1
|
Li CX, Talukder M, Xu YR, Zhu SY, Wang YX, Li JL. Cadmium causes cerebral mitochondrial dysfunction through regulating mitochondrial HSF1. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 360:124677. [PMID: 39127336 DOI: 10.1016/j.envpol.2024.124677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/15/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Mitochondria, as the powerhouse of the cell, play a vital role in maintaining cellular energy homeostasis and are known to be a primary target of cadmium (Cd) toxicity. The improper targeting of proteins to mitochondria can compromise the normal functions of the mitochondria. However, the precise mechanism by which protein localization contributes to the development of mitochondrial dysfunction induced by Cd is still not fully understood. For this research, Hy-Line white variety chicks (1-day-old) were used and equally distributed into 4 groups: the Control group (fed with a basic diet), the Cd35 group (basic diet with 35 mg/kg CdCl2), the Cd70 group (basic diet with 70 mg/kg CdCl2) and the Cd140 group (basic diet with 140 mg/kg CdCl2), respectively for 90 days. It was found that Cd caused the accumulation of heat shock factor 1 (HSF1) in the mitochondria, and the overexpression of HSF1 in the mitochondria led to mitochondrial dysfunction and neuronal damage. This process is due to the mitochondrial HSF1 (mtHSF1), causing mitochondrial fission through the upregulation of dynamin-related protein 1 (Drp1) content, while inhibiting oligomer formation of single-stranded DNA-binding protein 1 (SSBP1), resulting in the mitochondrial DNA (mtDNA) deletion. The findings unveil an unforeseen role of HSF1 in triggering mitochondrial dysfunction.
Collapse
Affiliation(s)
- Chen-Xi Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Milton Talukder
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, 8210, Bangladesh
| | - Ya-Ru Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Shi-Yong Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yu-Xiang Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
2
|
Zhu H, Yu Y, Li Y, Chang S, Liu Y. Puerarin ameliorates high glucose-induced MIN6 cell injury by activating PINK1/Parkin-mediated mitochondrial autophagy. Heliyon 2024; 10:e36176. [PMID: 39224278 PMCID: PMC11367457 DOI: 10.1016/j.heliyon.2024.e36176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
The dysfunction of pancreatic β-cells plays a pivotal role in the pathogenesis of type 2 diabetes mellitus (T2DM). Despite numerous studies demonstrating the anti-inflammatory and antioxidant properties of puerarin, the protective effects of puerarin on β-cells remain poorly understood. Hence, this study aimed to explore the effects of puerarin on β-cell dysfunction in a hyperglycemic environment via the PINK/Parkin-mediated mitochondrial autophagy pathway. The alterations in cell viability of MIN6 cells exposed to glucose concentrations of 5 mM, 10 mM, 20 mM, and 30 mM for 24 h, 48 h, and 72 h, respectively, were assessed using the CCK-8 assay to optimize the modeling conditions. Subsequently, cellular insulin secretion was measured using enzyme-linked immunosorbent assay (ELISA), apoptosis rate by flow cytometry, mitochondrial membrane potential alteration by JC-1, cellular ROS production by the DCFH-DA fluorescent probe, and fusion of cellular autophagosomes and lysosomes through adenoviral infection analysis. Furthermore, gene and protein expression levels of the PINK/Parkin-mediated mitochondrial autophagy pathway and mitochondrial apoptosis pathway were assessed using real-time quantitative polymerase chain reaction (RT-qPCR) and Western blot, respectively. Results indicated a significant decrease in MIN6 cell viability following 48 h of exposure to 30 mM glucose concentration. Puerarin intervention markedly attenuated ROS production, restored mitochondrial membrane potential, induced PINK/Parkin-mediated mitochondrial autophagy, suppressed activation of the mitochondrial apoptotic pathway, mitigated apoptosis, and enhanced insulin secretion in a high glucose (HG) environment. The findings of this investigation contribute to a deeper understanding of the precise mechanism underlying the protective effects of puerarin on β-cells and offer a theoretical foundation for advancing puerarin-based therapeutics aimed at ameliorating T2DM.
Collapse
Affiliation(s)
- Hongyang Zhu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - You Yu
- The First Affiliated Hospital Of Nanchang University, Nanchang, China
| | - Yuting Li
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Shiyao Chang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yuhui Liu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
3
|
Fu C, Cao N, Liu W, Zhang Z, Yang Z, Zhu W, Fan S. Crosstalk between mitophagy and innate immunity in viral infection. Front Microbiol 2022; 13:1064045. [PMID: 36590405 PMCID: PMC9800879 DOI: 10.3389/fmicb.2022.1064045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Mitochondria are important organelles involved in cell metabolism and programmed cell death in eukaryotic cells and are closely related to the innate immunity of host cells against viruses. Mitophagy is a process in which phagosomes selectively phagocytize damaged or dysfunctional mitochondria to form autophagosomes and is degraded by lysosomes, which control mitochondrial mass and maintain mitochondrial dynamics and cellular homeostasis. Innate immunity is an important part of the immune system and plays a vital role in eliminating viruses. Viral infection causes many physiological and pathological alterations in host cells, including mitophagy and innate immune pathways. Accumulating evidence suggests that some virus promote self-replication through regulating mitophagy-mediated innate immunity. Clarifying the regulatory relationships among mitochondria, mitophagy, innate immunity, and viral infection will shed new insight for pathogenic mechanisms and antiviral strategies. This review systemically summarizes the activation pathways of mitophagy and the relationship between mitochondria and innate immune signaling pathways, and then discusses the mechanisms of viruses on mitophagy and innate immunity and how viruses promote self-replication by regulating mitophagy-mediated innate immunity.
Collapse
Affiliation(s)
- Cheng Fu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Nan Cao
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Wenjun Liu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Zilin Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zihui Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wenhui Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China,*Correspondence: Wenhui Zhu,
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China,Shuangqi Fan,
| |
Collapse
|
4
|
Moore MP, Cunningham RP, Meers GM, Johnson SA, Wheeler AA, Ganga RR, Spencer NM, Pitt JB, Diaz-Arias A, Swi AIA, Hammoud GM, Ibdah JA, Parks EJ, Rector RS. Compromised hepatic mitochondrial fatty acid oxidation and reduced markers of mitochondrial turnover in human NAFLD. Hepatology 2022; 76:1452-1465. [PMID: 35000203 PMCID: PMC9270503 DOI: 10.1002/hep.32324] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS NAFLD and its more-advanced form, steatohepatitis (NASH), is associated with obesity and is an independent risk factor for cardiovascular, liver-related, and all-cause mortality. Available human data examining hepatic mitochondrial fatty acid oxidation (FAO) and hepatic mitochondrial turnover in NAFLD and NASH are scant. APPROACH AND RESULTS To investigate this relationship, liver biopsies were obtained from patients with obesity undergoing bariatric surgery and data clustered into four groups based on hepatic histopathological classification: Control (CTRL; no disease); NAFL (steatosis only); Borderline-NASH (steatosis with lobular inflammation or hepatocellular ballooning); and Definite-NASH (D-NASH; steatosis, lobular inflammation, and hepatocellular ballooning). Hepatic mitochondrial complete FAO to CO2 and the rate-limiting enzyme in β-oxidation (β-hydroxyacyl-CoA dehydrogenase activity) were reduced by ~40%-50% with D-NASH compared with CTRL. This corresponded with increased hepatic mitochondrial reactive oxygen species production, as well as dramatic reductions in markers of mitochondrial biogenesis, autophagy, mitophagy, fission, and fusion in NAFL and NASH. CONCLUSIONS These findings suggest that compromised hepatic FAO and mitochondrial turnover are intimately linked to increasing NAFLD severity in patients with obesity.
Collapse
Affiliation(s)
- Mary P. Moore
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
| | - Rory P. Cunningham
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
| | - Grace M. Meers
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
| | - Sarah A. Johnson
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| | - Andrew A. Wheeler
- Department of Surgery, University of Missouri, Columbia MO, USA, 65211
| | - Rama R. Ganga
- Department of Surgery, University of Missouri, Columbia MO, USA, 65211
| | - Nicole M. Spencer
- Department of Surgery, University of Missouri, Columbia MO, USA, 65211
| | - James B. Pitt
- Department of Surgery, University of Missouri, Columbia MO, USA, 65211
| | | | - Ahmed I. A. Swi
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| | - Ghassan M. Hammoud
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| | - Jamal A. Ibdah
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| | - Elizabeth J. Parks
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| | - R. Scott Rector
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO, USA, 65201
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA, 65211
- Department of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia MO, USA, 65211
| |
Collapse
|
5
|
Barathkumar S, Padhi RK, Parida PK, Marigoudar SR. In vivo appraisal of oxidative stress response, cell ultrastructural aberration and accumulation in Juvenile Scylla serrata exposed to uranium. CHEMOSPHERE 2022; 300:134561. [PMID: 35413368 DOI: 10.1016/j.chemosphere.2022.134561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 06/14/2023]
Abstract
In vivo studies were performed to evaluate the organ specific tissue accumulation and cellular toxicity of uranium to mud crab Scylla serrata. The specimens were acclimated in natural seawater and the exposure to 50-250 μg/L uranium was investigated up to 60 days. The present study examined the effects of concentration and duration of uranium exposure in the tissue of S. serrata at cellular and subcellular level using scanning electron microscopy and bright field transmission electron microscopy in addition to histological analysis. The results indicated that accumulation of U in S. serrata was organ specific and followed the order gills > hepatopancreas > muscle. The response of key antioxidant enzyme activities such as SOD, GPx and CAT in different organs of crabs indicated oxidative stress due to U in the ambient medium and tissue. At 50 and 100 μg/L of U exposure, individuals were able to acclimate the oxidative stress and withstand the uranium exposure. This acclimation could not be sustained at higher concentrations (250 μg/L), affecting the production of CAT in the tissues. Cellular and subcellular changes were observed in the hemocytes with reduction in their number in consonance with the antioxidant enzymes. Histological aberrations like lamellar disruption of gill, necrosis of hepatopancreas, disruption and rupture of muscle bundles were observed at different concentrations and were severe at higher concentration (250 μg/L). Necrosis was observed in the electron micrographs of tissues shortly after 15 days of exposure. SEM micrograph clearly shows disrupted lamellae, folding of marginal canal and reduction of inter lamellar spaces in the gills of crab exposed to high concentration of uranium. Mitochondrial anomalies are reported for the first time in the present study in addition to the subcellular changes and vacuoles on exposure uranium in the cells of gill and hepatopancreas.
