permissive only for specific amino acid residues and rejective for the other substrates extending our detailed understanding of the modes of action of ribosomal antibiotics.
Collapse
Affiliation(s)
- Egor A Syroegin
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Elena V Aleksandrova
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Yury S Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
13
|
Corda PO, Silva JV, Pereira SC, Barros A, Alves MG, Fardilha M. Bioinformatic Approach to Unveil Key Differentially Expressed Proteins in Human Sperm After Slow and Rapid Cryopreservation. Front Cell Dev Biol 2022; 9:759354. [PMID: 35145967 PMCID: PMC8821918 DOI: 10.3389/fcell.2021.759354] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/23/2021] [Indexed: 11/25/2022] Open
Abstract
Currently, two conventional freezing techniques are used in sperm cryopreservation: slow freezing (SF) and rapid freezing (RF). Despite the protocolar improvements, cryopreservation still induces significant alterations in spermatozoon that are poorly understood. Here, available proteomic data from human cryopreserved sperm was analyzed through bioinformatic tools to unveil key differentially expressed proteins (DEPs) that can be used as modulation targets or quality markers. From the included proteomic studies, 160 and 555 DEPs were collected for SF and RF groups, respectively. For each group, an integrative network was constructed using gene ontology and protein-protein interaction data to identify key DEPs. Among them, arylsulfatase A (ARSA) was highlighted in both freezing networks, and low ARSA levels have been associated with poor-sperm quality. Thus, ARSA was selected for further experimental investigation and its levels were assessed in cryopreserved samples by western blot. ARSA levels were significantly decreased in RF and SF samples (∼31.97 and ∼39.28%, respectively). The bioinformatic analysis also revealed that the DEPs were strongly associated with proteasomal and translation pathways. The purposed bioinformatic approach allowed the identification of potential key DEPs in freeze-thawed human spermatozoa. ARSA has the potential to be used as a marker to assess sperm quality after cryopreservation.
Collapse
Affiliation(s)
- Pedro O Corda
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Joana Vieira Silva
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal.,Department of Chemistry, QOPNA and LAQV, University of Aveiro, Aveiro, Portugal.,Clinical and Experimental Endocrinology, Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Sara C Pereira
- Clinical and Experimental Endocrinology, Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Alberto Barros
- Department of Chemistry, QOPNA and LAQV, University of Aveiro, Aveiro, Portugal.,Centre for Reproductive Genetics A. Barros, Porto, Portugal.,Department of Genetics, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Marco G Alves
- Clinical and Experimental Endocrinology, Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Margarida Fardilha
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| |
Collapse
|
14
|
Syroegin EA, Flemmich L, Klepacki D, Vazquez-Laslop N, Micura R, Polikanov YS. Structural basis for the context-specific action of the classic peptidyl transferase inhibitor chloramphenicol. Nat Struct Mol Biol 2022; 29:152-161. [PMID: 35165455 PMCID: PMC9071271 DOI: 10.1038/s41594-022-00720-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023]
Abstract
Ribosome-targeting antibiotics serve as powerful antimicrobials and as tools for studying the ribosome, the catalytic peptidyl transferase center (PTC) of which is targeted by many drugs. The classic PTC-acting antibiotic chloramphenicol (CHL) and the newest clinically significant linezolid (LZD) were considered indiscriminate inhibitors of protein synthesis that cause ribosome stalling at every codon of every gene being translated. However, recent discoveries have shown that CHL and LZD preferentially arrest translation when the ribosome needs to polymerize particular amino acid sequences. The molecular mechanisms that underlie the context-specific action of ribosome inhibitors are unknown. Here we present high-resolution structures of ribosomal complexes, with or without CHL, carrying specific nascent peptides that support or negate the drug action. Our data suggest that the penultimate residue of the nascent peptide directly modulates antibiotic affinity to the ribosome by either establishing specific interactions with the drug or by obstructing its proper placement in the binding site.
Collapse
Affiliation(s)
- Egor A Syroegin
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Laurin Flemmich
- Institute of Organic Chemistry, University of Innsbruck, Center of Molecular Biosciences Innsbruck, Innsbruck, Austria
| | - Dorota Klepacki
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Nora Vazquez-Laslop
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Ronald Micura
- Institute of Organic Chemistry, University of Innsbruck, Center of Molecular Biosciences Innsbruck, Innsbruck, Austria.
| | - Yury S Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA.
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
15
|
Chen RN, Kang SH, Li J, Lu LN, Luo XP, Wu L. Comparison and recent progress of molecular imprinting technology and dummy template molecular imprinting technology. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2021; 13:4538-4556. [PMID: 34570126 DOI: 10.1039/d1ay01014j] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Molecular imprinting technology for the preparation of polymers with specific molecular recognition function had become one of the current research hotspots. It has been widely applied in chromatographic separation, antibody and receptor mimetics, solid-phase extraction, bio-sensors, and other fields in the last decades. In this study, molecular imprinting technology was summarized from the points of templates and dummy templates, and four typical target analytes were selected to compare the differences between templates and dummy templates. The current status and prospects of molecular imprinting technology were also proposed.
Collapse
Affiliation(s)
| | | | - Jia Li
- Northwest Minzu University, China.
| | - Li-Na Lu
- Northwest Minzu University, China.
| | | | - Lan Wu
- Northwest Minzu University, China.
| |
Collapse
|
16
|
Kim T, Jeon J, Park JS, Park Y, Kim J, Noh H, Kim HS, Seo H. Matrix Metalloproteinase-8 Inhibitor Ameliorates Inflammatory Responses and Behavioral Deficits in LRRK2 G2019S Parkinson's Disease Model Mice. Biomol Ther (Seoul) 2021; 29:483-491. [PMID: 34045367 PMCID: PMC8411029 DOI: 10.4062/biomolther.2020.181] [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: 10/15/2020] [Revised: 03/15/2021] [Accepted: 04/28/2021] [Indexed: 11/14/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder that involves the loss of dopaminergic neurons in the substantia nigra (SN). Matrix metalloproteinases-8 (MMP-8), neutrophil collagenase, is a functional player in the progressive pathology of various inflammatory disorders. In this study, we administered an MMP-8 inhibitor (MMP-8i) in Leucine-rich repeat kinase 2 (LRRK2) G2019S transgenic mice, to determine the effects of MMP-8i on PD pathology. We observed a significant increase of ionized calcium-binding adapter molecule 1 (Iba1)-positive activated microglia in the striatum of LRRK2 G2019S mice compared to normal control mice, indicating enhanced neuro-inflammatory responses. The increased number of Iba1-positive activated microglia in LRRK2 G2019S PD mice was down-regulated by systemic administration of MMP-8i. Interestingly, this LRRK2 G2019S PD mice showed significantly reduced size of cell body area of tyrosine hydroxylase (TH) positive neurons in SN region and MMP-8i significantly recovered cellular atrophy shown in PD model indicating distinct neuro-protective effects of MMP-8i. Furthermore, MMP-8i administration markedly improved behavioral abnormalities of motor balancing coordination in rota-rod test in LRRK2 G2019S mice. These data suggest that MMP-8i attenuates the pathological symptoms of PD through anti-inflammatory processes.
Collapse
Affiliation(s)
- Taewoo Kim
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jeha Jeon
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jin-Sun Park
- Department of Molecular Medicine and Medical Research Institute, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Yeongwon Park
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jooeui Kim
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Haneul Noh
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine and Medical Research Institute, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Hyemyung Seo
- Department of Molecular & Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| |
Collapse
|
17
|
Mitochondria and Antibiotics: For Good or for Evil? Biomolecules 2021; 11:biom11071050. [PMID: 34356674 PMCID: PMC8301944 DOI: 10.3390/biom11071050] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 01/16/2023] Open
Abstract
The discovery and application of antibiotics in the common clinical practice has undeniably been one of the major medical advances in our times. Their use meant a drastic drop in infectious diseases-related mortality and contributed to prolonging human life expectancy worldwide. Nevertheless, antibiotics are considered by many a double-edged sword. Their extensive use in the past few years has given rise to a global problem: antibiotic resistance. This factor and the increasing evidence that a wide range of antibiotics can damage mammalian mitochondria, have driven a significant sector of the medical and scientific communities to advise against the use of antibiotics for purposes other to treating severe infections. Notwithstanding, a notorious number of recent studies support the use of these drugs to treat very diverse conditions, ranging from cancer to neurodegenerative or mitochondrial diseases. In this context, there is great controversy on whether the risks associated to antibiotics outweigh their promising beneficial features. The aim of this review is to provide insight in the topic, purpose for which the most relevant findings regarding antibiotic therapies have been discussed.
Collapse
|
18
|
Tengeler AC, Emmerzaal TL, Geenen B, Verweij V, van Bodegom M, Morava E, Kiliaan AJ, Kozicz T. Early-adolescent antibiotic exposure results in mitochondrial and behavioral deficits in adult male mice. Sci Rep 2021; 11:12875. [PMID: 34145328 PMCID: PMC8213690 DOI: 10.1038/s41598-021-92203-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/21/2021] [Indexed: 11/21/2022] Open
Abstract
Exposure to antibiotic treatment has been associated with increased vulnerability to various psychiatric disorders. However, a research gap exists in understanding how adolescent antibiotic therapy affects behavior and cognition. Many antibiotics that target bacterial translation may also affect mitochondrial translation resulting in impaired mitochondrial function. The brain is one of the most metabolically active organs, and hence is the most vulnerable to impaired mitochondrial function. We hypothesized that exposure to antibiotics during early adolescence would directly affect brain mitochondrial function, and result in altered behavior and cognition. We administered amoxicillin, chloramphenicol, or gentamicin in the drinking water to young adolescent male wild-type mice. Next, we assayed mitochondrial oxidative phosphorylation complex activities in the cerebral cortex, performed behavioral screening and targeted mass spectrometry-based acylcarnitine profiling in the cerebral cortex. We found that mice exposed to chloramphenicol showed increased repetitive and compulsive-like behavior in the marble burying test, an accurate and sensitive assay of anxiety, concomitant with decreased mitochondrial complex IV activity. Our results suggest that only adolescent chloramphenicol exposure leads to impaired brain mitochondrial complex IV function, and could therefore be a candidate driver event for increased anxiety-like and repetitive, compulsive-like behaviors.
