1
|
Arif SM, Khan I, Saeed M, Chaudhari SK, Ghorbanpour M, Hasan M, Mustafa G. Exploring omics solutions to reduce micro/nanoplastic toxicity in plants: A comprehensive overview. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 974:179220. [PMID: 40147233 DOI: 10.1016/j.scitotenv.2025.179220] [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: 01/04/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
The proliferation of plastic waste, particularly in the form of microplastics (MPs) and nanoplastics (NPs), has emerged as a significant environmental challenge with profound implications for agricultural ecosystems. These pervasive pollutants accumulate in soil, altering its physicochemical properties and disrupting microbial communities. MPs/NPs can infiltrate plant systems, leading to oxidative stress and cytotoxic effects, which in turn compromise essential physiological functions such as water uptake, nutrient absorption, and photosynthesis. This situation threatens crop yield and health, while also posing risks to human health and food security through potential accumulation in the food chain. Despite increasing awareness of this issue, substantial gaps still remain in our understanding of the physiological and molecular mechanisms that govern plant responses to MP/NP stress. This review employs integrative omics techniques including genomics, transcriptomics, proteomics, metabolomics, and epigenomics to elucidate these responses. High-throughput methodologies have revealed significant genetic and metabolic alterations that enable plants to mitigate the toxicity associated with MPs/NPs. The findings indicate a reconfiguration of metabolic pathways aimed at maintaining cellular homeostasis, activation of antioxidant mechanisms, and modulation of gene expression related to stress responses. Additionally, epigenetic modifications suggest that plants adapt to prolonged plastics exposure, highlighting unexplored avenues for targeted research. By integrating various omics approaches, a comprehensive understanding of molecular interactions and their effects on plant systems can be achieved. This review underscores potential targets for biotechnological and agronomic interventions aimed at enhancing plant resilience by identifying key stress-responsive genes, proteins, and metabolites. Ultimately, this work addresses critical knowledge gaps and highlights the importance of multi-omics strategies in developing sustainable solutions to mitigate the adverse effects of MP/NP pollution in agriculture, thereby ensuring the integrity of food systems and ecosystems.
Collapse
Affiliation(s)
- Samia Muhammad Arif
- Department of Plant Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Ilham Khan
- Department of Plant Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Muhammad Saeed
- Environmental Biology and Ecotoxicology Laboratory, Department of Environmental Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Sunbal Khalil Chaudhari
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Sargodha Campus, Sargodha 42100, Pakistan
| | - Mansour Ghorbanpour
- Department of Medicinal Plants, Faculty of Agriculture and Natural Resources, Arak University, Arak 38156-8-8349, Iran.
| | - Murtaza Hasan
- Department of Biotechnology, Faculty of Chemical and Biological Sciences, The Islamia University of Bahawalpur, 63100, Pakistan.
| | - Ghazala Mustafa
- Department of Plant Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|
2
|
Ali A, Saeed S, Hussain R, Afzal G, Siddique AB, Parveen G, Hasan M, Caprioli G. Synthesis and Characterization of Silica, Silver-Silica, and Zinc Oxide-Silica Nanoparticles for Evaluation of Blood Biochemistry, Oxidative Stress, and Hepatotoxicity in Albino Rats. ACS OMEGA 2023; 8:20900-20911. [PMID: 37332821 PMCID: PMC10269246 DOI: 10.1021/acsomega.3c01674] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/10/2023] [Indexed: 06/20/2023]
Abstract
Evaluation of nanoparticles (NPs) for biomedical applications has received a lot of attention for detailed study on pharmacokinetics prior to clinical application. In this study, pure C-SiO2 (crystalline silica) NPs and SiO2 nanocomposites with silver (Ag) and zinc oxide (ZnO) were prepared by utilizing different synthesis routes such as sol-gel and co-precipitation techniques. The prepared NPs showed highly crystalline nature as confirmed by X-ray diffraction analysis where average crystallite sizes of 35, 16, and 57 nm for C-SiO2, Ag-SiO2, and ZnO-SiO2 NPs, respectively, were calculated. Fourier transform infrared analysis confirmed the presence of functional groups related to the chemicals and procedures used for sample preparation. Due to agglomeration of the prepared NPs, the scanning electron microscope images showed large particle sizes when compared to their crystalline sizes. The optical properties of the prepared NPs such as absorption were obtained with UV-Vis spectroscopy. For in vivo biological evaluation, albino rats, both male and female, kept in different groups were exposed to NPs with 500 μg/kg dose. Hematological, serum biochemistry, histo-architecture, oxidative stress biomarkers, and antioxidant parameters in liver tissues along with various biomarkers for the evaluation of erythrocytes were estimated. The results on hemato-biochemistry, histopathological ailments, and oxidative stress parameters exhibited 95% alteration in the liver and erythrocytes of C-SiO2 NPs-treated rats while 75 and 60% alteration in the liver tissues of rats due to exposure to Ag-SiO2 and ZnO-SiO2 NPs, respectively, when compared with the albino rats of the control (untreated) group. Therefore, the current study showed that the prepared NPs had adverse effects on the liver and erythrocytes causing hepatotoxicity in the albino rats in respective order C-SiO2 > Ag SiO2 > ZnO-SiO2. As the C-SiO2 NPs appeared to be the most toxic, it has been concluded that coating SiO2 on Ag and ZnO reduced their toxicological impact on albino rats. Consequently, it is suggested that Ag-SiO2 and ZnO-SiO2 NPs are more biocompatible than C-SiO2 NPs.