Collapse
Affiliation(s)
- S Barathkumar
- National Centre for Coastal Research, Ministry of Earth Science, Chennai, Tamil Nadu, 600100, India; Indira Gandhi Centre for Atomic Research, Kalpakkam, Tamil Nadu, 603102, India
| | - R K Padhi
- Material Chemistry and Metal Fuel Cycle Group, Indira Gandhi Centre for Atomic Research, Kalpakkam, Tamil Nadu, 603102, India.
| | - P K Parida
- Metallurgy and Materials Group, Indira Gandhi Centre for Atomic Research, Kalpakkam, Tamil Nadu, 603102, India
| | - S R Marigoudar
- National Centre for Coastal Research, Ministry of Earth Science, Chennai, Tamil Nadu, 600100, India
| |
Collapse
|
6
|
Maksoud S. The DNA Double-Strand Break Repair in Glioma: Molecular Players and Therapeutic Strategies. Mol Neurobiol 2022; 59:5326-5365. [PMID: 35696013 DOI: 10.1007/s12035-022-02915-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/05/2022] [Indexed: 12/12/2022]
Abstract
Gliomas are the most frequent type of tumor in the central nervous system, which exhibit properties that make their treatment difficult, such as cellular infiltration, heterogeneity, and the presence of stem-like cells responsible for tumor recurrence. The response of this type of tumor to chemoradiotherapy is poor, possibly due to a higher repair activity of the genetic material, among other causes. The DNA double-strand breaks are an important type of lesion to the genetic material, which have the potential to trigger processes of cell death or cause gene aberrations that could promote tumorigenesis. This review describes how the different cellular elements regulate the formation of DNA double-strand breaks and their repair in gliomas, discussing the therapeutic potential of the induction of this type of lesion and the suppression of its repair as a control mechanism of brain tumorigenesis.
Collapse
Affiliation(s)
- Semer Maksoud
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
7
|
Von Schulze AT, Geiger PC. Heat and Mitochondrial Bioenergetics. CURRENT OPINION IN PHYSIOLOGY 2022. [DOI: 10.1016/j.cophys.2022.100553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
8
|
McCormick JJ, King KE, Rutherford MM, Meade RD, Notley SR, Akerman AP, Dokladny K, Kenny GP. Effect of extracellular hyperosmolality during normothermia and hyperthermia on the autophagic response in peripheral blood mononuclear cells from young men. J Appl Physiol (1985) 2022; 132:995-1004. [PMID: 35238651 DOI: 10.1152/japplphysiol.00661.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Heat-stress induced dehydration is associated with extracellular hyperosmolality. To counteract the associated stress, cells employ cytoprotective mechanisms, including autophagy, however, the autophagic response to hyperosmotic stress has yet to be evaluated in humans. Thus, we investigated autophagy and associated cellular stress pathways (the heat shock response [HSR], apoptosis, and the acute inflammatory response) to isosmotic and hyperosmotic conditions with and without hyperthermia in twelve young men (mean [SD]; 25 [5] years). Participants received a 90-min intravenous infusion of either isosmotic (ISO; 0.9% NaCl; serum osmolality of 293 [4] mOsm/kg) or hyperosmotic (HYP; 3.0% NaCl; 300 [6] mOsm/kg) saline, followed by passive whole-body heating using a water perfused suit to increase esophageal temperature by ~0.8⁰C. Peripheral blood mononuclear cells were harvested at baseline (pre-infusion), post-infusion, and after heating, and changes in protein content were analyzed via Western blotting. Post-infusion, the LC3-II/I ratio was higher in HYP compared to ISO infusion (p<0.001), although no other protein changes were observed (all p>0.050). Following passive heating, autophagy increased in HYP, as demonstrated by an increase in LC3-II from baseline (p=0.004) and an elevated LC3-II/I ratio compared to ISO (p=0.035), and a decrease in p62 when compared to the ISO condition (p=0.019). This was accompanied by an elevation in cleaved caspase-3 following heating in the HYP condition (p<0.010), however, the HSR and acute inflammatory response did not change under any condition (all p>0.050). Taken together, our findings indicate that serum hyperosmolality induces autophagy and apoptotic signaling during mild hyperthermia with minimal autophagic activation during normothermia.
Collapse
Affiliation(s)
- James J McCormick
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Kelli E King
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Maura M Rutherford
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Robert D Meade
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada.,Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, United States
| | - Sean R Notley
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Ashley P Akerman
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Karol Dokladny
- Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico, United States
| | - Glen P Kenny
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Canada.,Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
9
|
The complex interplay between autophagy and cell death pathways. Biochem J 2022; 479:75-90. [PMID: 35029627 DOI: 10.1042/bcj20210450] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/26/2022]
Abstract
Autophagy is a universal cellular homeostatic process, required for the clearance of dysfunctional macromolecules or organelles. This self-digestion mechanism modulates cell survival, either directly by targeting cell death players, or indirectly by maintaining cellular balance and bioenergetics. Nevertheless, under acute or accumulated stress, autophagy can also contribute to promote different modes of cell death, either through highly regulated signalling events, or in a more uncontrolled inflammatory manner. Conversely, apoptotic or necroptotic factors have also been implicated in the regulation of autophagy, while specific factors regulate both processes. Here, we survey both earlier and recent findings, highlighting the intricate interaction of autophagic and cell death pathways. We, Furthermore, discuss paradigms, where this cross-talk is disrupted, in the context of disease.
Collapse
|
10
|
Abd El-Aziz YS, Leck LYW, Jansson PJ, Sahni S. Emerging Role of Autophagy in the Development and Progression of Oral Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:6152. [PMID: 34944772 PMCID: PMC8699656 DOI: 10.3390/cancers13246152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a cellular catabolic process, which is characterized by degradation of damaged proteins and organelles needed to supply the cell with essential nutrients. At basal levels, autophagy is important to maintain cellular homeostasis and development. It is also a stress responsive process that allows the cells to survive when subjected to stressful conditions such as nutrient deprivation. Autophagy has been implicated in many pathologies including cancer. It is well established that autophagy plays a dual role in different cancer types. There is emerging role of autophagy in oral squamous cell carcinoma (OSCC) development and progression. This review will focus on the role played by autophagy in relation to different aspects of cancer progression and discuss recent studies exploring the role of autophagy in OSCC. It will further discuss potential therapeutic approaches to target autophagy in OSCC.
Collapse
Affiliation(s)
- Yomna S. Abd El-Aziz
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, NSW 2064, Australia
- Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta 31527, Egypt
| | - Lionel Y. W. Leck
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, NSW 2064, Australia
- Cancer Drug Resistance and Stem Cell Program, University of Sydney, Sydney, NSW 2006, Australia
| | - Patric J. Jansson
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, NSW 2064, Australia
- Cancer Drug Resistance and Stem Cell Program, University of Sydney, Sydney, NSW 2006, Australia
| | - Sumit Sahni
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, NSW 2064, Australia
| |
Collapse
|
11
|
Guan R, Li Z, Dai X, Zou W, Yu X, Liu H, Chen Q, Teng W, Liu P, Liu X, Dong S. Electroacupuncture at GV20‑GB7 regulates mitophagy to protect against neurological deficits following intracerebral hemorrhage via inhibition of apoptosis. Mol Med Rep 2021; 24:492. [PMID: 33955500 PMCID: PMC8127033 DOI: 10.3892/mmr.2021.12131] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 02/24/2021] [Indexed: 01/07/2023] Open
Abstract
The acupuncture penetrating line of Baihui (GV20) to Qubin (GB7) spans the parietal, frontal and temporal lobes. The present study aimed to elucidate the mechanism by which electroacupuncture (EA) at GV20‑GB7 regulates mitophagy in intracerebral hemorrhage (ICH) and whether it serves a neuroprotective role. A whole blood‑induced ICH model was used. Mitophagy‑regulating proteins, including BCL/adenovirus E1B 19 kDa‑interacting protein 3 (BNIP3), PTEN‑induced putative kinase 1 (PINK1), Parkin and apoptosis‑associated proteins were detected by western blotting; autophagy following ICH was evaluated by immunofluorescent techniques; morphological characteristics of mitophagy were observed using transmission electron microscopy; and TUNEL assay was performed to determine the number of apoptotic cells. Immunohistochemistry was used to detect p53 expression. The protective role of EA (GV20‑GB7) via enhanced mitophagy and suppressed apoptosis in ICH was further confirmed by decreased modified neurological severity score. The results showed that EA (GV20‑GB7) treatment upregulated mitochondrial autophagy following ICH and inhibited apoptotic cell death. The mechanism underlying EA (GV20‑GB7) treatment may involve inhibition of p53, an overlapping protein of autophagy and apoptosis. EA (GV20‑GB7) treatment decreased neurobehavioral deficits following ICH but pretreatment with 3‑methyladenine counteracted the beneficial effects of EA (GV20‑GB7) treatment. In conclusion, EA (GV20‑GB7) improved recovery from ICH by regulating the balance between mitophagy and apoptosis.
Collapse
Affiliation(s)
- Ruiqiao Guan
- Department of Integrated Chinese and Western Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- The Third Department of Acupuncture and Moxibustion, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- Clinical Key Laboratory of Integrated Chinese and Western Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- Department of Traditional Chinese Medicine, London South Bank University, London SE1 6RD, UK
- The Clinic of Traditional Chinese Medicine, London Confucius Institute of Traditional Chinese Medicine, London SE1 0AA, UK
| | - Zhihao Li
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Chinese Medicine, Shanghai 200437, P.R. China
| | - Xiaohong Dai
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- The Third Department of Acupuncture and Moxibustion, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Wei Zou
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- The Third Department of Acupuncture and Moxibustion, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Xueping Yu
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- The Third Department of Acupuncture and Moxibustion, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Hao Liu
- Department of Acupuncture and Moxibustion, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 315099, P.R. China
| | - Qiuxin Chen
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- The Third Department of Acupuncture and Moxibustion, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- Clinical Key Laboratory of Integrated Chinese and Western Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Wei Teng
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- The Third Department of Acupuncture and Moxibustion, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Peng Liu
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- The Third Department of Acupuncture and Moxibustion, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Xiaoying Liu
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- The Third Department of Acupuncture and Moxibustion, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Shanshan Dong
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- The Third Department of Acupuncture and Moxibustion, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- Clinical Key Laboratory of Integrated Chinese and Western Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
12
|
Von Schulze AT, Deng F, Fuller KNZ, Franczak E, Miller J, Allen J, McCoin CS, Shankar K, Ding WX, Thyfault JP, Geiger PC. Heat Treatment Improves Hepatic Mitochondrial Respiratory Efficiency via Mitochondrial Remodeling. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab001. [PMID: 33629069 PMCID: PMC7886620 DOI: 10.1093/function/zqab001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 01/06/2023]
Abstract
Nonacholic fatty liver disease, or hepatic steatosis, is the most common liver disorder affecting the western world and currently has no pharmacologic cure. Thus, many investigations have focused on alternative strategies to treat or prevent hepatic steatosis. Our laboratory has shown that chronic heat treatment (HT) mitigates glucose intolerance, insulin resistance, and hepatic steatosis in rodent models of obesity. Here, we investigate the direct bioenergetic mechanism(s) surrounding the metabolic effects of HT on hepatic mitochondria. Utilizing mitochondrial proteomics and respiratory function assays, we show that one bout of acute HT (42°C for 20 min) in male C57Bl/6J mice (n = 6/group) triggers a hepatic mitochondrial heat shock response resulting in acute reductions in respiratory capacity, degradation of key mitochondrial enzymes, and induction of mitophagy via mitochondrial ubiquitination. We also show that chronic bouts of HT and recurrent activation of the heat shock response enhances mitochondrial quality and respiratory function via compensatory adaptations in mitochondrial organization, gene expression, and transport even during 4 weeks of high-fat feeding (n = 6/group). Finally, utilizing a liver-specific heat shock protein 72 (HSP72) knockout model, we are the first to show that HSP72, a protein putatively driving the HT metabolic response, does not play a significant role in the hepatic mitochondrial adaptation to acute or chronic HT. However, HSP72 is required for the reductions in blood glucose observed with chronic HT. Our data are the first to suggest that chronic HT (1) improves hepatic mitochondrial respiratory efficiency via mitochondrial remodeling and (2) reduces blood glucose in a hepatic HSP72-dependent manner.