Collapse
Affiliation(s)
- Anouk C Tengeler
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands
| | - Tim L Emmerzaal
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands.,Department of Clinical Genomics, Mayo Clinic, 200 First St. SW, Rochester, MN, USA
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands
| | - Vivienne Verweij
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands
| | - Miranda van Bodegom
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands
| | - Eva Morava
- Department of Clinical Genomics, Mayo Clinic, 200 First St. SW, Rochester, MN, USA
| | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands
| | - Tamas Kozicz
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Centre for Medical Neuroscience, Preclinical Imaging Centre PRIME, Nijmegen, The Netherlands. .,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
19
|
Chen CW, Pavlova JA, Lukianov DA, Tereshchenkov AG, Makarov GI, Khairullina ZZ, Tashlitsky VN, Paleskava A, Konevega AL, Bogdanov AA, Osterman IA, Sumbatyan NV, Polikanov YS. Binding and Action of Triphenylphosphonium Analog of Chloramphenicol upon the Bacterial Ribosome. Antibiotics (Basel) 2021; 10:390. [PMID: 33916420 PMCID: PMC8066774 DOI: 10.3390/antibiotics10040390] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/20/2022] Open
Abstract
Chloramphenicol (CHL) is a ribosome-targeting antibiotic that binds to the peptidyl transferase center (PTC) of the bacterial ribosome and inhibits peptide bond formation. As an approach for modifying and potentially improving the properties of this inhibitor, we explored ribosome binding and inhibitory properties of a semi-synthetic triphenylphosphonium analog of CHL-CAM-C4-TPP. Our data demonstrate that this compound exhibits a ~5-fold stronger affinity for the bacterial ribosome and higher potency as an in vitro protein synthesis inhibitor compared to CHL. The X-ray crystal structure of the Thermus thermophilus 70S ribosome in complex with CAM-C4-TPP reveals that, while its amphenicol moiety binds at the PTC in a fashion identical to CHL, the C4-TPP tail adopts an extended propeller-like conformation within the ribosome exit tunnel where it establishes multiple hydrophobic Van der Waals interactions with the rRNA. The synthesized compound represents a promising chemical scaffold for further development by medicinal chemists because it simultaneously targets the two key functional centers of the bacterial ribosome-PTC and peptide exit tunnel.
Collapse
Affiliation(s)
- Chih-Wei Chen
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Julia A. Pavlova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (J.A.P.); (Z.Z.K.); (V.N.T.); (A.A.B.)
| | - Dmitrii A. Lukianov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 143028 Skolkovo, Russia;
| | - Andrey G. Tereshchenkov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Gennady I. Makarov
- Laboratory of Multiscale Modeling of Multicomponent Materials, South Ural State University, 454080 Chelyabinsk, Russia;
| | - Zimfira Z. Khairullina
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (J.A.P.); (Z.Z.K.); (V.N.T.); (A.A.B.)
| | - Vadim N. Tashlitsky
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (J.A.P.); (Z.Z.K.); (V.N.T.); (A.A.B.)
| | - Alena Paleskava
- Petersburg Nuclear Physics Institute, National Research Center (NRC) “Kurchatov Institute”, 188300 Gatchina, Russia; (A.P.); (A.L.K.)
- Peter the Great St.Petersburg Polytechnic University, 195251 Saint Petersburg, Russia
| | - Andrey L. Konevega
- Petersburg Nuclear Physics Institute, National Research Center (NRC) “Kurchatov Institute”, 188300 Gatchina, Russia; (A.P.); (A.L.K.)
- Peter the Great St.Petersburg Polytechnic University, 195251 Saint Petersburg, Russia
- National Research Center (NRC) “Kurchatov Institute”, 123182 Moscow, Russia
| | - Alexey A. Bogdanov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (J.A.P.); (Z.Z.K.); (V.N.T.); (A.A.B.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Ilya A. Osterman
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (J.A.P.); (Z.Z.K.); (V.N.T.); (A.A.B.)
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 143028 Skolkovo, Russia;
| | - Natalia V. Sumbatyan
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (J.A.P.); (Z.Z.K.); (V.N.T.); (A.A.B.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Yury S. Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
20
|
Qu X, Guo S, Yan L, Zhu H, Li H, Shi Z. TNFα-Erk1/2 signaling pathway-regulated SerpinE1 and SerpinB2 are involved in lipopolysaccharide-induced porcine granulosa cell proliferation. Cell Signal 2020; 73:109702. [PMID: 32619562 DOI: 10.1016/j.cellsig.2020.109702] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023]
Abstract
Lipopolysaccharide (LPS) is an inhibitory factor that causes hormonal imbalance and subsequently affects ovarian function and fertility in mammals. Previous studies have shown that the exposure of granulosa cells (GC) to LPS leads to steroidogenesis dysfunction. However, the effects of LPS on the viability of GC remain largely unclear. In the present study, we aimed to address this question and unveil the underlying molecular mechanisms using cultured porcine GC. Results showed that GC proliferation and tumor necrosis factor α (TNFα) secretion were significantly increased after exposure to LPS, and these effects were completely reversed by blocking the TNFα sheddase, ADAM17. Moreover, GC proliferation induced by LPS was mimicked by treatment with recombinant TNFα. In addition, SerpinE1 and SerpinB2 expression levels increased in GC after treatment with LPS or recombinant TNFα, whereas blocking the Erk1/2 pathway completely abolished these effects and also inhibited GC proliferation. Further, consistent with the effects of blocking the Erk1/2 pathway, cell proliferation was completely inhibited by knocking down SerpinE1 or SerpinB2 in the presence of LPS or recombinant TNFα. Mitochondrial membrane potential (MMP) polarization in GC was increased by LPS or recombinant TNFα treatment, and these changes were completely negated by Erk1/2 inhibition, but not by SerpinE1 or SerpinB2 knockdown. Taken together, these results suggested that the TNFα-mediated upregulation of SerpinE1 and SerpinB2, through activation of the Erk1/2 pathway plays a crucial role in LPS-stimulated GC proliferation, and the increase in GC MMP may synergistically influence this process.
Collapse
Affiliation(s)
- Xiaolu Qu
- College of Animal Science and technology, Jilin Agricultural University, Changchun 130118, China; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Key laboratory of Animal Breeding and Reproduction, Nanjing 210014, China
| | - Shuangshuang Guo
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Key laboratory of Animal Breeding and Reproduction, Nanjing 210014, China
| | - Leyan Yan
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Key laboratory of Animal Breeding and Reproduction, Nanjing 210014, China
| | - Huanxi Zhu
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Key laboratory of Animal Breeding and Reproduction, Nanjing 210014, China
| | - Hui Li
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Key laboratory of Animal Breeding and Reproduction, Nanjing 210014, China.
| | - Zhendan Shi
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Key laboratory of Animal Breeding and Reproduction, Nanjing 210014, China.
| |
Collapse
|
21
|
Panuzzo C, Jovanovski A, Pergolizzi B, Pironi L, Stanga S, Fava C, Cilloni D. Mitochondria: A Galaxy in the Hematopoietic and Leukemic Stem Cell Universe. Int J Mol Sci 2020; 21:ijms21113928. [PMID: 32486249 PMCID: PMC7312164 DOI: 10.3390/ijms21113928] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 12/17/2022] Open
Abstract
Mitochondria are the main fascinating energetic source into the cells. Their number, shape, and dynamism are controlled by the cell’s type and current behavior. The perturbation of the mitochondrial inward system via stress response and/or oncogenic insults could activate several trafficking molecular mechanisms with the intention to solve the problem. In this review, we aimed to clarify the crucial pathways in the mitochondrial system, dissecting the different metabolic defects, with a special emphasis on hematological malignancies. We investigated the pivotal role of mitochondria in the maintenance of hematopoietic stem cells (HSCs) and their main alterations that could induce malignant transformation, culminating in the generation of leukemic stem cells (LSCs). In addition, we presented an overview of LSCs mitochondrial dysregulated mechanisms in terms of (1) increasing in oxidative phosphorylation program (OXPHOS), as a crucial process for survival and self-renewal of LSCs,(2) low levels of reactive oxygen species (ROS), and (3) aberrant expression of B-cell lymphoma 2 (Bcl-2) with sustained mitophagy. Furthermore, these peculiarities may represent attractive new “hot spots” for mitochondrial-targeted therapy. Finally, we remark the potential of the LCS metabolic effectors to be exploited as novel therapeutic targets.
Collapse
Affiliation(s)
- Cristina Panuzzo
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
- Correspondence: (C.P.); (D.C.)
| | - Aleksandar Jovanovski
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
| | - Barbara Pergolizzi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
| | - Lucrezia Pironi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
| | - Serena Stanga
- Department of Neuroscience Rita Levi Montalcini, 10124 Turin, Italy;
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, Italy
| | - Carmen Fava
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (A.J.); (B.P.); (L.P.); (C.F.)