Collapse
Affiliation(s)
- Arooj Ali
- Institute
of Physics, Faculty of Physical & Mathematical Sciences, The Islamia University of Bahawalpur, Bahawalpur, Punjab 63100, Pakistan
| | - Saba Saeed
- Institute
of Physics, Faculty of Physical & Mathematical Sciences, The Islamia University of Bahawalpur, Bahawalpur, Punjab 63100, Pakistan
| | - Riaz Hussain
- Department
of Pathology, Faculty of Veterinary & Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Punjab 63100, Pakistan
| | - Gulnaz Afzal
- Department
of Zoology, Faculty of Chemical & Biological Sciences, The Islamia University of Bahawalpur, Bahawalpur, Punjab 63100, Pakistan
| | - Abu Baker Siddique
- Department
of Microbiology, Faculty of Life Sciences, Government College University, Faisalabad, Punjab 38000, Pakistan
| | - Gulnaz Parveen
- Department
of Botany, Faculty of Science, Women University
Swabi, Swabi, Khyber Pakhtunkhwa 23430, Pakistan
| | - Murtaza Hasan
- Department
of Biotechnology, Faculty of Chemical & Biological Sciences, The Islamia University of Bahawalpur, Bahawalpur, Punjab 63100, Pakistan
- College
of Chemistry and Chemical Engineering, Zhongkai
University of Agriculture and Engineering, Guangzhou 510225, China
| | - Giovanni Caprioli
- Chemistry
Interdisciplinary Project (CHip), School of Pharmacy, University of Camerino, Camerino 62032, Italy
| |
Collapse
|
3
|
Ma F, Li H, Huo H, Han Q, Liao J, Zhang H, Li Y, Pan J, Hu L, Guo J, Tang Z. N-acetyl-L-cysteine alleviates FUNDC1-mediated mitophagy by regulating mitochondrial dynamics in type 1 diabetic nephropathy canine. Life Sci 2023; 313:121278. [PMID: 36521547 DOI: 10.1016/j.lfs.2022.121278] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/29/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Diabetic nephropathy (DN) is a major complication of type 1 diabetes mellitus, and hyperglycemia and hypertension are the main risk factors for the development of DN. N-Acetyl-Cysteine (NAC) has a variety of effects, interfering with the production and scavenging of free radicals and regulating the metabolic activity of tissue cells. However, the efficacy of NAC on DN treatment is unclear. Thus, this study investigated the protective mechanism of NAC combined with insulin on renal injury in dogs with DN. The forty dogs were selected and divided into control group, DM group, INS group, INS + NAC group and NAC group to establish the model for a trial period of 4 months. The results revealed that INS + NAC was effective in reducing and stabilizing blood glucose levels. Biochemical results showed that INS + NAC treatment significantly regulated the stability of UREA, CREA and fructosamine indicators. Meanwhile, histopathology staining showed significant glomerular wrinkling and fibrosis in the DM group, which could be reversed after INS + NAC treatment. In addition, INS + NAC could restore mitochondria homeostasis by upregulating the levels of mitochondrial fission (MFN1, MFN2 and OPA1) and inhibiting of mitochondrial fusion (DRP1, FIS1 and MFF) related indicators. Further studies revealed that INS + NAC regulated the expression levels of renal BNIP3, NIX and FUNDC1 in the DM group, thereby alleviating mitophagy. Collectively, these results suggested that NAC combined with insulin protects DN by regulating the mitochondrial dynamics and FUNDC1-mediated mitophagy.