Collapse
Affiliation(s)
- Alex T Von Schulze
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Fengyan Deng
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Kelly N Z Fuller
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Edziu Franczak
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Josh Miller
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Julie Allen
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Colin S McCoin
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Kartik Shankar
- Pediatrics, Section of Nutrition, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Wen-Xing Ding
- Pharmacology, Toxicology & Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - John P Thyfault
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Paige C Geiger
- Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, KS 66160, USA,Address correspondence to P.C.G. (e-mail: )
| |
Collapse
|
13
|
Yan Z, Zhan J, Qi W, Lin J, Huang Y, Xue X, Pan X. The Protective Effect of Luteolin in Glucocorticoid-Induced Osteonecrosis of the Femoral Head. Front Pharmacol 2020; 11:1195. [PMID: 32903480 PMCID: PMC7435053 DOI: 10.3389/fphar.2020.01195] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/22/2020] [Indexed: 01/22/2023] Open
Abstract
Glucocorticoid-induced osteonecrosis of the femoral head (GIONFH) is a frequently occurring type of nontraumatic osteonecrosis. A failure of the timely treatment can eventually result in the collapse of the subchondral bone structure. Luteolin (Lut), a compound extracted from Rhizoma Drynariae, is reported to possess multiple pharmacological properties including anticancer, antioxidant, antiapoptosis, and antiinflammatory properties. However, whether Lut has a protective effect on the development of GIONFH remains unclear. In this study, we evaluated the effect of Lut on Dexamethasone (Dex)-induced STAT1/caspase3 pathway in vitro and evaluated GIONFH model in vivo. In vitro, Lut inhibited the upregulation of Dex-induced phospho-STAT1, cleaved caspase9, and cleaved caspase3. In addition, Lut inhibited Dex-induced expression of Bax and cytochrome c and increased the expression of B cell lymphoma-2(Bcl-2). In vivo, Lut decreased the proportion of empty lacunae in rats with GIONFH. Taken together, these findings indicate that Lut may have therapeutic potential in the treatment of GIONFH. Further, this effect might be achieved by suppressing mitochondrial apoptosis of osteoblasts via inhibition of STAT1 activity.
Collapse
Affiliation(s)
- Zijian Yan
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthpaedics, Wenzhou, China.,The Second School of Medicine, WenZhou Medical University, Wenzhou, China
| | - Jingdi Zhan
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthpaedics, Wenzhou, China.,The Second School of Medicine, WenZhou Medical University, Wenzhou, China
| | - Weihui Qi
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthpaedics, Wenzhou, China.,The Second School of Medicine, WenZhou Medical University, Wenzhou, China
| | - Jian Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yijiang Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinghe Xue
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyun Pan
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
Milani A, Basirnejad M, Bolhassani A. Heat-shock proteins in diagnosis and treatment: an overview of different biochemical and immunological functions. Immunotherapy 2020; 11:215-239. [PMID: 30730280 DOI: 10.2217/imt-2018-0105] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Heat-shock proteins (HSPs) have been involved in different functions including chaperone activity, protein folding, apoptosis, autophagy and immunity. The HSP families have powerful effects on the stimulation of innate immune responses through Toll-like receptors and scavenger receptors. Moreover, HSP-mediated phagocytosis directly enhances the processing and presentation of internalized antigens via the endocytic pathway in adaptive immune system. These properties of HSPs have been used for development of prophylactic and therapeutic vaccines against infectious and noninfectious diseases. Several studies also demonstrated the relationship between HSPs and drug resistance as well as their use as a novel biomarker for detecting tumors in patients. The present review describes different roles of HSPs in biology and medicine especially biochemical and immunological aspects.
Collapse
Affiliation(s)
- Alireza Milani
- Department of Hepatitis & AIDS, Pasteur Institute of Iran, Tehran, Iran.,Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | | | - Azam Bolhassani
- Department of Hepatitis & AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
15
|
Cao Y, Han X, Pan H, Jiang Y, Peng X, Xiao W, Rong J, Chen F, He J, Zou L, Tang Y, Pei Y, Zheng J, Wang J, Zhong J, Hong X, Liu Z, Zheng Z. Emerging protective roles of shengmai injection in septic cardiomyopathy in mice by inducing myocardial mitochondrial autophagy via caspase-3/Beclin-1 axis. Inflamm Res 2020; 69:41-50. [PMID: 31712853 DOI: 10.1007/s00011-019-01292-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/04/2019] [Accepted: 10/11/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Sepsis, a life-threatening systemic syndrome related to inflammatory response, usually accompanied by major organ dysfunctions. The aim of the present study was to elucidate the role by which Shengmai injection (SMI) acts to septic cardiomyopathy. METHODS Initially, the induced mice with septic cardiomyopathy were treated with SMI or normal saline (NS) with oe-caspase-3, and HL-1 cells were treated with oe-Beclin-1 and oe-caspase-3 and then cultured with lipopolysaccharide (LPS). Subsequently, we measured the cardiac troponin I (cTnI) level, and expression of mitochondrial autophagy protein (parkin and pink1) and myocardial cell autophagy-related proteins (LC3-II and LC3-I). Additionally, we identified the cleavage of Beclin-1 by caspase-3 and detected the changes of mitochondrial membrane potential, level of reactive oxygen species (ROS), and apoptosis of myocardial cells in myocardial tissues of mice. RESULTS It has been demonstrated that SMI contributed to the increase of myocardial mitochondrial autophagy, reduction of cTnI level, and elevation of mitochondrial membrane potential in septic cardiomyopathy mice. Both in vitro and in vivo experiments showed that caspase-3 promoted cleavage of Beclin-1 and release of ROS, whereas repressed lipopolysaccharide (LPS)-induced mitochondrial autophagy. Furthermore, the facilitation of myocardial mitochondrial autophagy and protection of myocardial mitochondria by SMI through inhibition of cleavage Beclin-1 by caspase-3 in septic cardiomyopathy mice were also proved by in vivo experiments. CONCLUSION Taken together, SMI could protect myocardial mitochondria by promoting myocardial mitochondrial autophagy in septic cardiomyopathy via inhibition of cleavage of Beclin-1 by caspase-3. Our study demonstrates that SMI could represent a novel target for treatment of septic cardiomyopathy.
Collapse
Affiliation(s)
- Yan Cao
- Department of Emergency, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), Changsha, 410000, People's Republic of China.,Chest Pain Center of Hunan, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China
| | - Xiaotong Han
- Department of Emergency, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), Changsha, 410000, People's Republic of China.,Chest Pain Center of Hunan, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China
| | - Hongwei Pan
- Chest Pain Center of Hunan, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China.,Department of Cardiology, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China
| | - Yu Jiang
- Hunan Provincial Institute of Emergency Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410000, People's Republic of China.,Hunan Provincial Key Laboratory of Emergency and Critical Care Metabolomics, Changsha, 410000, People's Republic of China
| | - Xiang Peng
- Chest Pain Center of Hunan, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China.,Department of Cardiology, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China
| | - Weiwei Xiao
- Department of Emergency, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), Changsha, 410000, People's Republic of China.,Chest Pain Center of Hunan, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China
| | - Jingjing Rong
- Department of Cardiology, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China
| | - Fang Chen
- Department of Emergency, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), Changsha, 410000, People's Republic of China.,Hunan Provincial Institute of Emergency Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410000, People's Republic of China
| | - Jin He
- Department of Cardiology, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China
| | - Lianhong Zou
- Chest Pain Center of Hunan, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China.,Department of Cardiology, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China
| | - Yi Tang
- Department of Cardiology, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China
| | - Yanfang Pei
- Department of Emergency, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), Changsha, 410000, People's Republic of China
| | - Jiao Zheng
- Institute of Clinical Pharmacology Research, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410000, People's Republic of China
| | - Jia Wang
- Department of Research, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410000, People's Republic of China
| | - Jie Zhong
- Department of Research, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410000, People's Republic of China
| | - Xiuqing Hong
- Department of Research, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410000, People's Republic of China
| | - Zhengyu Liu
- Chest Pain Center of Hunan, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China. .,Department of Cardiology, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China.
| | - Zhaofen Zheng
- Chest Pain Center of Hunan, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China. .,Department of Cardiology, Hunan Provincial People's Hospital (The Frist Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410000, People's Republic of China.
| |
Collapse
|
16
|
Abosheasha MA, Abd El Khalik EAM, El-Gowily AH. Indispensable Role of Protein Turnover in Autophagy, Apoptosis and Ubiquitination Pathways. HEAT SHOCK PROTEINS 2020:447-468. [DOI: 10.1007/7515_2020_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
17
|
Das CK, Banerjee I, Mandal M. Pro-survival autophagy: An emerging candidate of tumor progression through maintaining hallmarks of cancer. Semin Cancer Biol 2019; 66:59-74. [PMID: 31430557 DOI: 10.1016/j.semcancer.2019.08.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/30/2019] [Accepted: 08/16/2019] [Indexed: 12/13/2022]
Abstract
Autophagy is an evolutionary conserved catabolic process that regulates the cellular homeostasis by targeting damaged cellular contents and organelles for lysosomal degradation and sustains genomic integrity, cellular metabolism, and cell survival during diverse stress and adverse conditions. Recently, the role of autophagy is extremely debated in the regulation of cancer initiation and progression. Although autophagy has a dichotomous role in the regulation of cancer, growing numbers of studies largely indicate the pro-survival role of autophagy in cancer progression and metastasis. In this review, we discuss the detailed mechanisms of autophagy, the role of pro-survival autophagy that positively drives several classical as well as emerging hallmarks of cancer for tumorigenic progression, and also we address various autophagy inhibitors that could be harnessed against pro-survival autophagy for effective cancer therapeutics. Finally, we highlight some outstanding problems that need to be deciphered extensively in the future to unravel the role of autophagy in tumor progression.