- Correspondence: (C.P.); (D.C.)
| |
Collapse
|
22
|
Gupta A, Ökesli-Armlovich A, Morgens D, Bassik MC, Khosla C. A genome-wide analysis of targets of macrolide antibiotics in mammalian cells. J Biol Chem 2020; 295:2057-2067. [PMID: 31915244 DOI: 10.1074/jbc.ra119.010770] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 01/05/2020] [Indexed: 01/04/2023] Open
Abstract
Macrolide antibiotics, such as erythromycin and josamycin, are natural polyketide products harboring 14- to 16-membered macrocyclic lactone rings to which various sugars are attached. These antibiotics are used extensively in the clinic because of their ability to inhibit bacterial protein synthesis. More recently, some macrolides have been shown to also possess anti-inflammatory and other therapeutic activities in mammalian cells. To better understand the targets and effects of this drug class in mammalian cells, we used a genome-wide shRNA screen in K562 cancer cells to identify genes that modulate cellular sensitivity to josamycin. Among the most sensitizing hits were proteins involved in mitochondrial translation and the mitochondrial unfolded protein response, glycolysis, and the mitogen-activated protein kinase signaling cascade. Further analysis revealed that cells treated with josamycin or other antibacterial agents exhibited impaired oxidative phosphorylation and metabolic shifts to glycolysis. Interestingly, we observed that knockdown of the mitogen-activated protein kinase kinase kinase 4 (MAP3K4) gene, which contributes to p38 mitogen-activated protein kinase signaling, sensitized cells only to josamycin but not to other antibacterial agents. There is a growing interest in better characterizing the therapeutic effects and toxicities of antibiotics in mammalian cells to guide new applications in both cellular and clinical studies. To our knowledge, this is the first report of an unbiased genome-wide screen to investigate the effects of a clinically used antibiotic on human cells.
Collapse
Affiliation(s)
- Amita Gupta
- Department of Chemical Engineering, Stanford University, Stanford, California 94305; Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, California 94305
| | - Aye Ökesli-Armlovich
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, California 94305; Department of Chemistry, Stanford University, Stanford, California 94305
| | - David Morgens
- Department of Genetics, Stanford University, Stanford, California 94305
| | - Michael C Bassik
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, California 94305; Department of Genetics, Stanford University, Stanford, California 94305
| | - Chaitan Khosla
- Department of Chemical Engineering, Stanford University, Stanford, California 94305; Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, California 94305; Department of Chemistry, Stanford University, Stanford, California 94305; Department of Biochemistry, Stanford University, Stanford, California 94305.
| |
Collapse
|
23
|
Zada SL, Baruch BB, Simhaev L, Engel H, Fridman M. Chemical Modifications Reduce Auditory Cell Damage Induced by Aminoglycoside Antibiotics. J Am Chem Soc 2020; 142:3077-3087. [PMID: 31958945 DOI: 10.1021/jacs.9b12420] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although aminoglycoside antibiotics are effective against Gram-negative infections, these drugs often cause irreversible hearing damage. Binding to the decoding site of the eukaryotic ribosomes appears to result in ototoxicity, but there is evidence that other effects are involved. Here, we show how chemical modifications of apramycin and geneticin, considered among the least and most toxic aminoglycosides, respectively, reduce auditory cell damage. Using molecular dynamics simulations, we studied how modified aminoglycosides influence the essential freedom of movement of the decoding site of the ribosome, the region targeted by aminoglycosides. By determining the ratio of a protein translated in mitochondria to that of a protein translated in the cytoplasm, we showed that aminoglycosides can paradoxically elevate rather than reduce protein levels. We showed that certain aminoglycosides induce rapid plasma membrane permeabilization and that this nonribosomal effect can also be reduced through chemical modifications. The results presented suggest a new paradigm for the development of safer aminoglycoside antibiotics.
Collapse
Affiliation(s)
- Sivan Louzoun Zada
- School of Chemistry, Raymond and Beverley Sackler Faculty of Exact Sciences , Tel Aviv University , Tel Aviv , Israel , 6997801
| | - Bar Ben Baruch
- School of Chemistry, Raymond and Beverley Sackler Faculty of Exact Sciences , Tel Aviv University , Tel Aviv , Israel , 6997801
| | - Luba Simhaev
- Blavatnik Center for Drug Discovery , Tel Aviv University , Tel Aviv , 6997801 , Israel
| | - Hamutal Engel
- Blavatnik Center for Drug Discovery , Tel Aviv University , Tel Aviv , 6997801 , Israel
| | - Micha Fridman
- School of Chemistry, Raymond and Beverley Sackler Faculty of Exact Sciences , Tel Aviv University , Tel Aviv , Israel , 6997801
| |
Collapse
|
24
|
Wu X, Cai Y, Lu S, Xu K, Shi X, Yang L, Huang Z, Xu P. Intra-articular Injection of Chloramphenicol Reduces Articular Cartilage Degeneration in a Rabbit Model of Osteoarthritis. Clin Orthop Relat Res 2019; 477:2785-2797. [PMID: 31764352 PMCID: PMC6907289 DOI: 10.1097/corr.0000000000001016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 10/08/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is characterized by degeneration of articular cartilage. Studies have found that enhancement of autophagy, an intracellular catabolic process, may limit the pathologic progression of OA. Chloramphenicol is a potent activator of autophagy; however, the effects of chloramphenicol on articular cartilage are unknown. QUESTIONS/PURPOSES Using human OA knee chondrocytes in vitro, we asked, does chloramphenicol (1) activate autophagy in chondrocytes; (2) protect chondrocytes from IL-1β-induced apoptosis; and (3) reduce the expression of matrix metallopeptidase (MMP)-13 and IL-6 (markers associated with articular cartilage degradation and joint inflammation). Using an in vivo rabbit model of OA, we asked, does an intra-articular injection of chloramphenicol in the knee (4) induce autophagy; (5) reduce OA severity; and (6) reduce MMP-13 expression? METHODS Human chondrocytes were extracted from 10 men with OA undergoing TKA. After treatment with 25 μg/mL, 50 μg/mL, or 100μg/mL chloramphenicol, the autophagy of chondrocytes was detected with Western blotting, transmission electron microscopy, or an autophagy detection kit. There were four groups in our study: one group was untreated, one was treated with 100 μg/mL chloramphenicol, another was treated with 10 ng/mL of IL-1β, and the final group was treated with 10 ng/mL of IL-1β and 100 μg/mL of chloramphenicol. All groups were treated for 48 hours; cell apoptosis was detected with Western blotting and flow cytometry. Inflammation marker IL-6 in the cell culture supernatant was detected with an ELISA. Articular cartilage degradation-related enzyme MMP-13 was analyzed with Western blotting. A rabbit model of OA was induced by intra-articular injection of type II collagenase in 20 male 3-month-old New Zealand White rabbits' right hind leg knees; the left hind leg knees served as controls. Rabbits were treated by intra-articular injection of saline or chloramphenicol once a week for 8 weeks. Autophagy of the articular cartilage was detected with Western blotting and transmission electron microscopy. Degeneration of articular cartilage was analyzed with Safranin O-fast green staining and the semi-quantitative index Osteoarthritis Research Society International (OARSI) grading system. Degeneration of articular cartilage was evaluated using the OARSI grading system. The expression of MMP-13 in articular cartilage was detected with immunohistochemistry. RESULTS Chloramphenicol activated autophagy in vitro in the chondrocytes of humans with OA and in an in vivo rabbit model of OA. Chloramphenicol inhibited IL-1-induced apoptosis (flow cytometry results with chloramphenicol, 25.33 ± 3.51%, and without chloramphenicol, 44.00 ± 3.61%, mean difference, 18.67% [95% CI 10.60 to 26.73]; p = 0.003) and the production of proinflammatory cytokine IL-6 (ELISA results, with chloramphenicol, 720.00 ± 96.44 pg/mL, without chloramphenicol, 966.67 ± 85.05 pg/mL; mean difference 74.24 pg/mL [95% CI 39.28 to 454.06]; p = 0.029) in chondrocytes. After chloramphenicol treatment, the severity of cartilage degradation was reduced in the treatment group (OARSI 6.80 ± 2.71) compared with the control group (12.30 ± 2.77), (mean difference 5.50 [95% CI 1.50 to 9.50]; p = 0.013). Furthermore, chloramphenicol treatment also decreased the production of MMP-13 in vitro and in vivo. CONCLUSIONS Chloramphenicol reduced the severity of cartilage degradation in a type II collagen-induced rabbit model of OA, which may be related to induction of autophagy and inhibition of MMP-13 and IL-6. CLINICAL RELEVANCE Our study suggests that an intra-articular injection of chloramphenicol may reduce degeneration of articular cartilage and that induction of autophagy may be a method for treating OA. The animal model we used was type II collagen-induced OA, which was different from idiopathic OA and post-traumatic OA. Therefore, we need to use other types of OA models (idiopathic OA or a surgically induced OA model) to further verify its effect, and the side effects of chloramphenicol also need to be considered, such as myelosuppression.
Collapse
Affiliation(s)
- Xiaoqing Wu
- X. Wu, Y. Cai, K. Xu, L. Yang, P. Xu, Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, China S. Lu, Department of Genetics and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, China X. Shi, Department of Oncology, Qianfoshan Hospital, Shandong University, Jinan, China Z. Huang, Department of Joint Surgery, Shiquan County Hospital of Traditional Chinese Medicine, Ankang, China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Sun AW, Bulterys PL, Bartberger MD, Jorth PA, O'Boyle BM, Virgil SC, Miller JF, Stoltz BM. Incorporation of a chiral gem-disubstituted nitrogen heterocycle yields an oxazolidinone antibiotic with reduced mitochondrial toxicity. Bioorg Med Chem Lett 2019; 29:2686-2689. [PMID: 31383589 PMCID: PMC6711789 DOI: 10.1016/j.bmcl.2019.07.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 07/08/2019] [Accepted: 07/13/2019] [Indexed: 11/20/2022]
Abstract
gem-Disubstituted N-heterocycles are rarely found in drugs, despite their potential to improve the drug-like properties of small molecule pharmaceuticals. Linezolid, a morpholine heterocycle-containing oxazolidinone antibiotic, exhibits significant side effects associated with human mitochondrial protein synthesis inhibition. We synthesized a gem-disubstituted linezolid analogue that when compared to linezolid, maintains comparable (albeit slightly diminished) activity against bacteria, comparable in vitro physicochemical properties, and a decrease in undesired mitochondrial protein synthesis (MPS) inhibition. This research contributes to the structure-activity-relationship data surrounding oxazolidinone MPS inhibition, and may inspire investigations into the utility of gem-disubstituted N-heterocycles in medicinal chemistry.
Collapse
Affiliation(s)
- Alexander W Sun
- The Warren and Katharine Schlinger Laboratory for Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Philip L Bulterys
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Michael D Bartberger
- The Warren and Katharine Schlinger Laboratory for Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Peter A Jorth
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Brendan M O'Boyle
- The Warren and Katharine Schlinger Laboratory for Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Scott C Virgil
- The Warren and Katharine Schlinger Laboratory for Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Jeff F Miller
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Brian M Stoltz
- The Warren and Katharine Schlinger Laboratory for Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States.
| |
Collapse
|
26
|
Andjelković A, Mordas A, Bruinsma L, Ketola A, Cannino G, Giordano L, Dhandapani PK, Szibor M, Dufour E, Jacobs HT. Expression of the Alternative Oxidase Influences Jun N-Terminal Kinase Signaling and Cell Migration. Mol Cell Biol 2018; 38:e00110-18. [PMID: 30224521 PMCID: PMC6275184 DOI: 10.1128/mcb.00110-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/11/2018] [Accepted: 09/11/2018] [Indexed: 12/25/2022] Open
Abstract
Downregulation of Jun N-terminal kinase (JNK) signaling inhibits cell migration in diverse model systems. In Drosophila pupal development, attenuated JNK signaling in the thoracic dorsal epithelium leads to defective midline closure, resulting in cleft thorax. Here we report that concomitant expression of the Ciona intestinalis alternative oxidase (AOX) was able to compensate for JNK pathway downregulation, substantially correcting the cleft thorax phenotype. AOX expression also promoted wound-healing behavior and single-cell migration in immortalized mouse embryonic fibroblasts (iMEFs), counteracting the effect of JNK pathway inhibition. However, AOX was not able to rescue developmental phenotypes resulting from knockdown of the AP-1 transcription factor, the canonical target of JNK, nor its targets and had no effect on AP-1-dependent transcription. The migration of AOX-expressing iMEFs in the wound-healing assay was differentially stimulated by antimycin A, which redirects respiratory electron flow through AOX, altering the balance between mitochondrial ATP and heat production. Since other treatments affecting mitochondrial ATP did not stimulate wound healing, we propose increased mitochondrial heat production as the most likely primary mechanism of action of AOX in promoting cell migration in these various contexts.
Collapse
Affiliation(s)
- Ana Andjelković
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Amelia Mordas
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Lyon Bruinsma
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Annika Ketola
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Giuseppe Cannino
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Luca Giordano
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Praveen K Dhandapani
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Marten Szibor
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Eric Dufour
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Howard T Jacobs
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
27
|
Hoque SAM, Kawai T, Zhu Z, Shimada M. Mitochondrial Protein Turnover Is Critical for Granulosa Cell Proliferation and Differentiation in Antral Follicles. J Endocr Soc 2018; 3:324-339. [PMID: 30652133 PMCID: PMC6330174 DOI: 10.1210/js.2018-00329] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 11/27/2018] [Indexed: 12/28/2022] Open
Abstract
Granulosa cell (GC) proliferation is essential for follicular development. FSH is a key factor in GC proliferation, and a continuous supply of high levels of ATP is necessary for cell proliferation. However, genes encoding proteins of the glycolytic pathways are poorly expressed in GCs. Therefore, we hypothesized that mitochondrial gene expression and protein synthesis play a primary role in ATP production during GC proliferation. To test this hypothesis, we performed an in vivo study of GCs collected from 23-day-old mice ovaries with or without equine chorionic gonadotropin (eCG) priming. It was observed that mitochondrial activity with membrane potential, expression of protein-coding genes (Nd1-6, Cytb, Atpase6,8) and transcription-related genes (Polrmt, Tfam, Tfb2m), copy number of mitochondrial (mt-)DNA, and protein synthesis were increased in GCs after 24 hours of eCG injection and mostly maintained elevated up to 48 hours. Therefore, we performed in vitro culture of GCs in DMEM medium supplemented with FSH, testosterone, and serum and containing different glucose concentrations with or without d-chloramphenicol (CRP) for 24 hours. GC proliferation and ATP production were observed to be independent of glucose concentration. Furthermore, FSH-induced mitochondrial activity with membrane potential, ATP content, BrdU-incorporated cell proliferation, intensity of mt-ND1 and mt-ND6 proteins, and expressions of marker genes for proliferation and differentiation were significantly decreased by CRP treatment. These results revealed the crucial role of mitochondria in the supply of ATP and the necessity of mitochondrial gene expression and protein synthesis in not only the proliferation but also the differentiation of GCs during follicular development.
Collapse
Affiliation(s)
- S A Masudul Hoque
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Tomoko Kawai
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Zhendong Zhu
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,College of Animal Science and Technology, Northwest A&F University, Shaanxi, China
| | - Masayuki Shimada
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| |
Collapse
|
28
|
Yu JJ, Lee DH, Gallagher SP, Kenney MC, Boisvert CJ. Mitochondrial Impairment in Antibiotic Induced Toxic Optic Neuropathies. Curr Eye Res 2018; 43:1199-1204. [DOI: 10.1080/02713683.2018.1504086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Jeffrey J. Yu
- Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Daniel H. Lee
- Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Shea P. Gallagher
- Department of Ophthalmology, University of California, Irvine, CA, USA
| | | | | |
Collapse
|
29
|
Kogachi K, Ter-Zakarian A, Asanad S, Sadun A, Karanjia R. Toxic medications in Leber's hereditary optic neuropathy. Mitochondrion 2018; 46:270-277. [PMID: 30081212 DOI: 10.1016/j.mito.2018.07.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/23/2018] [Accepted: 07/31/2018] [Indexed: 12/18/2022]
Abstract
Leber's hereditary optic neuropathy (LHON) is a maternally inherited mitochondrial disorder characterized by acute bilateral vision loss. The pathophysiology involves reactive oxygen species (ROS), which can be affected by medications. This article reviews the evidence for medications with demonstrated and theoretical effects on mitochondrial function, specifically in relation to increased ROS production. The data reviewed provides guidance when selecting medications for individuals with LHON mutations (carriers) and are susceptible to conversion to affected. However, as with all medications, the proven benefits of these therapies must be weighed against, in some cases, purely theoretical risks for this unique patient population.
Collapse
Affiliation(s)
- Kaitlin Kogachi
- Doheny Eye Institute, 1355 San Pablo Street, Los Angeles, CA 90033, USA.