Collapse
Affiliation(s)
- Chandan Kanta Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Indranil Banerjee
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India.
| |
Collapse
|
18
|
Bloemberg D, Quadrilatero J. Autophagy, apoptosis, and mitochondria: molecular integration and physiological relevance in skeletal muscle. Am J Physiol Cell Physiol 2019; 317:C111-C130. [PMID: 31017800 DOI: 10.1152/ajpcell.00261.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Apoptosis and autophagy are processes resulting from the integration of cellular stress and death signals. Their individual importance is highlighted by the lethality of various mouse models missing apoptosis or autophagy-related genes. In addition to their independent roles, significant overlap exists with respect to the signals that stimulate these processes as well as their effector consequences. While these cellular systems exemplify the programming redundancies that underlie many fundamental biological mechanisms, their intertwined relationship means that dysfunction can promote pathology. Although both autophagic and apoptotic signaling are active in skeletal muscle during various diseases and atrophy, their specific roles here are somewhat unique. Given our growing understanding of how specific changes at the cellular level impact whole-organism physiology, there is an equally growing interest in pharmacological manipulation of apoptosis and/or autophagy for altering human physiology and health.
Collapse
Affiliation(s)
- Darin Bloemberg
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| |
Collapse
|
19
|
Zhang X, Du J, Guo Z, Yu J, Gao Q, Yin W, Zhu S, Gu Z, Zhao Y. Efficient Near Infrared Light Triggered Nitric Oxide Release Nanocomposites for Sensitizing Mild Photothermal Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801122. [PMID: 30775223 PMCID: PMC6364593 DOI: 10.1002/advs.201801122] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 11/15/2018] [Indexed: 04/14/2023]
Abstract
Mild photothermal therapy (PTT), as a new anticancer therapeutic strategy, faces big challenges of limited therapeutic accuracy and side-effects due to uneven heat distribution. Here, near infrared triggered nitric oxide (NO) release nanocomposites based on bismuth sulfide (Bi2S3) nanoparticles and bis-N-nitroso compounds (BNN) are constructed for NO-enhanced mild photothermal therapy. Upon 808 nm irradiation, the high photothermal conversion efficiency and on-demand NO release are realized simultaneously. Due to the unique properties of NO, enhanced antitumor efficacy of mild PTT based on BNN-Bi2S3 nanocomposites is achieved in vitro and in vivo. Mechanism studies reveal that the exogenous NO from BNN-Bi2S3 could not only impair the autophagic self-repairing ability of tumor cells in situ, but also diffuse to the surrounding cells to enhance the therapeutic effect. This work points out a strategy to overcome the difficulties in mild PTT, and has potentials for further exploitation of NO-sensitized synergistic cancer therapy.
Collapse
Affiliation(s)
- Xiao Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy Physics and National Center for Nanosciences and TechnologyChinese Academy of SciencesBeijing100049China
| | - Jiangfeng Du
- Department of Medical ImagingShanxi Medical UniversityTaiyuanShanxi030001China
| | - Zhao Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy Physics and National Center for Nanosciences and TechnologyChinese Academy of SciencesBeijing100049China
- College of Materials Science and Optoelectronic TechnologyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Jie Yu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy Physics and National Center for Nanosciences and TechnologyChinese Academy of SciencesBeijing100049China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of EducationSchool of Life Science and TechnologyXidian UniversityXi'anShaanxi710126China
| | - Qin Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy Physics and National Center for Nanosciences and TechnologyChinese Academy of SciencesBeijing100049China
| | - Wenyan Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy Physics and National Center for Nanosciences and TechnologyChinese Academy of SciencesBeijing100049China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy Physics and National Center for Nanosciences and TechnologyChinese Academy of SciencesBeijing100049China
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy Physics and National Center for Nanosciences and TechnologyChinese Academy of SciencesBeijing100049China
- College of Materials Science and Optoelectronic TechnologyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy Physics and National Center for Nanosciences and TechnologyChinese Academy of SciencesBeijing100049China
- College of Materials Science and Optoelectronic TechnologyUniversity of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
20
|
LC3/GABARAPs drive ubiquitin-independent recruitment of Optineurin and NDP52 to amplify mitophagy. Nat Commun 2019; 10:408. [PMID: 30679426 PMCID: PMC6345886 DOI: 10.1038/s41467-019-08335-6] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 12/21/2018] [Indexed: 12/17/2022] Open
Abstract
Current models of selective autophagy dictate that autophagy receptors, including Optineurin and NDP52, link cargo to autophagosomal membranes. This is thought to occur via autophagy receptor binding to Atg8 homologs (LC3/GABARAPs) through an LC3 interacting region (LIR). The LIR motif within autophagy receptors is therefore widely recognised as being essential for selective sequestration of cargo. Here we show that the LIR motif within OPTN and NDP52 is dispensable for Atg8 recruitment and selectivity during PINK1/Parkin mitophagy. Instead, Atg8s play a critical role in mediating ubiquitin-independent recruitment of OPTN and NDP52 to growing phagophore membranes via the LIR motif. The additional recruitment of OPTN and NDP52 amplifies mitophagy through an Atg8-dependent positive feedback loop. Rather than functioning in selectivity, our discovery of a role for the LIR motif in mitophagy amplification points toward a general mechanism by which Atg8s can recruit autophagy factors to drive autophagosome growth and amplify selective autophagy.
Collapse
|
21
|
Parousis A, Carter HN, Tran C, Erlich AT, Mesbah Moosavi ZS, Pauly M, Hood DA. Contractile activity attenuates autophagy suppression and reverses mitochondrial defects in skeletal muscle cells. Autophagy 2018; 14:1886-1897. [PMID: 30078345 PMCID: PMC6152519 DOI: 10.1080/15548627.2018.1491488] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 06/05/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022] Open
Abstract
Macroautophagy/autophagy is a survival mechanism that facilitates protein turnover in post-mitotic cells in a lysosomal-dependent process. Mitophagy is a selective form of autophagy, which arbitrates the selective recognition and targeting of aberrant mitochondria for degradation. Mitochondrial content in cells is the net balance of mitochondrial catabolism via mitophagy, and organelle biogenesis. Although the latter process has been well described, mitophagy in skeletal muscle is less understood, and it is currently unknown how these two opposing mechanisms converge during contractile activity. Here we show that chronic contractile activity (CCA) in muscle cells induced mitochondrial biogenesis and coordinately enhanced the expression of TFEB (transcription factor EB) and PPARGC1A/PGC-1α, master regulators of lysosome and mitochondrial biogenesis, respectively. CCA also enhanced the expression of PINK1 and the lysosomal protease CTSD (cathepsin D). Autophagy blockade with bafilomycin A1 (BafA) reduced mitochondrial state 3 and 4 respiration, increased ROS production and enhanced the accumulation of MAP1LC3B-II/LC3-II and SQSTM1/p62. CCA ameliorated this mitochondrial dysfunction during defective autophagy, increased PPARGC1A, normalized LC3-II levels and reversed mitochondrially-localized SQSTM1 toward control levels. NAC emulated the LC3-II reductions induced by contractile activity, signifying that a decrease in oxidative stress could represent a mechanism of autophagy normalization brought about by CCA. CCA enhances mitochondrial biogenesis and lysosomal activity, and normalizes autophagy flux during autophagy suppression, partly via ROS-dependent mechanisms. Thus, contractile activity represents a potential therapeutic intervention for diseases in which autophagy is inhibited, such as vacuolar myopathies in skeletal muscle, by establishing a healthy equilibrium of anabolic and catabolic pathways. ABBREVIATIONS AMPK: AMP-activated protein kinase; BafA: bafilomycin A1; BNIP3L: BCL2/adenovirus E1B interacting protein 3-like; CCA: chronic contractile activity; COX4I1: cytochrome c oxidase subunit 4I1; DMEM: Dulbecco's modified Eagle's medium; GFP: green fluorescent protein; LSD: lysosomal storage diseases; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MTORC1: mechanistic target of rapamycin kinase complex 1; NAC: N-acetylcysteine; PPARGC1A: peroxisome proliferative activated receptor, gamma, coactivator 1 alpha; PINK1: PTEN induced putative kinase 1; ROS: reactive oxygen species; SOD2: superoxide dismutase 2, mitochondrial; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB.
Collapse
Affiliation(s)
- Alexa Parousis
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Heather N. Carter
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Claudia Tran
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Avigail T. Erlich
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Zahra S. Mesbah Moosavi
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Marion Pauly
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - David A. Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| |
Collapse
|
22
|
Gu X, Han D, Chen W, Zhang L, Lin Q, Gao J, Fanning S, Han B. SIRT1-mediated FoxOs pathways protect against apoptosis by promoting autophagy in osteoblast-like MC3T3-E1 cells exposed to sodium fluoride. Oncotarget 2018; 7:65218-65230. [PMID: 27564107 PMCID: PMC5323150 DOI: 10.18632/oncotarget.11573] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/15/2016] [Indexed: 11/25/2022] Open
Abstract
Fluorine may result in damage to teeth, bones and other body tissues, and is a serious public health problem. SIRT1 deacetylates FOXOs, which brings about apoptosis and autophagy promotion or suppression. Fluorine may induce cell apoptosis, however, the role of autophagy in apoptosis induced by fluorine is still poorly understood, and the interaction between SIRT1 and FOXOs should be further illustrated. Therefore, this study investigated the mechanisms underlying the NaF- induced apoptosis and autophagy in osteoblast-like MC3T3-E1 cells in vitro through activating or inhibiting SIRT1. Via RT-PCR, western blot, flow cytometry assays, fluorescence and laser confocal microscopy, it was found that NaF induced both cell apoptosis and autophagy. Results also showed that NaF up-regulated SIRT1 expression in a dose-dependent manner. The autophagy of MC3T3-E1 was also up- regulated indirectly whilst apoptosis was significantly attenuated when incubated with the SIRT1 activator SRT1720. When SIRT1 inhibitor Ex-527 was used, the latter effects were reversed. Furthermore, SIRT1 increased deacetylation of FoxO1 and promoted the up-regulation of its target substrate Rab7, as well as increase of Bnip3 which was substrate of FoxO3, and we hypothesize that these pathways may cause an increase in autophagic flux and a reduction in apoptosis. In conclusion, SIRT1-induced autophagy enhancement protects against fluoride-induced apoptosis through autophagy induction in MC3T3-E1 cells, which may be associated with a SIRT1-FoxO1-Rab7 axis and a SIRT1-FoxO3-Binp3 axis. The role of SIRT1 in selecting between cell survival and death provides a potential therapeutic strategy in fluorosis.