| | - Anna Ter-Zakarian
- Doheny Eye Institute, 1355 San Pablo Street, Los Angeles, CA 90033, USA
| | - Samuel Asanad
- Doheny Eye Institute, 1355 San Pablo Street, Los Angeles, CA 90033, USA; Doheny Eye Center, Department of Ophthalmology, David Geffen School of Medicine at UCLA, 800 South Fairmount Avenue, Suite 215, Pasadena, CA 91105, USA
| | - Alfredo Sadun
- Doheny Eye Institute, 1355 San Pablo Street, Los Angeles, CA 90033, USA; Doheny Eye Center, Department of Ophthalmology, David Geffen School of Medicine at UCLA, 800 South Fairmount Avenue, Suite 215, Pasadena, CA 91105, USA
| | - Rustum Karanjia
- Doheny Eye Institute, 1355 San Pablo Street, Los Angeles, CA 90033, USA; Doheny Eye Center, Department of Ophthalmology, David Geffen School of Medicine at UCLA, 800 South Fairmount Avenue, Suite 215, Pasadena, CA 91105, USA; The Ottawa Eye Institute, University of Ottawa, 501 Smyth Rd, Ottawa, ON K1H 8M2, Canada; Ottawa Hospital Research Institute, 1053 Carling Avenue, Ottawa, ON K1Y 4E9, Canada
| |
Collapse
|
30
|
Byrnes J, Ganetzky R, Lightfoot R, Tzeng M, Nakamaru-Ogiso E, Seiler C, Falk MJ. Pharmacologic modeling of primary mitochondrial respiratory chain dysfunction in zebrafish. Neurochem Int 2018; 117:23-34. [PMID: 28732770 PMCID: PMC5773416 DOI: 10.1016/j.neuint.2017.07.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 07/11/2017] [Accepted: 07/16/2017] [Indexed: 02/07/2023]
Abstract
Mitochondrial respiratory chain (RC) disease is a heterogeneous and highly morbid group of energy deficiency disorders for which no proven effective therapies exist. Robust vertebrate animal models of primary RC dysfunction are needed to explore the effects of variation in RC disease subtypes, tissue-specific manifestations, and major pathogenic factors contributing to each disorder, as well as their pre-clinical response to therapeutic candidates. We have developed a series of zebrafish (Danio rerio) models that inhibit, to variable degrees, distinct aspects of RC function, and enable quantification of animal development, survival, behaviors, and organ-level treatment effects as well as effects on mitochondrial biochemistry and physiology. Here, we characterize four pharmacologic inhibitor models of mitochondrial RC dysfunction in early larval zebrafish, including rotenone (complex I inhibitor), azide (complex IV inhibitor), oligomycin (complex V inhibitor), and chloramphenicol (mitochondrial translation inhibitor that leads to multiple RC complex dysfunction). A range of concentrations and exposure times of each RC inhibitor were systematically evaluated on early larval development, animal survival, integrated behaviors (touch and startle responses), organ physiology (brain death, neurologic tone, heart rate), and fluorescence-based analyses of mitochondrial physiology in zebrafish skeletal muscle. Pharmacologic RC inhibitor effects were validated by spectrophotometric analysis of Complex I, II and IV enzyme activities, or relative quantitation of ATP levels in larvae. Outcomes were prioritized that utilize in vivo animal imaging and quantitative behavioral assessments, as may optimally inform the translational potential of pre-clinical drug screens for future clinical study in human mitochondrial disease subjects. The RC complex inhibitors each delayed early embryo development, with short-term exposures of these three agents or chloramphenicol from 5 to 7 days post fertilization also causing reduced larval survival and organ-specific defects ranging from brain death, behavioral and neurologic alterations, reduced mitochondrial membrane potential in skeletal muscle (rotenone), and/or cardiac edema with visible blood pooling (oligomycin). Remarkably, we demonstrate that treating animals with probucol, a nutrient-sensing signaling network modulating drug that has been shown to yield therapeutic effects in a range of other RC disease cellular and animal models, both prevented acute rotenone-induced brain death in zebrafish larvae, and significantly rescued early embryo developmental delay from either rotenone or oligomycin exposure. Overall, these zebrafish pharmacologic RC function inhibition models offer a unique opportunity to gain novel insights into diverse developmental, survival, organ-level, and behavioral defects of varying severity, as well as their individual response to candidate therapies, in a highly tractable and cost-effective vertebrate animal model system.
Collapse
Affiliation(s)
- James Byrnes
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Rebecca Ganetzky
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Richard Lightfoot
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Michael Tzeng
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Eiko Nakamaru-Ogiso
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Christoph Seiler
- Aquatics Core Facility, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, United States
| | - Marni J Falk
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States.
| |
Collapse
|
31
|
Han SY, Jeong YJ, Choi Y, Hwang SK, Bae YS, Chang YC. Mitochondrial dysfunction induces the invasive phenotype, and cell migration and invasion, through the induction of AKT and AMPK pathways in lung cancer cells. Int J Mol Med 2018; 42:1644-1652. [PMID: 29916527 DOI: 10.3892/ijmm.2018.3733] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 05/08/2018] [Indexed: 11/05/2022] Open
Abstract
Mitochondria are well known for their important roles in oxidative phosphorylation, amino acid metabolism, fatty acid oxidation and ion homeostasis. Although the effects of mitochondrial dysfunction on tumorigenesis in various cancer cells have been reported, the correlation between mitochondrial dysfunction and epithelial‑to‑mesenchymal transition (EMT) in lung cancer development and metastasis has not been well elucidated. In the present study, the effects of mitochondrial dysfunction on EMT and migration in lung cancer cells were investigated using inhibitors of mitochondrial respiration, oligomycin A and antimycin A. Oligomycin A and antimycin A induced distinct mesenchymal‑like morphological features in H23, H1793 and A549 lung cancer cells. In addition, they decreased the expression levels of the epithelial marker protein E‑cadherin, but increased the expression levels of the mesenchymal marker proteins Vimentin, Snail and Slug. The results of immunofluorescence staining indicated that oligomycin A and antimycin A downregulated cortical E‑cadherin expression and upregulated the expression of Vimentin. In addition, oligomycin A and antimycin A increased the migration and invasion of A549 lung cancer cells, and promoted the expression levels of phosphorylated (p)‑protein kinase B (AKT) and p‑AMP‑activated protein kinase (AMPK). Notably, the production of reactive oxygen species by oligomycin A and antimycin A did not affect the expression of EMT protein markers. Conversely, treatment with the AKT inhibitor wortmannin and the AMPK inhibitor Compound C upregulated E‑cadherin and downregulated Vimentin expression. These results suggested that oligomycin A and antimycin A may induce migration and invasion of lung cancer cells by inducing EMT via the upregulation of p‑AKT and p‑AMPK expression.
Collapse
Affiliation(s)
- Si-Yoon Han
- Department of Cell Biology, Catholic University of Daegu School of Medicine, Daegu 42472, Republic of Korea
| | - Yun-Jeong Jeong
- Department of Cell Biology, Catholic University of Daegu School of Medicine, Daegu 42472, Republic of Korea
| | - Yongsoo Choi
- Systems Biotechnology Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Gangwon 25451, Republic of Korea
| | - Soon-Kyung Hwang
- Department of Cell Biology, Catholic University of Daegu School of Medicine, Daegu 42472, Republic of Korea
| | - Young-Seuk Bae
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group,College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Young-Chae Chang
- Department of Cell Biology, Catholic University of Daegu School of Medicine, Daegu 42472, Republic of Korea
| |
Collapse
|
32
|
Mahamat O, Christopher T, Gilbert A, Teke Gerald N, Albert K. Immunological In Vivo and In Vitro Investigations of Aqueous Extract of Stem Bark of Pterocarpus erinaceus Poir (Fabaceae). Am J Med Sci 2018; 356:56-63. [PMID: 29807644 DOI: 10.1016/j.amjms.2018.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 02/08/2018] [Accepted: 02/15/2018] [Indexed: 01/26/2023]
Abstract
BACKGROUND Macrophages are the first cells to recognize invading foreign bodies and are central to cell mediated and humoral immunity. Therefore, the activation of macrophages is a key event for effective innate and adaptive immunity. Pterocarpus erinaceus has been reported to control infectious diseases, but the mechanism remains to be elucidated. In this study, we demonstrated the immune-modulatory effect of aqueous extract of P. erinaceus using human macrophages and lymphocytes, as well as mice. METHODS Hot water was used to extract P. erinaceus from the stem bark. Its effect on lymphocytes was measured by evaluating proliferative response and delayed hypersensitivity. Phagocytic activity of macrophages were measured based on neutral red uptake assay, nitric oxide production and myeloperoxidase and phosphatase acid activity. Hematopoietic and infectious activities were analyzed using the effect on infectious stress and chloramphenicol-induced leucopenic mice model. RESULTS Aqueous extract showed stronger stimulatory effects on the neutral red uptake, production of nitric oxide and phosphatase acid activity in lipopolysaccharide-activated macrophages. In addition, aqueous extract significantly stimulated the proliferation of phytohemagglutinin-activated lymphocytes, enhanced delayed hypersensitivity response to erythrocytes and attenuated infection-induced fever. Furthermore, aqueous extract also significantly increased the rate of recovery of white blood cell levels in chloramphenicol-induced leucopenia mice. CONCLUSIONS The results suggest that aqueous extract of P. erinaceus stem bark is able to modulate the immune system and has potential effects in clinical conditions when an immune-enhancing and anti-infectious effect is desired.
Collapse
Affiliation(s)
- Oumar Mahamat
- Department of Biological Sciences, Faculty of Sciences, University of Bamenda, Bamenda, Cameroon.
| | - Tume Christopher
- Department of Biochemistry, Faculty of Sciences, University of Dschang, Dschang, Cameroon
| | - Ateufack Gilbert
- Department of Animal Biology, Faculty of Sciences, University of Dschang, Dschang, Cameroon
| | - Ngo Teke Gerald
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Bamenda, Bamenda, Cameroon
| | - Kamanyi Albert
- Department of Animal Biology, Faculty of Sciences, University of Dschang, Dschang, Cameroon
| |
Collapse
|
33
|
Mitochondrial biogenesis and metabolic hyperactivation limits the application of MTT assay in the estimation of radiation induced growth inhibition. Sci Rep 2018; 8:1531. [PMID: 29367754 PMCID: PMC5784148 DOI: 10.1038/s41598-018-19930-w] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/05/2018] [Indexed: 01/19/2023] Open
Abstract
Metabolic viability based high throughput assays like MTT and MTS are widely used in assessing the cell viability. However, alteration in both mitochondrial content and metabolism can influence the metabolic viability of cells and radiation is a potential mitochondrial biogenesis inducer. Therefore, we tested if MTT assay is a true measure of radiation induced cell death in widely used cell lines. Radiation induced cellular growth inhibition was performed by enumerating cell numbers and metabolic viability using MTT assay at 24 and 48 hours (hrs) after exposure. The extent of radiation induced reduction in cell number was found to be larger than the decrease in MTT reduction in all the cell lines tested. We demonstrated that radiation induces PGC-1α and TFAM to stimulate mitochondrial biogenesis leading to increased levels of SDH-A and enhanced metabolic viability. Radiation induced disturbance in calcium (Ca2+) homeostasis also plays a crucial role by making the mitochondria hyperactive. These findings suggest that radiation induces mitochondrial biogenesis and hyperactivation leading to increased metabolic viability and MTT reduction. Therefore, conclusions drawn on radiation induced growth inhibition based on metabolic viability assays are likely to be erroneous as it may not correlate with growth inhibition and/or loss of clonogenic survival.
Collapse
|
34
|
Tian F, Wang C, Tang M, Li J, Cheng X, Zhang S, Ji D, Huang Y, Li H. The antibiotic chloramphenicol may be an effective new agent for inhibiting the growth of multiple myeloma. Oncotarget 2018; 7:51934-51942. [PMID: 27437770 PMCID: PMC5239525 DOI: 10.18632/oncotarget.10623] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 06/29/2016] [Indexed: 01/08/2023] Open
Abstract
Chloramphenicol is an old antibiotic that also inhibits mammalian mitochondrial protein synthesis. Our studies demonstrated that chloramphenicol is highly cytotoxic to myeloma cells, acting in a dose- and time-dependent manner. Chloramphenicol sharply suppressed ATP levels in myeloma cells at concentrations ≥ 25 μg/mL. Colorimetric and clonogenic assays indicate that chloramphenicol inhibits growth of myeloma cell lines at concentrations ≥ 50 μg/mL, and inhibits primary myeloma cell growth at concentrations ≥ 25 μg/mL. Flow cytometry and Western blotting showed that chloramphenicol induces myeloma cell apoptosis at concentrations ≥ 50 μg/mL. Chloramphenicol increased levels of cytochrome c, cleaved caspase-9 and cleaved caspase-3, suggesting that myeloma cell apoptosis occurs through the mitochondria-mediated apoptosis pathway. It thus appears chloramphenicol is not only an old antibiotic, it is also a potential cytotoxic agent effective against myeloma cells. This suggests chloramphenicol may be an effective "new" drug for the treatment of myeloma.
Collapse
Affiliation(s)
- Faqing Tian
- Department of Hematology, Longgang District People's Hospital of Shenzhen, Guangdong, China
| | - Chunyan Wang
- Department of Rheumatology, Lanzhou University Second Hospital, Gansu, China
| | - Meiqin Tang
- Department of Hematology, Longgang District People's Hospital of Shenzhen, Guangdong, China
| | - Juheng Li
- Department of Hematology, Longgang District People's Hospital of Shenzhen, Guangdong, China
| | - Xiaohui Cheng
- Department of Hematology, Longgang District People's Hospital of Shenzhen, Guangdong, China
| | - Sihan Zhang
- Department of Rheumatology, Longgang District People's Hospital of Shenzhen, Guangdong, China
| | - Delan Ji
- Department of Hematology, Longgang District People's Hospital of Shenzhen, Guangdong, China
| | - Yingcai Huang
- Department of Hematology, Longgang District People's Hospital of Shenzhen, Guangdong, China
| | - Huiqing Li
- Department of Hematology, Longgang District People's Hospital of Shenzhen, Guangdong, China
| |
Collapse
|
35
|
Fujii S, Muraoka S, Miyamoto A, Sakurai K. [Linezolid-induced Apoptosis through Mitochondrial Damage and Role of Superoxide Dismutase-1 in Human Monocytic Cell Line U937]. YAKUGAKU ZASSHI 2018; 138:73-81. [PMID: 29311467 DOI: 10.1248/yakushi.17-00133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cytopenia is a major adverse event associated with linezolid therapy. The objective of this study was to examine whether the cytotoxicity of linezolid to eukaryotic cells was associated with mitochondrial dysfunction and apoptosis-like cell death in human leukemic monocyte lymphoma cell line U937. Apoptosis-like cell death was clearly observed when cells were incubated with linezolid, depending on the duration and linezolid concentration. Mitochondrial membrane potential of cells treated with linezolid collapsed in a short period of time, but the number of mitochondria did not decrease. Cytotoxicity of linezolid was relieved by the knockdown of superoxide dismutase-1 in U937 cells. On the other hand, no autophagy was observed in cells treated with linezolid. These results suggest that mitochondrial damages would be linked to the induction of apoptosis in U937 cells treated with linezolid and that its mechanism does not involve autophagy.
Collapse
Affiliation(s)
- Satoshi Fujii
- Department of Hospital Pharmacy, Sapporo Medical University Hospital.,Department of Life Science, Hokkaido Pharmaceutical University School of Pharmacy
| | - Sanae Muraoka
- Department of Life Science, Hokkaido Pharmaceutical University School of Pharmacy
| | - Atsushi Miyamoto
- Department of Hospital Pharmacy, Sapporo Medical University Hospital
| | - Koichi Sakurai
- Department of Life Science, Hokkaido Pharmaceutical University School of Pharmacy
| |
Collapse
|
36
|
Tsai CH, Li CH, Cheng YW, Lee CC, Liao PL, Lin CH, Huang SH, Kang JJ. The inhibition of lung cancer cell migration by AhR-regulated autophagy. Sci Rep 2017; 7:41927. [PMID: 28195146 PMCID: PMC5307309 DOI: 10.1038/srep41927] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 01/03/2017] [Indexed: 12/11/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that is highly expressed in multiple organs and tissues. Whereas AhR mediates the metabolism of xenobiotic and endogenous compounds, its novel function in cancer epithelial-mesenchymal transition (EMT) remains controversial. Autophagy also participates in tumour progression through its functions in cell homeostasis and facilitates adaptation to EMT progression. In the present study, we found that AhR-regulated autophagy positively modulates EMT in non-small cell lung cancer cells. The motility of A549, H1299, and CL1-5 cells were correlated with different AhR expression levels. Invasive potential and cell morphology also changed when AhR protein expression was altered. Moreover, AhR levels exerted a contrasting effect on autophagy potential. Autophagy was higher in CL1-5 and H1299 cells with lower AhR levels than in A549 cells. Both AhR overexpression and autophagy inhibition decreased CL1-5 metastasis in vivo. Furthermore, AhR promoted BNIP3 ubiquitination for proteasomal degradation. AhR silencing in A549 cells also reduced BNIP3 ubiquitination. Taken together, these results provide a novel insight into the cross-linking between AhR and autophagy, we addressed the mechanistic BNIP3 modulation by endogenous AhR, which affect cancer cell EMT progression.
Collapse
Affiliation(s)
- Chi-Hao Tsai
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Hao Li
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wen Cheng
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chen-Chen Lee
- Department of Microbiology and Immunology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Po-Lin Liao
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Hui Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Shih-Hsuan Huang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jaw-Jou Kang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
37
|
Novel Detection Strategy To Rapidly Evaluate the Efficacy of Antichlamydial Agents. Antimicrob Agents Chemother 2017; 61:AAC.02202-16. [PMID: 27855081 DOI: 10.1128/aac.02202-16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 11/06/2016] [Indexed: 12/24/2022] Open
Abstract
Chlamydia trachomatis infections present a major heath burden worldwide. The conventional method used to detect C. trachomatis is laborious. In the present study, a novel strategy was utilized to evaluate the impact of antimicrobial agents on the growth of C. trachomatis and its expression of ompA promoter-driven green fluorescence protein (GFP). We demonstrate that this GFP reporter system gives a robust fluorescent display of C. trachomatis growth in human cervical epithelial cells and, further, that GFP production directly correlates to changes in ompA expression following sufficient exposure to antimicrobials. Validation with azithromycin, the first-line macrolide drug used for the treatment of C. trachomatis infection, highlights the advantages of this method over the traditional method because of its simplicity and versatility. The results indicate both that ompA is highly responsive to antimicrobials targeting the transcription and translation of C. trachomatis and that there is a correlation between changing GFP levels and C. trachomatis growth. This proof-of-concept study also reveals that the ompA-GFP system can be easily adapted to rapidly assess antimicrobial effectiveness in a high-throughput format.
Collapse
|
38
|
Stefano GB, Samuel J, Kream RM. Antibiotics May Trigger Mitochondrial Dysfunction Inducing Psychiatric Disorders. Med Sci Monit 2017; 23:101-106. [PMID: 28063266 PMCID: PMC5240889 DOI: 10.12659/msm.899478] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Clinical usage of several classes of antibiotics is associated with moderate to severe side effects due to the promotion of mitochondrial dysfunction. We contend that this may be due to perturbation of unique evolutionary relationships that link selective biochemical and molecular aspects of mitochondrial biology to conserved enzymatic processes derived from bacterial progenitors. Operationally, stereo-selective conformational matching between mitochondrial respiratory complexes, cytosolic and nuclear signaling complexes appears to support the conservation of a critically important set of chemical messengers required for existential regulation of homeostatic cellular processes. Accordingly, perturbation of normative mitochondrial function by select classes of antibiotics is certainly reflective of the high degree of evolutionary pressure designed to maintain ongoing bidirectional signaling processes between cellular compartments. These issues are of critical importance in evaluating potentially severe side effects of antibiotics on complex behavioral functions mediated by CNS neuronal groups. The CNS is extremely dependent on delivery of molecular oxygen for maintaining a required level of metabolic activity, as reflected by the high concentration of neuronal mitochondria. Thus, it is not surprising to find several distinct behavioral abnormalities conforming to established psychiatric criteria that are associated with antibiotic usage in humans. The manifestation of acute and/or chronic psychiatric conditions following antibiotic usage may provide unique insights into key etiological factors of major psychiatric syndromes that involve rundown of cellular bioenergetics via mitochondrial dysfunction. Thus, a potential window of opportunity exists for development of novel therapeutic agents targeting diminished mitochondrial function as a factor in severe behavioral disorders.