Collapse
Affiliation(s)
- Xiaolong Gu
- College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing 100193, P R China
| | - Dandan Han
- College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing 100193, P R China
| | - Wei Chen
- College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing 100193, P R China
| | - Limei Zhang
- College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing 100193, P R China
| | - Qianyun Lin
- College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing 100193, P R China
| | - Jian Gao
- College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing 100193, P R China
| | - Séamus Fanning
- UCD-Centre for Food Safety, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Bo Han
- College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing 100193, P R China
| |
Collapse
|
23
|
Liang S, Jin J, Lin B, Gong J, Li Y, He Q. Rapamycin Induces Autophagy and Reduces the Apoptosis of Podocytes Under a Stimulated Condition of Immunoglobulin A Nephropathy. Kidney Blood Press Res 2017; 42:177-187. [PMID: 28427080 DOI: 10.1159/000475484] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/17/2017] [Indexed: 11/19/2022] Open
Abstract
Backgroud/Aims: The aim of this study was to investigate the potential renoprotective effect of rapamycin on the autophagy of podocytes treated with the supernatant of mesangial cells cultured with aggregated IgA1 (aIgA1) from immunoglobulin A nephropathy (IgAN) patients. METHODS Monomeric IgA1 (mIgA1) was isolated from the serum of IgAN patients or healthy volunteers, and then transformed to aIgA1 by heating. Subsequently, the aIgA1-mesangial cell supernatant was prepared by collecting the medium of mouse mesangial cells (MSC1097) cultured with aIgA1 (100 mg/L) from different IgAN patients or healthy volunteers for 48 h. Subsequently mouse podocytes (MPC5) were exposed to the supernatant of the aIgA1-mesangial cells for 24 h, using 100 mg/L aIgA1 from healthy volunteers as the control group or 100 mg/L aIgA1 from IgAN patients as the IgANs group, in RPMI 1640 medium. The MPC5 cells in the IgANs+Rap group were cultured with rapamycin (10 nmol/L) and the supernatant of MSC-1097 cells cultured with aIgA1 from IgAN patients in RPMI 1640 medium. Autophagy was assessed by western blot analysis (LC3, p62), electron microscopy, and immunofluorescence staining (LC3, p62, and CD63). The apoptosis of podocytes was evaluated by flow cytometry, and the expression of apoptosis-associated proteins cleaved-caspase-3 and caspase-3 were determined by western blot analysis. RESULTS Deficient autophagy, which was evident by decreased LC3-II and CD63 levels, caused accumulation of p62, and fewer autophagosomes were observed in the MPC5 cells cultured with the IgAN supernatant, along with stronger expression of cleaved caspase-3 and a higher apoptosis rate. Inhibition of autophagy was alleviated in the IgANs+Rap group. The LC3-II/LC3-I ratio increased by almost 30%, the accumulated p62 amount was reduced by 50%, and the number of autophagosomes per podocyte increased to about 7 times that of the IgAN groups. These results were confirmed by immunofluorescence staining. In addition, the apoptosis rate of MPC5 cells decreased from 19.88% in the IgAN group to 16.78% in the IgANs+Rap group, which was accompanied by a weaker expression level of cleaved caspase-3. CONCLUSIONS Rapamycin can reduce the apoptosis of podocytes by inducing autophagy in IgAN.
Collapse
|
24
|
Heat stress–induced autophagy promotes lactate secretion in cultured immature boar Sertoli cells by inhibiting apoptosis and driving SLC2A3 , LDHA , and SLC16A1 expression. Theriogenology 2017; 87:339-348. [DOI: 10.1016/j.theriogenology.2016.09.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 09/06/2016] [Accepted: 09/07/2016] [Indexed: 01/07/2023]
|
25
|
Xing F, Zhan Q, He Y, Cui J, He S, Wang G. 1800MHz Microwave Induces p53 and p53-Mediated Caspase-3 Activation Leading to Cell Apoptosis In Vitro. PLoS One 2016; 11:e0163935. [PMID: 27689798 PMCID: PMC5045209 DOI: 10.1371/journal.pone.0163935] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 04/17/2016] [Indexed: 12/23/2022] Open
Abstract
Recent studies have reported that exposure of mammalian cells to microwave radiation may have adverse effects such as induction of cell apoptosis. However, the molecular mechanisms underlying microwave induced mammalian cell apoptosis are not fully understood. Here, we report a novel mechanism: exposure to 1800MHz microwave radiation induces p53-dependent cell apoptosis through cytochrome c-mediated caspase-3 activation pathway. We first measured intensity of microwave radiation from several electronic devices with an irradiation detector. Mouse NIH/3T3 and human U-87 MG cells were then used as receivers of 1800MHz electromagnetic radiation (EMR) at a power density of 1209 mW/m2. Following EMR exposure, cells were analyzed for viability, intracellular reactive oxygen species (ROS) generation, DNA damage, p53 expression, and caspase-3 activity. Our analysis revealed that EMR exposure significantly decreased viability of NIH/3T3 and U-87 MG cells, and increased caspase-3 activity. ROS burst was observed at 6 h and 48 h in NIH/3T3 cells, while at 3 h in U-87 MG cells. Hoechst 33258 staining and in situ TUNEL assay detected that EMR exposure increased DNA damage, which was significantly restrained in the presence of N-acetyl-L-cysteine (NAC, an antioxidant). Moreover, EMR exposure increased the levels of p53 protein and p53 target gene expression, promoted cytochrome c release from mitochondrion, and increased caspase-3 activity. These events were inhibited by pretreatment with NAC, pifithrin-α (a p53 inhibitor) and caspase inhibitor. Collectively, our findings demonstrate, for the first time, that 1800MHz EMR induces apoptosis-related events such as ROS burst and more oxidative DNA damage, which in turn promote p53-dependent caspase-3 activation through release of cytochrome c from mitochondrion. These findings thus provide new insights into physiological mechanisms underlying microwave-induced cell apoptosis.
Collapse
Affiliation(s)
- Fuqiang Xing
- SCNU-ZJU Joint Research Center of Photonics, South China Academy of Advanced Optoelectronics, South China Normal University (SCNU), 510006 Guangzhou, China
- Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China
| | - Qiuqiang Zhan
- SCNU-ZJU Joint Research Center of Photonics, South China Academy of Advanced Optoelectronics, South China Normal University (SCNU), 510006 Guangzhou, China
| | - Yiduo He
- Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China
| | - Jiesheng Cui
- Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China
| | - Sailing He
- SCNU-ZJU Joint Research Center of Photonics, South China Academy of Advanced Optoelectronics, South China Normal University (SCNU), 510006 Guangzhou, China
- Department of Electromagnetic Engineering, Royal Institute of Technology (KTH), 10044 Stockholm, Sweden
- * E-mail: (SH); (GW)
| | - Guanyu Wang
- Department of Biology, South University of Science and Technology of China (SUSTC), Shenzhen 518055, China
- * E-mail: (SH); (GW)
| |
Collapse
|
26
|
Yang X, Niu B, Wang L, Chen M, Kang X, Wang L, Ji Y, Zhong J. Autophagy inhibition enhances colorectal cancer apoptosis induced by dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235. Oncol Lett 2016; 12:102-106. [PMID: 27347108 PMCID: PMC4906634 DOI: 10.3892/ol.2016.4590] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 05/17/2016] [Indexed: 12/31/2022] Open
Abstract
Phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway performs a central role in tumorigenesis and is constitutively activated in many malignancies. As a novel dual PI3K/mTOR inhibitor currently undergoing evaluation in a phase I/II clinical trial, NVP-BEZ235 indicates a significant antitumor efficacy in diverse solid tumors, including colorectal cancer (CRC). Autophagy is a catabolic process that maintains cellular homeostasis and reduces diverse stresses through lysosomal recycling of the unnecessary and damaged cell components. This process is also observed to antagonize the antitumor efficacy of PI3K/mTOR inhibitor agents such as NVP-BEZ235, via apoptosis inhibition. In the present study, we investigated anti-proliferative and apoptosis-inducing ability of NVP-BEZ235 in SW480 cells and the crosstalk between autophagy and apoptosis in SW480 cells treated with NVP-BEZ235 in combination with an autophagy inhibitor. The results revealed that, NVP-BEZ235 effectively inhibit the growth of SW480 cells by targeting the PI3K/mTOR signaling pathway and induced apoptosis. The inhibition of autophagy with 3-methyladenine or chloroquine inhibitors in combination with NVP-BEZ235 in SW480 cells enhanced the apoptotic rate as componets to NVP-BEZ235 alone. In conclusion, the findings provide a rationale for chemotherapy targeting the PI3K/mTOR signaling pathway presenting a potential therapeutic strategy to enhance the efficacy of dual PI3K/mTOR inhibitor NVP-BEZ235 in combination with an autophagy inhibitor in CRC treatment and treatment of other tumors.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Bingxuan Niu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Libo Wang
- Department of Gastroenterology, The First Affiliated Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Meiling Chen
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Xiaochun Kang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Luonan Wang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Yinghua Ji
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Jiateng Zhong
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
27
|
Deng X, Deng T, Ni Y, Zhan Y, Huang W, Liu J, Liao C. Cytochrome c modulates the mitochondrial signaling pathway and polymorphonuclear neutrophil apoptosis in bile duct-ligated rats. Exp Ther Med 2016; 12:333-342. [PMID: 27347059 DOI: 10.3892/etm.2016.3313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/04/2016] [Indexed: 12/15/2022] Open
Abstract
It has been observed that polymorphonuclear neutrophils (PMN) increase in number and function during obstructive jaundice (OJ). However, the precise mechanisms underlying PMN apoptosis during OJ remain poorly understood. The aim of the present study was to investigate the modulation of cytochrome c (Cytc) on the mitochondrial signaling pathway in bile duct-ligated (BDL) rats and the effect on PMN apoptosis following the intravenous administration of Cytc. Rats were randomly divided into four groups: A control group, a sham group, a BDL group and a BDL + Cytc group (rats with common bile duct ligation as well as Cytc intravenous injection). Blood samples were collected from the inferior vein cava for biochemical analysis and separation of the PMN. PMN apoptosis was evaluated using flow cytometry. The mitochondrial membrane potential (ΔΨm) of PMN was detected by rhodamine-123 staining. The Cytc protein expression levels were examined using western blotting. PMN mitochondria were observed using transmission electron microscopy. The results of the present study revealed that the PMN apoptosis rate in rats decreased gradually from 12 to 72 h following BDL to levels that were significantly lower than those of the control group and the sham group. Compared with the corresponding time point of the BDL group, the BDL + Cytc group showed a significantly increased PMN apoptosis rate. The mean fluorescence intensity (MFI) of ΔΨm decreased from 12 to 72 h following BDL, and was significantly increased compared with the control and sham groups. MFI in the BDL + Cytc group was higher compared with that in the BDL group. Cytc expression levels increased in the mitochondria and decreased in the cytoplasm from the 12 to 72 h in the BDL group, which was significantly different from that in the control and sham groups at the corresponding time points. Compared with the BDL group, Cytc expression levels in the cytoplasm for the BDL + Cytc group tended to gradually and significantly increase. Morphological changes in PMN mitochondria were marginal in BDL rats and marked in the BDL + Cytc group. In the BDL rats, PMN apoptosis was inhibited, a process induced by the mitochondrial apoptotic signaling pathway in which Cytc has an important role. High ΔΨm in the mitochondria and decreased Cytc expression levels in the cytoplasm result in PMN apoptosis inhibition. Intravenous injection of Cytc may help compensate for the lack of Cytc proteins in the cytoplasm, inducing PMN apoptosis following BDL.
Collapse
Affiliation(s)
- Xuesong Deng
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China; Department of Hepatobiliary Surgery, The Second People's Hospital of Shenzhen (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong 518035, P.R. China
| | - Tongming Deng
- Department of General Surgery, Baoan Central Hospital of Shenzhen, Shenzhen, Guangdong 518102, P.R. China
| | - Yong Ni
- Department of Hepatobiliary Surgery, The Second People's Hospital of Shenzhen (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong 518035, P.R. China
| | - Yongqiang Zhan
- Department of Hepatobiliary Surgery, The Second People's Hospital of Shenzhen (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong 518035, P.R. China
| | - Wenlong Huang
- Department of Hepatobiliary Surgery, The Second People's Hospital of Shenzhen (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong 518035, P.R. China
| | - Jianfeng Liu
- Department of Hepatobiliary Surgery, The Second People's Hospital of Shenzhen (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong 518035, P.R. China
| | - Caixian Liao
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
28
|
Novoderezhkina EA, Zhivotovsky BD, Gogvadze VG. Induction of unspecific permeabilization of mitochondrial membrane and its role in cell death. Mol Biol 2016. [DOI: 10.1134/s0026893316010167] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Deng X, Zhang F, Wang L, Rui W, Long F, Zhao Y, Chen D, Ding W. Airborne fine particulate matter induces multiple cell death pathways in human lung epithelial cells. Apoptosis 2015; 19:1099-112. [PMID: 24722831 DOI: 10.1007/s10495-014-0980-5] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Our group was the first one reporting that autophagy could be triggered by airborne fine particulate matter (PM) with a mean diameter of less than 2.5 μm (PM2.5) in human lung epithelial A549 cells, which could potentially lead to cell death. In the present study, we further explored the potential interactions between autophagy and apoptosis because it was well documented that PM2.5 could induce apoptosis in A549 cells. Much to our surprise, we found that PM2.5-exposure caused oxidative stress, resulting in activation of multiple cell death pathways in A549 cells, that is, the tumor necrosis factor-alpha (TNF-α)-induced pathway as evidenced by TNF-α secretion and activation of caspase-8 and -3, the intrinsic apoptosis pathway as evidenced by increased expression of pro-apoptotic protein Bax, decreased expression of anti-apoptotic protein Bcl-2, disruption of mitochondrial membrane potential, and activation of caspase-9 and -3, and autophagy as evidenced by an increased number of double-membrane vesicles, accompanied by increases of conversion and punctuation of microtubule-associated proteins light chain 3 (LC3) and expression of Beclin 1. It appears that reactive oxygen species (ROS) function as signaling molecules for all the three pathways because pretreatment with N-acetylcysteine, a scavenger of ROS, almost completely abolished TNF-α secretion and significantly reduced the number of apoptotic and autophagic cells. In another aspect, inhibiting autophagy with 3-methyladenine, a specific autophagy inhibitor, enhanced PM2.5-induced apoptosis and cytotoxicity. Intriguingly, neutralization of TNF-α with an anti-TNF-α special antibody not only abolished activation of caspase-8, but also drastically reduced LC3-II conversion. Thus, the present study has provided novel insights into the mechanism of cytotoxicity and even pathogenesis of diseases associated with PM2.5 exposure.
Collapse
Affiliation(s)
- Xiaobei Deng
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Tan K, Fujimoto M, Takii R, Takaki E, Hayashida N, Nakai A. Mitochondrial SSBP1 protects cells from proteotoxic stresses by potentiating stress-induced HSF1 transcriptional activity. Nat Commun 2015; 6:6580. [PMID: 25762445 PMCID: PMC4558571 DOI: 10.1038/ncomms7580] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 02/09/2015] [Indexed: 12/13/2022] Open
Abstract
Heat-shock response is an adaptive response to proteotoxic stresses including heat shock, and is regulated by heat-shock factor 1 (HSF1) in mammals. Proteotoxic stresses challenge all subcellular compartments including the mitochondria. Therefore, there must be close connections between mitochondrial signals and the activity of HSF1. Here, we show that heat shock triggers nuclear translocation of mitochondrial SSBP1, which is involved in replication of mitochondrial DNA, in a manner dependent on the mitochondrial permeability transition pore ANT–VDAC1 complex and direct interaction with HSF1. HSF1 recruits SSBP1 to the promoters of genes encoding cytoplasmic/nuclear and mitochondrial chaperones. HSF1–SSBP1 complex then enhances their induction by facilitating the recruitment of a chromatin-remodelling factor BRG1, and supports cell survival and the maintenance of mitochondrial membrane potential against proteotoxic stresses. These results suggest that the nuclear translocation of mitochondrial SSBP1 is required for the regulation of cytoplasmic/nuclear and mitochondrial proteostasis against proteotoxic stresses. Heat shock induces proteotoxic stress, and the cellular response is mediated by heat-shock factor 1 (HSF1). Here, Tan et al. show that following heat shock, mitochondrial SSBP1 translocates to the nucleus and binds HSF1 to enhance the expression of chaperones and support the maintenance of mitochondrial function.
Collapse
Affiliation(s)
- Ke Tan
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| | - Mitsuaki Fujimoto
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| | - Ryosuke Takii
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| | - Eiichi Takaki
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| | - Naoki Hayashida
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| | - Akira Nakai
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| |
Collapse
|
31
|
Katagiri N, Kuroda T, Kishimoto H, Hayashi Y, Kumazawa T, Kimura K. The nucleolar protein nucleophosmin is essential for autophagy induced by inhibiting Pol I transcription. Sci Rep 2015; 5:8903. [PMID: 25754892 PMCID: PMC4354046 DOI: 10.1038/srep08903] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 02/10/2015] [Indexed: 12/19/2022] Open
Abstract
Various cellular stresses activate autophagy, which is involved in lysosomal degradation of cytoplasmic materials for maintaining nutrient homeostasis and eliminating harmful components. Here, we show that RNA polymerase I (Pol I) transcription inhibition induces nucleolar disruption and autophagy. Treatment with autophagy inhibitors or siRNA specific for autophagy-related (ATG) proteins inhibited autophagy but not nucleolar disruption induced by Pol I transcription inhibition, which suggested that nucleolar disruption was upstream of autophagy. Furthermore, treatment with siRNA specific for nucleolar protein nucleophosmin (NPM) inhibited this type of autophagy. This showed that NPM was involved in autophagy when the nucleolus was disrupted by Pol I inhibition. In contrast, NPM was not required for canonical autophagy induced by nutrient starvation, as it was not accompanied by nucleolar disruption. Thus, our results revealed that, in addition to canonical autophagy, there may be NPM-dependent autophagy associated with nucleolar disruption.
Collapse
Affiliation(s)
- Naohiro Katagiri
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennnoudai, Tsukuba 305-8577, Japan
| | - Takao Kuroda
- Center of Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennnoudai, Tsukuba 305-8577, Japan
| | - Hiroyuki Kishimoto
- 1] Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennnoudai, Tsukuba 305-8577, Japan [2] Center of Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennnoudai, Tsukuba 305-8577, Japan
| | - Yuki Hayashi
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennnoudai, Tsukuba 305-8577, Japan
| | - Takuya Kumazawa
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennnoudai, Tsukuba 305-8577, Japan
| | - Keiji Kimura
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennnoudai, Tsukuba 305-8577, Japan
| |
Collapse
|
32
|
Wu H, Che X, Zheng Q, Wu A, Pan K, Shao A, Wu Q, Zhang J, Hong Y. Caspases: a molecular switch node in the crosstalk between autophagy and apoptosis. Int J Biol Sci 2014; 10:1072-83. [PMID: 25285039 PMCID: PMC4183927 DOI: 10.7150/ijbs.9719] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/20/2014] [Indexed: 12/16/2022] Open
Abstract
Autophagy and apoptosis are two important catabolic processes contributing to the maintenance of cellular and tissue homeostasis. Autophagy controls the turnover of protein aggregates and damaged organelles within cells, while apoptosis is the principal mechanism by which unwanted cells are dismantled and eliminated from organisms. Despite marked differences between these two pathways, they are highly interconnected in determining the fate of cells. Intriguingly, caspases, the primary drivers of apoptotic cell death, play a critical role in mediating the complex crosstalk between autophagy and apoptosis. Pro-apoptotic signals can converge to activate caspases to execute apoptotic cell death. In addition, activated caspases can degrade autophagy proteins (i.e., Beclin-1, Atg5, and Atg7) to shut down the autophagic response. Moreover, caspases can convert pro-autophagic proteins into pro-apoptotic proteints to trigger apoptotic cell death instead. It is clear that caspases are important in both apoptosis and autophagy, thus a detailed deciphering of the role of caspases in these two processes is still required to clarify the functional relationship between them. In this article, we provide a current overview of caspases in its interplay between autophagy and apoptosis. We emphasized that defining the role of caspases in autophagy-apoptosis crosstalk will provide a framework for more precise manipulation of these two processes during cell death.
Collapse
Affiliation(s)
- Haijian Wu
- 1. Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoru Che
- 2. Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Qiaoli Zheng
- 3. Clinical Research Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - An Wu
- 1. Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kun Pan
- 4. Department of Neurological Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Anwen Shao
- 1. Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qun Wu
- 1. Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- 1. Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuan Hong
- 1. Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
33
|
Dirks-Naylor AJ, Kouzi SA, Bero JD, Tran NTK, Yang S, Mabolo R. Effects of acute doxorubicin treatment on hepatic proteome lysine acetylation status and the apoptotic environment. World J Biol Chem 2014; 5:377-386. [PMID: 25225604 PMCID: PMC4160530 DOI: 10.4331/wjbc.v5.i3.377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/02/2014] [Accepted: 06/27/2014] [Indexed: 02/05/2023] Open
Abstract
AIM: To determine if doxorubicin (Dox) alters hepatic proteome acetylation status and if acetylation status was associated with an apoptotic environment.
METHODS: Doxorubicin (20 mg/kg; Sigma, Saint Louis, MO; n = 8) or NaCl (0.9%; n = 7) was administered as an intraperitoneal injection to male F344 rats, 6-wk of age. Once animals were treated with Dox or saline, all animals were fasted until sacrifice 24 h later.
RESULTS: Dox treatment decreased proteome lysine acetylation likely due to a decrease in histone acetyltransferase activity. Proteome deacetylation may likely not be associated with a proapoptotic environment. Dox did not increase caspase-9, -8, or -3 activation nor poly (adenosine diphosphate-ribose) polymerase-1 cleavage. Dox did stimulate caspase-12 activation, however, it likely did not play a role in apoptosis induction.