Collapse
Affiliation(s)
- George B Stefano
- Department of Research, MitoGenetics Research Institute, Farmingdale, NY, USA
| | - Joshua Samuel
- Department of Research, MitoGenetics Research Institute, Farmingdale, NY, USA
| | - Richard M Kream
- Department of Research, MitoGenetics Research Institute, Farmingdale, NY, USA
| |
Collapse
|
39
|
Dinos GP, Athanassopoulos CM, Missiri DA, Giannopoulou PC, Vlachogiannis IA, Papadopoulos GE, Papaioannou D, Kalpaxis DL. Chloramphenicol Derivatives as Antibacterial and Anticancer Agents: Historic Problems and Current Solutions. Antibiotics (Basel) 2016; 5:antibiotics5020020. [PMID: 27271676 PMCID: PMC4929435 DOI: 10.3390/antibiotics5020020] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 05/17/2016] [Accepted: 05/24/2016] [Indexed: 12/19/2022] Open
Abstract
Chloramphenicol (CAM) is the D-threo isomer of a small molecule, consisting of a p-nitrobenzene ring connected to a dichloroacetyl tail through a 2-amino-1,3-propanediol moiety. CAM displays a broad-spectrum bacteriostatic activity by specifically inhibiting the bacterial protein synthesis. In certain but important cases, it also exhibits bactericidal activity, namely against the three most common causes of meningitis, Haemophilus influenzae, Streptococcus pneumoniae and Neisseria meningitidis. Resistance to CAM has been frequently reported and ascribed to a variety of mechanisms. However, the most important concerns that limit its clinical utility relate to side effects such as neurotoxicity and hematologic disorders. In this review, we present previous and current efforts to synthesize CAM derivatives with improved pharmacological properties. In addition, we highlight potentially broader roles of these derivatives in investigating the plasticity of the ribosomal catalytic center, the main target of CAM.
Collapse
Affiliation(s)
- George P Dinos
- Department of Biochemistry, School of Medicine, University of Patras, GR-26504 Patras, Greece.
| | | | - Dionissia A Missiri
- Laboratory of Synthetic Organic Chemistry, Department of Chemistry, University of Patras, GR-26504 Patras, Greece.
| | | | - Ioannis A Vlachogiannis
- Department of Biochemistry, School of Medicine, University of Patras, GR-26504 Patras, Greece.
| | - Georgios E Papadopoulos
- Department of Biochemistry and Biotechnology, University of Thessaly, Ploutonos 26, GR-41221 Larissa, Greece.
| | - Dionissios Papaioannou
- Laboratory of Synthetic Organic Chemistry, Department of Chemistry, University of Patras, GR-26504 Patras, Greece.
| | - Dimitrios L Kalpaxis
- Department of Biochemistry, School of Medicine, University of Patras, GR-26504 Patras, Greece.
| |
Collapse
|
40
|
Emancipating Chlamydia: Advances in the Genetic Manipulation of a Recalcitrant Intracellular Pathogen. Microbiol Mol Biol Rev 2016; 80:411-27. [PMID: 27030552 DOI: 10.1128/mmbr.00071-15] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chlamydia species infect millions of individuals worldwide and are important etiological agents of sexually transmitted disease, infertility, and blinding trachoma. Historically, the genetic intractability of this intracellular pathogen has hindered the molecular dissection of virulence factors contributing to its pathogenesis. The obligate intracellular life cycle of Chlamydia and restrictions on the use of antibiotics as selectable markers have impeded the development of molecular tools to genetically manipulate these pathogens. However, recent developments in the field have resulted in significant gains in our ability to alter the genome of Chlamydia, which will expedite the elucidation of virulence mechanisms. In this review, we discuss the challenges affecting the development of molecular genetic tools for Chlamydia and the work that laid the foundation for recent advancements in the genetic analysis of this recalcitrant pathogen.
Collapse
|
41
|
Stefano GB, Kream RM. Dysregulated mitochondrial and chloroplast bioenergetics from a translational medical perspective (Review). Int J Mol Med 2016; 37:547-55. [PMID: 26821064 PMCID: PMC4771107 DOI: 10.3892/ijmm.2016.2471] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/22/2016] [Indexed: 02/06/2023] Open
Abstract
Mitochondria and chloroplasts represent endosymbiotic models of complex organelle development, driven by intense evolutionary pressure to provide exponentially enhanced ATP-dependent energy production functionally linked to cellular respiration and photosynthesis. Within the realm of translational medicine, it has become compellingly evident that mitochondrial dysfunction, resulting in compromised cellular bioenergetics, represents a key causative factor in the etiology and persistence of major diseases afflicting human populations. As a pathophysiological consequence of enhanced oxygen utilization that is functionally uncoupled from the oxidative phosphorylation of ADP, significant levels of reactive oxygen species (ROS) may be generated within mitochondria and chloroplasts, which may effectively compromise cellular energy production following prolonged stress/inflammatory conditions. Empirically determined homologies in biochemical pathways, and their respective encoding gene sequences between chloroplasts and mitochondria, suggest common origins via entrapped primordial bacterial ancestors. From evolutionary and developmental perspectives, the elucidation of multiple biochemical and molecular relationships responsible for errorless bioenergetics within mitochondrial and plastid complexes will most certainly enhance the depth of translational approaches to ameliorate or even prevent the destructive effects of multiple disease states. The selective choice of discussion points contained within the present review is designed to provide theoretical bases and translational insights into the pathophysiology of human diseases from a perspective of dysregulated mitochondrial bioenergetics with special reference to chloroplast biology.
Collapse
|
42
|
Abstract
Mitochondrial dysfunction underlies many human disorders, including those that affect the visual system. The retinal ganglion cells, whose axons form the optic nerve, are often damaged by mitochondrial-related diseases which result in blindness. Both mitochondrial DNA (mtDNA) and nuclear gene mutations impacting many different mitochondrial processes can result in optic nerve disease. Of particular importance are mutations that impair mitochondrial network dynamics (fusion and fission), oxidative phosphorylation (OXPHOS), and formation of iron-sulfur complexes. Current genetic knowledge can inform genetic counseling and suggest strategies for novel gene-based therapies. Identifying new optic neuropathy-causing genes and defining the role of current and novel genes in disease will be important steps toward the development of effective and potentially neuroprotective therapies.
Collapse
Affiliation(s)
- Janey L Wiggs
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, Massachusetts 02114;
| |
Collapse
|
43
|
Liu CW, Li CH, Peng YJ, Cheng YW, Chen HW, Liao PL, Kang JJ, Yeng MH. Snail regulates Nanog status during the epithelial-mesenchymal transition via the Smad1/Akt/GSK3β signaling pathway in non-small-cell lung cancer. Oncotarget 2015; 5:3880-94. [PMID: 25003810 PMCID: PMC4116528 DOI: 10.18632/oncotarget.2006] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The epithelial–mesenchymal transition (EMT), a crucial step in cancer metastasis, is important in transformed cancer cells with stem cell-like properties. In this study, we established a Snail-overexpressing cell model for non-small-cell lung cancer (NSCLC) and investigated its underlying mechanism. We also identified the downstream molecular signaling pathway that contributes to the role of Snail in regulating Nanog expression. Our data shows that high levels of Snail expression correlate with metastasis and high levels of Nanog expression in NSCLC. NSCLC cells expressing Snail are characterized by active EMT characteristics and exhibit an increased ability to migrate, chemoresistance, sphere formation, and stem cell-like properties. We also investigated the signals required for Snail-mediated Nanog expression. Our data demonstrate that LY294002, SB431542, LDN193189, and Noggin pretreatment inhibit Snail-induced Nanog expression during EMT. This study shows a significant correlation between Snail expression and phosphorylation of Smad1, Akt, and GSK3β. In addition, pretreatment with SB431542, LDN193189, or Noggin prevented Snail-induced Smad1 and Akt hyperactivation and reactivated GSK3β. Moreover, LY294002 pretreatment prevented Akt hyperactivation and reactivated GSK3β without altering Smad1 activation. These findings provide a novel mechanistic insight into the important role of Snail in NSCLC during EMT and indicate potentially useful therapeutic targets for NSCLC.
Collapse
Affiliation(s)
- Chen-Wei Liu
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | - Jaw-Jou Kang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | |
Collapse
|
44
|
|
45
|
Shiomi M, Miyamae M, Takemura G, Kaneda K, Inamura Y, Onishi A, Koshinuma S, Momota Y, Minami T, Figueredo VM. Induction of autophagy restores the loss of sevoflurane cardiac preconditioning seen with prolonged ischemic insult. Eur J Pharmacol 2013; 724:58-66. [PMID: 24374197 DOI: 10.1016/j.ejphar.2013.12.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 12/17/2013] [Accepted: 12/18/2013] [Indexed: 12/30/2022]
Abstract
Sevoflurane preconditioning against myocardial ischemia-reperfusion injury is lost if the ischemic insult is too long. Emerging evidence suggests that induction of autophagy may also confer cardioprotection against ischemia-reperfusion injury. We examined whether induction of autophagy prolongs sevoflurane preconditioning protection during a longer ischemic insult. Isolated guinea pigs hearts were subjected to 30 or 45 min ischemia, followed by 120 min reperfusion (control). Anesthetic preconditioning was elicited with 2% sevoflurane for 10 min prior to ischemia (SEVO-30, SEVO-45). Chloramphenicol (autophagy upregulator, 300 µM) was administered starting 20 min before ischemia and throughout reperfusion in SEVO-45 (SEVO-45+CAP). To inhibit autophagy, 3-methyladenine (10 μM) was administered during sevoflurane administration in SEVO-45+CAP. Infarct size was determined by triphenyltetrazolium chloride stain. Tissue samples were obtained before ischemia to determine autophagy-related protein (microtubule-associated protein light chain I and II: LC3-I, II), Akt and glycogen synthase kinase 3β (GSK3β) expression using Western blot analysis. The effect of autophagy on calcium-induced mitochondrial permeability transition pore (MPTP) opening in isolated calcein-loaded mitochondria was assessed. Electron microscopy was used to detect autophagosomes. Infarct size was significantly reduced in SEVO-30, but not in SEVO-45. Chloramphenicol restored sevoflurane preconditioning lost by 45 min ischemia. There were more abundant autophagozomes and LC3-II expression was significantly increased in SEVO-45+CAP. Induction of autophagy before ischemia enhanced GSK3β phosphorylation and inhibition of calcium-induced MPTP opening. These effects were abolished by 3-methyladenine. Pre-ischemic induction of autophagy restores sevoflurane preconditioning lost by longer ischemic insult. This effect is associated with enhanced inhibition of MPTP by autophagy.