CONCLUSION: Early effects of Dox involve hepatic proteome lysine deacetylation and caspase-12 activation under these experimental conditions.
Collapse
|
34
|
Abstract
Antiviral innate immune responses and apoptosis are the two major factors limiting viral infections. Successful viral infection requires the virus to take advantage of the cellular machinery to bypass cellular defenses. Accumulated evidences show that autophagy plays a crucial role in cell-to-virus interaction. Here, we focus on how viruses subvert mitophagy to favor viral replication by mitigating innate immune responses and apoptotic signaling.
Collapse
Affiliation(s)
- Mao Xia
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | | | | | | |
Collapse
|
35
|
Min JJ, Huo XL, Xiang LY, Qin YQ, Chai KQ, Wu B, Jin L, Wang XT. Protective effect of Dl-3n-butylphthalide on learning and memory impairment induced by chronic intermittent hypoxia-hypercapnia exposure. Sci Rep 2014; 4:5555. [PMID: 24990154 PMCID: PMC4080197 DOI: 10.1038/srep05555] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 06/13/2014] [Indexed: 12/20/2022] Open
Abstract
Cognitive impairment is a common finding in patients with chronic obstructive pulmonary disease (COPD), but little attention has been focused on therapeutic intervention for this complication. Chronic intermittent hypoxia hypercapnia (CIHH) exposure is considered to be responsible for the pathogenesis of COPD. Dl-3n-Butylphthalide (NBP), extracted from Apium graveolens Linn, has displayed a broad spectrum of neuroprotective properties. Our study aimed to investigate the potential of NBP on CIHH-induced cognitive deficits. The cognitive function of rats after CIHH exposure was evaluated by the Morris water maze, which showed that the NBP treated group performed better in the navigation test. NBP activated BDNF and phosphorylated CREB, the both are responsible for neuroprotection. Additionally, NBP decreased CIHH induced apoptosis. Moreover, NBP further induced the expression of HIF-1α, accompanied by the up-regulation of the autophagy proteins Bnip3, Beclin-1 and LC3-II. Finally, NBP also reversed the decreased expression of SIRT1 and PGC-1α, but the expression of Tfam, Cox II and mtDNA remained unchanged. These results suggested that the neuroprotective effects of NBP under CIHH condition possibly occurred through the inhibition of apoptosis, promotion of hypoxia-induced autophagy, and activation of the SIRT1/PGC-1α signalling pathway, while stimulation of mitochondrial biogenesis may not be a characteristic response.
Collapse
Affiliation(s)
- Jing-jing Min
- The Center of Neurology and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
- The First People's Hospital of Huzhou, Huzhou 313000, China
- These authors contributed equally to this work
| | - Xin-long Huo
- The Center of Neurology and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
- These authors contributed equally to this work
| | - ling-yun Xiang
- The Center of Neurology and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yan-qing Qin
- The Center of Neurology and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Ke-qin Chai
- The Center of Neurology and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Bin Wu
- Wenzhou Medical University, Wenzhou 325027, China
| | - Lu Jin
- The Center of Neurology and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiao-tong Wang
- The Center of Neurology and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
36
|
Dirks-Naylor AJ, Kouzi SA, Bero JD, Phan DT, Taylor HN, Whitt SD, Mabolo R. Doxorubicin alters the mitochondrial dynamics machinery and mitophagy in the liver of treated animals. Fundam Clin Pharmacol 2014; 28:633-42. [PMID: 24666153 DOI: 10.1111/fcp.12073] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 02/17/2014] [Accepted: 03/07/2014] [Indexed: 12/21/2022]
Affiliation(s)
| | - Samir A. Kouzi
- Wingate University School of Pharmacy; Wingate NC 28174 USA
| | - Joseph D. Bero
- Wingate University School of Pharmacy; Wingate NC 28174 USA
| | - Diep T. Phan
- Wingate University School of Pharmacy; Wingate NC 28174 USA
| | | | | | - Raean Mabolo
- Wingate University School of Pharmacy; Wingate NC 28174 USA
| |
Collapse
|
37
|
Neutelings T, Lambert CA, Nusgens BV, Colige AC. Effects of mild cold shock (25°C) followed by warming up at 37°C on the cellular stress response. PLoS One 2013; 8:e69687. [PMID: 23936078 PMCID: PMC3720612 DOI: 10.1371/journal.pone.0069687] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 06/17/2013] [Indexed: 01/12/2023] Open
Abstract
Temperature variations in cells, tissues and organs may occur in a number of circumstances. We report here that reducing temperature of cells in culture to 25°C for 5 days followed by a rewarming to 37°C affects cell biology and induces a cellular stress response. Cell proliferation was almost arrested during mild hypothermia and not restored upon returning to 37°C. The expression of cold shock genes, CIRBP and RBM3, was increased at 25°C and returned to basal level upon rewarming while that of heat shock protein HSP70 was inversely regulated. An activation of pro-apoptotic pathways was evidenced by FACS analysis and increased Bax/Bcl2 and BclX(S/L) ratios. Concomitant increased expression of the autophagosome-associated protein LC3II and AKT phosphorylation suggested a simultaneous activation of autophagy and pro-survival pathways. However, a large proportion of cells were dying 24 hours after rewarming. The occurrence of DNA damage was evidenced by the increased phosphorylation of p53 and H2AX, a hallmark of DNA breaks. The latter process, as well as apoptosis, was strongly reduced by the radical oxygen species (ROS) scavenger, N-acetylcysteine, indicating a causal relationship between ROS, DNA damage and cell death during mild cold shock and rewarming. These data bring new insights into the potential deleterious effects of mild hypothermia and rewarming used in various research and therapeutical fields.
Collapse
Affiliation(s)
- Thibaut Neutelings
- Laboratory of Connective Tissue Biology, Interdisciplinary Grouping of Applied Genoproteomic-Research, University of Liège, Liège, Belgium.
| | | | | | | |
Collapse
|
38
|
Chang Z, Shi G, Jin J, Guo H, Guo X, Luo F, Song Y, Jia X. Dual PI3K/mTOR inhibitor NVP-BEZ235-induced apoptosis of hepatocellular carcinoma cell lines is enhanced by inhibitors of autophagy. Int J Mol Med 2013; 31:1449-56. [PMID: 23588698 DOI: 10.3892/ijmm.2013.1351] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/08/2013] [Indexed: 12/21/2022] Open
Abstract
Dysregulation of the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling has been found in several types of human cancer, including hepatocellular carcinoma (HCC). NVP-BEZ235 is a novel, orally bioavailable dual PI3K/mTOR inhibitor that has exhibited promising activity against HCC in preclinical models. Autophagy is a cellular lysosomal degradation pathway essential for the regulation of cell survival and death to maintain homeostasis. This process is negatively regulated by mTOR signaling and often counteracts the efficacy of certain cancer therapeutic agents. In this study, we explored the role of autophagy in apoptosis induced by NVP-BEZ235 in two HCC cell lines, Hep3B and PLC/PRF/5, and identified the mechanism of combinatorial treatment. NVP-BEZ235 was effective in inhibiting the growth of the two HCC cell lines possibly though induction of apoptosis. NVP-BEZ235 also potently increased the expression of LC3-II and decreased the expression of p62, indicating induction of autophagy. When NVP-BEZ235 was used in combination with Atg5 siRNA or the autophagy inhibitor 3-methyladenine (3-MA), enhancement of the inhibitory effects on the growth of HCC cells was detected. In addition, enhanced induction of apoptosis was observed in cells exposed to the combination of NVP-BEZ235 and Atg5 siRNA or 3-MA. Thus, induction of autophagy by NVP-BEZ235 may be a survival mechanism that counteracts its anticancer effects. Based on these data, we suggest a strategy to enhance the anticancer efficacy of BEZ235 by blockade of autophagy. Thus, our study provides a rationale for the clinical development of combinations of NVP-BEZ235 and autophagy inhibitors for the treatment of HCC and other malignancies.
Collapse
Affiliation(s)
- Zhexing Chang
- Department of Oncology, Beihua Affiliated Hospital, Jilin, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Liu D, Yang J, Wang L. Cadmium induces ultrastructural changes in the hepatopancreas of the freshwater crab Sinopotamon henanense. Micron 2013; 47:24-32. [DOI: 10.1016/j.micron.2013.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 01/04/2013] [Accepted: 01/17/2013] [Indexed: 02/08/2023]
|
40
|
Inao T, Harashima N, Monma H, Okano S, Itakura M, Tanaka T, Tajima Y, Harada M. Antitumor effects of cytoplasmic delivery of an innate adjuvant receptor ligand, poly(I:C), on human breast cancer. Breast Cancer Res Treat 2012; 134:89-100. [PMID: 22203435 DOI: 10.1007/s10549-011-1930-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 12/17/2011] [Indexed: 11/26/2022]
Abstract
Innate adjuvant receptors are expressed in immune cells and some types of cancers. If antitumor therapies targeting these receptors are established, it is likely that they will be therapeutically beneficial because antitumor effects and immune-cell activation can be induced simultaneously. In this study, we tested this possibility of using an innate adjuvant receptor ligand, polyinosinic-polycytidylic acid [poly(I:C)], to treat human breast cancer cell lines. Three breast cancer cell lines (MCF-7, MDA-MB-231, and BT-549) were used in this study. Poly(I:C) was transfected into these cancer cells to stimulate melanoma differentiation-associated gene (MDA) 5, which is a cytoplasmic adjuvant receptor. Poly(I:C) transfection significantly reduced the viability of all cell lines in a manner partially dependent on MDA5. Flow cytometeric analyses and immunoblot assays revealed that the antitumor effect depended on both caspase-dependent apoptosis and c-Myc- and cyclinD1-dependent growth arrest. Interestingly, poly(I:C) transfection was accompanied by autophagy, which is thought to protect cancer cells from apoptosis after poly(I:C) transfection. In a xenograft mouse model, local transfection of poly(I:C) significantly inhibited the growth of xenografted MDA-MB-231 cells. Our findings indicate that cytoplasmic delivery of poly(I:C) can induce apoptosis and growth arrest of human breast cancer cells, and that therapy-associated autophagy prevents apoptosis. The results of this study suggest that the innate adjuvant receptors are promising targets and that their ligands could serve as antitumor reagents, which have the potential to simultaneously induce antitumor effects and activate immune cells.
Collapse
Affiliation(s)
- Touko Inao
- Department of Immunology, Shimane University Faculty of Medicine, Izumo, Shimane, 693-8501, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Lin CJ, Lee CC, Shih YL, Lin CH, Wang SH, Chen TH, Shih CM. Inhibition of mitochondria- and endoplasmic reticulum stress-mediated autophagy augments temozolomide-induced apoptosis in glioma cells. PLoS One 2012; 7:e38706. [PMID: 22745676 PMCID: PMC3382156 DOI: 10.1371/journal.pone.0038706] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 05/09/2012] [Indexed: 11/25/2022] Open
Abstract
Autophagy is a crucial process for cells to maintain homeostasis and survival through degradation of cellular proteins and organelles, including mitochondria and endoplasmic reticula (ER). We previously demonstrated that temozolomide (TMZ), an alkylating agent for brain tumor chemotherapy, induced reactive oxygen species (ROS)/extracellular signal-regulated kinase (ERK)-mediated autophagy to protect glioma cells from apoptosis. In this study, we investigated the role of mitochondrial damage and ER stress in TMZ-induced cytotoxicity. Mitochondrial depolarization and mitochondrial permeability transition pore (MPTP) opening were observed as a prelude to TMZ-induced autophagy, and these were followed by the loss of mitochondrial mass. Electron transport chain (ETC) inhibitors, such as rotenone (a complex I inhibitor), sodium azide (a complex IV inhibitor), and oligomycin (a complex V inhibitor), or the MPTP inhibitor, cyclosporine A, decreased mitochondrial damage-mediated autophagy, and therefore increased TMZ-induced apoptosis. TMZ treatment triggered ER stress with increased expression of GADD153 and GRP78 proteins, and deceased pro-caspase 12 protein. ER stress consequently induced autophagy through c-Jun N-terminal kinases (JNK) and Ca2+ signaling pathways. Combination of TMZ with 4-phenylbutyrate (4-PBA), an ER stress inhibitor, augmented TMZ-induced cytotoxicity by inhibiting autophagy. Taken together, our data indicate that TMZ induced autophagy through mitochondrial damage- and ER stress-dependent mechanisms to protect glioma cells. This study provides evidence that agents targeting mitochondria or ER may be potential anticancer strategies.
Collapse
Affiliation(s)
- Chien-Ju Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chin-Cheng Lee
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yung-Luen Shih
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Huang Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Hao Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Thay-Hsiung Chen
- Division of Cardiovascular Surgery, Department of Surgery, Cathay General Hospital, Taipei, Taiwan
| | - Chwen-Ming Shih
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Center for Reproductive Medicine and Sciences, Taipei Medical University Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
42
|
Reversine suppresses oral squamous cell carcinoma via cell cycle arrest and concomitantly apoptosis and autophagy. J Biomed Sci 2012; 19:9. [PMID: 22283874 PMCID: PMC3299600 DOI: 10.1186/1423-0127-19-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 01/27/2012] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The effective therapies for oral cancer patients of stage III and IV are generally surgical excision and radiation combined with adjuvant chemotherapy using 5-Fu and Cisplatin. However, the five-year survival rate is still less than 30% in Taiwan. Therefore, evaluation of effective drugs for oral cancer treatment is an important issue. Many studies indicated that aurora kinases (A, B and C) were potential targets for cancer therapies. Reversine was proved to be a novel aurora kinases inhibitor with lower toxicity recently. In this study, the potentiality for reversine as an anticancer agent in oral squamous cell carcinoma (OSCC) was evaluated. METHODS Effects of reversine on cell growth, cell cycle progress, apoptosis, and autophagy were evaluated mainly by cell counting, flow cytometry, immunoblot, and immunofluorescence. RESULTS The results demonstrated that reversine significantly suppressed the proliferation of two OSCC cell lines (OC2 and OCSL) and markedly rendered cell cycle arrest at G2/M stage. Reversine also induced cell death via both caspase-dependent and -independent apoptosis. In addition, reversine could inhibit Akt/mTORC1 signaling pathway, accounting for its ability to induce autophagy. CONCLUSIONS Taken together, reversine suppresses growth of OSCC via multiple mechanisms, which may be a unique advantage for developing novel therapeutic regimens for treatment of oral cancer in the future.
Collapse
|
43
|
Mai S, Muster B, Bereiter-Hahn J, Jendrach M. Autophagy proteins LC3B, ATG5 and ATG12 participate in quality control after mitochondrial damage and influence lifespan. Autophagy 2012; 8:47-62. [PMID: 22170153 DOI: 10.4161/auto.8.1.18174] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial health is maintained by the quality control mechanisms of mitochondrial dynamics (fission and fusion) and mitophagy. Decline of these processes is thought to contribute to aging and neurodegenerative diseases. To investigate the role of mitochondrial quality control in aging on the cellular level, human umbilical vein endothelial cells (HUVEC) were subjected to mitochondria-targeted damage by combining staining of mitochondria and irradiation. This treatment induced a short boost of reactive oxygen species, which resulted in transient fragmentation of mitochondria followed by mitophagy, while mitochondrial dynamics were impaired. Furthermore, targeted mitochondrial damage upregulated autophagy factors LC3B, ATG5 and ATG12. Consequently these proteins were overexpressed in HUVEC as an in vitro aging model, which significantly enhanced the replicative life span up to 150% and the number of population doublings up to 200%, whereas overexpression of LAMP-1 did not alter the life span. Overexpression of LC3B, ATG5 and ATG12 resulted in an improved mitochondrial membrane potential, enhanced ATP production and generated anti-apoptotic effects, while ROS levels remained unchanged and the amount of oxidized proteins increased. Taken together, these data relate LC3B, ATG5 and ATG12 to mitochondrial quality control after oxidative damage, and to cellular longevity.
Collapse
Affiliation(s)
- Sören Mai
- Kinematic Cell Research Group; Institute for Cell Biology and Neuroscience, Center of Excellence Frankfurt-Macromolecular Complexes, Goethe University, Frankfurt/Main, Germany
| | | | | | | |
Collapse
|
44
|
Zhang Y, Calderwood SK. Autophagy, protein aggregation and hyperthermia: a mini-review. Int J Hyperthermia 2011; 27:409-14. [PMID: 21756038 DOI: 10.3109/02656736.2011.552087] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE We aim to explore the role of macroautophagy in cellular responses to hyperthermia. Protein damage incurred during hyperthermia can either lead to cell death or may be repaired by polypeptide quality control pathways including: (1) the deterrence of protein unfolding by molecular chaperones and (2) proteolysis of the denatured proteins within the proteasome. A third pathway of protein quality control is triggered by formation of protein aggregates in the heat shocked cell. This is the macroautophagy pathway in which protein aggregates are transported to specialised organelles called autolysosomes capable of degrading the aggregates. The consequences for cell viability of triggering this pathway are complex and may involve cell death, although under many circumstances, including exposure of cells to hyperthermia, autophagy leads to enhanced cell survival. We have discussed mechanisms by which cells detect protein aggregates and recruit them into the macroautophagy pathway as well as the potential role of inhibiting this process in hyperthermia. CONCLUSIONS Directed macroautophagy, with its key role in protein quality control, seems an attractive target for a therapy such as hyperthermia that functions principally through denaturing the proteome. However, much work is needed to decode the mechanisms of thermal stress-mediated macroautophagy and their role in survival/death of cancer cells before recommendations can be made on targeting this pathway in combination with hyperthermia.
Collapse
Affiliation(s)
- Yue Zhang
- Molecular and Cellular Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School , 99 Brookline Avenue, Boston, MA 02215 , USA
| | | |
Collapse
|
45
|
Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science 2011; 333:1109-12. [PMID: 21868666 PMCID: PMC3405151 DOI: 10.1126/science.1201940] [Citation(s) in RCA: 881] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alterations of mitochondrial functions are linked to multiple degenerative or acute diseases. As mitochondria age in our cells, they become progressively inefficient and potentially toxic, and acute damage can trigger the permeabilization of mitochondrial membranes to initiate apoptosis or necrosis. Moreover, mitochondria have an important role in pro-inflammatory signaling. Autophagic turnover of cellular constituents, be it general or specific for mitochondria (mitophagy), eliminates dysfunctional or damaged mitochondria, thus counteracting degeneration, dampening inflammation, and preventing unwarranted cell loss. Decreased expression of genes that regulate autophagy or mitophagy can cause degenerative diseases in which deficient quality control results in inflammation and the death of cell populations. Thus, a combination of mitochondrial dysfunction and insufficient autophagy may contribute to multiple aging-associated pathologies.
Collapse
Affiliation(s)
- Douglas R. Green
- Department of Immunology, St Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Lorenzo Galluzzi
- INSERM, U848, F-94805 Villejuif, France
- Institut Gustave Roussy, F94805 Villejuif, France
- Université Paris-Sud, Paris 11, F-94805 Villejuif, France
| | - Guido Kroemer
- INSERM, U848, F-94805 Villejuif, France
- Metabolomics Platform, Institut Gustave Roussy, F-94805 Villejuif, France
- Centre de Recherche des Cordeliers, F-75005 Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, F-75908 Paris, France
- Université Paris Descartes, Paris 5, F-75270 Paris, France
| |
Collapse
|
46
|
Zhou F, Yang Y, Xing D. Bcl-2 and Bcl-xL play important roles in the crosstalk between autophagy and apoptosis. FEBS J 2010; 278:403-13. [PMID: 21182587 DOI: 10.1111/j.1742-4658.2010.07965.x] [Citation(s) in RCA: 315] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Autophagy and apoptosis play important roles in the development, cellular homeostasis and, especially, oncogenesis of mammals. They may be triggered by common upstream signals, resulting in combined autophagy and apoptosis. In other instances, they may be mutually exclusive. Recent studies have suggested possible molecular mechanisms for crosstalk between autophagy and apoptosis. Bcl-2 and Bcl-xL, the well-characterized apoptosis guards, appear to be important factors in autophagy, inhibiting Beclin 1-mediated autophagy by binding to Beclin 1. In addition, Beclin 1, Bcl-2 and Bcl-xL can cooperate with Atg5 or Ca(2+) to regulate both autophagy and apoptosis. Thus, Bcl-2 and Bcl-xL represent a molecular link between autophagy and apoptosis. Here, we discuss the possible roles of Bcl-2 and Bcl-xL in apoptosis and autophagy, and the crosstalk between them.
Collapse
Affiliation(s)
- Feifan Zhou
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | | | | |
Collapse
|
47
|
Abstract
Hyperthermia has been known to induce malformations in numerous animal models as well being associated with human abnormalities. This was apparent particularly when the hyperthermia exposure was during the early stages of neural development. Although it was recognized relatively early that these exposures induced cell death, the specific molecular mechanism of how a brief heat exposure was translated in to specific cellular functions remains largely unknown. While our understanding of the events that govern how cells react to heat, or stresses in general, has increased, there is much that remains undiscovered. In this brief review, animal and clinical observations are outlined as are some of the scientific explorations that were undertaken to characterize, define, and better understand the morphological, biochemical, and molecular effects of hyperthermia on the developing embryo.
Collapse
Affiliation(s)
- Gregory D Bennett
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska 68198-5805, USA.
| |
Collapse
|