Collapse
Affiliation(s)
- Mayumi Shiomi
- Department of Anesthesiology, Osaka Medical College, Osaka, Japan
| | - Masami Miyamae
- Department of Internal Medicine, Osaka Dental University, 8-1 Kuzuha hanazono-cho Hirakata, Osaka 573-1121, Japan.
| | - Genzou Takemura
- Department of Cardiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kazuhiro Kaneda
- Department of Anesthesiology, Osaka Dental University, Osaka, Japan
| | | | - Anna Onishi
- Department of Anesthesiology, Osaka Dental University, Osaka, Japan
| | | | - Yoshihiro Momota
- Department of Anesthesiology, Osaka Dental University, Osaka, Japan
| | - Toshiaki Minami
- Department of Anesthesiology, Osaka Medical College, Osaka, Japan
| | - Vincent M Figueredo
- Institute for Heart and Vascular Health, Einstein Medical Center, and Jefferson Medical College, Philadelphia, USA
| |
Collapse
|
46
|
Marullo R, Werner E, Degtyareva N, Moore B, Altavilla G, Ramalingam SS, Doetsch PW. Cisplatin induces a mitochondrial-ROS response that contributes to cytotoxicity depending on mitochondrial redox status and bioenergetic functions. PLoS One 2013; 8:e81162. [PMID: 24260552 PMCID: PMC3834214 DOI: 10.1371/journal.pone.0081162] [Citation(s) in RCA: 565] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 10/09/2013] [Indexed: 11/30/2022] Open
Abstract
Cisplatin is one of the most effective and widely used anticancer agents for the treatment of several types of tumors. The cytotoxic effect of cisplatin is thought to be mediated primarily by the generation of nuclear DNA adducts, which, if not repaired, cause cell death as a consequence of DNA replication and transcription blockage. However, the ability of cisplatin to induce nuclear DNA (nDNA) damage per se is not sufficient to explain its high degree of effectiveness nor the toxic effects exerted on normal, post-mitotic tissues. Oxidative damage has been observed in vivo following exposure to cisplatin in several tissues, suggesting a role for oxidative stress in the pathogenesis of cisplatin-induced dose-limiting toxicities. However, the mechanism of cisplatin-induced generation of ROS and their contribution to cisplatin cytotoxicity in normal and cancer cells is still poorly understood. By employing a panel of normal and cancer cell lines and the budding yeast Saccharomyces cerevisiae as model system, we show that exposure to cisplatin induces a mitochondrial-dependent ROS response that significantly enhances the cytotoxic effect caused by nDNA damage. ROS generation is independent of the amount of cisplatin-induced nDNA damage and occurs in mitochondria as a consequence of protein synthesis impairment. The contribution of cisplatin-induced mitochondrial dysfunction in determining its cytotoxic effect varies among cells and depends on mitochondrial redox status, mitochondrial DNA integrity and bioenergetic function. Thus, by manipulating these cellular parameters, we were able to enhance cisplatin cytotoxicity in cancer cells. This study provides a new mechanistic insight into cisplatin-induced cell killing and may lead to the design of novel therapeutic strategies to improve anticancer drug efficacy.
Collapse
Affiliation(s)
- Rossella Marullo
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia, United States of America
- Department of Medical Oncology, University of Messina, Messina, Italy
| | - Erica Werner
- Department of Biochemistry, Emory University, Atlanta, Georgia, United States of America
| | - Natalya Degtyareva
- Department of Biochemistry, Emory University, Atlanta, Georgia, United States of America
| | - Bryn Moore
- Department of Biochemistry, Emory University, Atlanta, Georgia, United States of America
- Geisiner Medical Center, Danville, Pennsylvania, United States of America
| | | | - Suresh S. Ramalingam
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia, United States of America
| | - Paul W. Doetsch
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia, United States of America
- Department of Biochemistry, Emory University, Atlanta, Georgia, United States of America
- Geisiner Medical Center, Danville, Pennsylvania, United States of America
- Department of Radiation Oncology, Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
47
|
Xu S, Battaglia L, Bao X, Fan H. Chloramphenicol acetyltransferase as a selection marker for chlamydial transformation. BMC Res Notes 2013; 6:377. [PMID: 24060200 PMCID: PMC3849861 DOI: 10.1186/1756-0500-6-377] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 09/11/2013] [Indexed: 12/04/2022] Open
Abstract
Background Chlamydia is a common bacterial pathogen responsible for many diseases. Methods for transforming this important organism using a β-lactamase as a selection marker have been developed very recently. However, the National Institutes of Health Guidelines for Research Involving Recombinant DNA Molecules do not permit transformation experiments with β-lactamase gene-containing vectors for certain human chlamydial pathogens. Therefore, a different selection marker is urgently needed for transformation of those chlamydiae. Results After transformation of plasmid-free Chlamydia trachomatis with pGFP:SW2, which carries a β-lactamase and a chloramphenicol acetyltransferase gene fused to a green fluorescence protein gene, transformants were obtained by selection with either ampicillin or chloramphenicol. Stable chloramphenicol-resistant, but ampicillin-sensitive, transformants were obtained using a pGFP:SW2 derivative without the β-lactamase. All transformants expressed green fluorescence protein and had glycogen synthesis activity restored. Conclusions Chloramphenicol resistance may be used as a selection marker for genetic experiments in Chlamydia. This eliminates the requirement for the use of β-lactamase, of which dissemination to some C. trachomatis serovars may jeopardize clinical treatment of chlamydial infections in pregnant women. Chloramphenicol acetyltransferase may also serve as a useful secondary selection marker for genetic analyses in β-lactamase-transformed chlamydial strains.
Collapse
Affiliation(s)
- Shuang Xu
- Department of Pharmacology, Rutgers University Robert Wood Johnson Medical School, Piscataway, NJ, USA.
| | | | | | | |
Collapse
|
48
|
Zheng X, Xia C, Chen Z, Huang J, Gao F, Li G, Zhang B. Requirement of the phosphatidylinositol 3-kinase/Akt signaling pathway for the effect of nicotine on interleukin-1beta-induced chondrocyte apoptosis in a rat model of osteoarthritis. Biochem Biophys Res Commun 2012; 423:606-12. [PMID: 22713471 DOI: 10.1016/j.bbrc.2012.06.045] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 06/10/2012] [Indexed: 11/19/2022]
Abstract
Chondrocyte apoptosis is mainly responsible for the progressive degeneration of cartilage in osteoarthritis (OA). Interleukin-1beta (IL-1β) was widely used as a modulating and chondrocyte apoptosis-inducing agent. Nicotine is able to confer resistance to apoptosis and promote cell survival in some cell lines, but its regulatory mechanism is ambiguous. We aimed to investigate the effect of nicotine on IL-1β-induced chondrocyte apoptosis and the mechanism underlying how nicotine antagonizes IL-1β-induced apoptosis of rat chondrocytes. Chondrocytes isolated from newborn rat joints were exposed to IL-1β. The cell viability was analyzed by the MTT (3-(4,5-dimethylthiazol-2yl)-2,5-diphenyl tetrazolium bromide) assay, and the apoptotic cells were counted with DAPI staining. The levels of Akt, phosphorylated-Akt (p-Akt) and downstream protein targets of Akt were detected by western blotting. The results showed that nicotine neutralized the effect of IL-1β on chondrocytes by activating PI3K/Akt signaling pathways, including the PI3K/Akt/Bcl-2 pathway, to block IL-1β-induced cell apoptosis and the PI3K/Akt/p70S6K (p70S6 kinase)/S6 pathway for promoting protein synthesis, modulating its downstream effectors such as TIMP-1 and MMP-13. Activation of the PI3K/Akt pathway is, in part, required for the effect of nicotine on IL-1β-induced chondrocyte apoptosis in a rat model of osteoarthritis.
Collapse
Affiliation(s)
- Xinpeng Zheng
- Zhongshan Hospital, School of Medicine, University of Xiamen, Xiamen, Fujian, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Genetic Manipulation of Coxiella burnetii. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 984:249-71. [DOI: 10.1007/978-94-007-4315-1_13] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
50
|
OXPHOS toxicogenomics and Parkinson's disease. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2011; 728:98-106. [DOI: 10.1016/j.mrrev.2011.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 05/16/2011] [Accepted: 06/30/2011] [Indexed: 12/21/2022]
|