Collapse
Affiliation(s)
- Feiyang Ma
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Huayu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Haihua Huo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Qingyue Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Jiaqiang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Jianying Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| |
Collapse
|
4
|
Will Cannabigerol Trigger Neuroregeneration after a Spinal Cord Injury? An In Vitro Answer from NSC-34 Scratch-Injured Cells Transcriptome. Pharmaceuticals (Basel) 2022; 15:ph15020117. [PMID: 35215230 PMCID: PMC8875351 DOI: 10.3390/ph15020117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury affects the lives of millions of people around the world, often causing disability and, in unfortunate circumstances, death. Rehabilitation can partly improve outcomes and only a small percentage of patients, typically the least injured, can hope to return to normal living conditions. Cannabis sativa is gaining more and more interest in recent years, even though its beneficial properties have been known for thousands of years. Cannabigerol (CBG), extracted from C. sativa, is defined as the “mother of all cannabinoids” and its properties range from anti-inflammatory to antioxidant and neuroprotection. Using NSC-34 cells to model spinal cord injury in vitro, our work evaluated the properties of CBG treatments in motor neuron regeneration. While pre-treatment can modulate oxidative stress and increase antioxidant enzyme genes, such as Tnx1, decreasing Nos1 post-treatment seems to induce regeneration genes by triggering different pathways, such as Gap43 via p53 acetylation by Ep300 and Ddit3 and Xbp1 via Bdnf signaling, along with cytoskeletal remodeling signaling genes Nrp1 and Map1b. Our results indicate CBG as a phytocompound worth further investigation in the field of neuronal regeneration.
Collapse
|
5
|
Prados ME, Correa-Sáez A, Unciti-Broceta JD, Garrido-Rodríguez M, Jimenez-Jimenez C, Mazzone M, Minassi A, Appendino G, Calzado MA, Muñoz E. Betulinic Acid Hydroxamate is Neuroprotective and Induces Protein Phosphatase 2A-Dependent HIF-1α Stabilization and Post-transcriptional Dephosphorylation of Prolyl Hydrolase 2. Neurotherapeutics 2021; 18:1849-1861. [PMID: 34339019 PMCID: PMC8608974 DOI: 10.1007/s13311-021-01089-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 02/04/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder characterized by unwanted choreatic movements, behavioral and psychiatric disturbances, and dementia. The activation of the hypoxic response pathway through the pharmacological inhibition of hypoxia-inducing factor (HIF) prolyl-hydroxylases (PHDs) is a promising approach for neurodegenerative diseases, including HD. Herein, we have studied the mechanism of action of the compound Betulinic acid hydroxamate (BAH), a hypoximimetic derivative of betulinic acid, and its efficacy against striatal neurodegeneration using complementary approaches. Firstly, we showed the molecular mechanisms through which BAH modifies the activity of the PHD2 prolyl hydroxylase, thus directly affecting HIF-1α stability. BAH treatment reduces PHD2 phosphorylation on Ser-125 residue, responsible for the control of its hydrolase activity. HIF activation by BAH is inhibited by okadaic acid and LB-100 indicating that a protein phosphatase 2A (PP2A) is implicated in the mechanism of action of BAH. Furthermore, in striatal cells bearing a mutated form of the huntingtin protein, BAH stabilized HIF-1α protein, induced Vegf and Bnip3 gene expression and protected against mitochondrial toxin-induced cytotoxicity. Pharmacokinetic analyses showed that BAH has a good brain penetrability and experiments performed in a mouse model of striatal neurodegeneration induced by 3-nitropropionic acid showed that BAH improved the clinical symptoms. In addition, BAH also prevented neuronal loss, decreased reactive astrogliosis and microglial activation, inhibited the upregulation of proinflammatory markers, and improved antioxidant defenses in the brain. Taken together, our results show BAH's ability to activate the PP2A/PHD2/HIF pathway, which may have important implications in the treatment of HD and perhaps other neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Alejandro Correa-Sáez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain
- Department of Cellular Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Hospital Reina Sofia, Cordoba, Spain
| | | | - Martín Garrido-Rodríguez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain
- Department of Cellular Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Hospital Reina Sofia, Cordoba, Spain
| | - Carla Jimenez-Jimenez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain
- Department of Cellular Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Hospital Reina Sofia, Cordoba, Spain
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB-KULeuven, 3000, Leuven, Belgium
| | - Alberto Minassi
- Department of Drug Science, University of Piemonte Orientale, Novara, Italy
| | - Giovanni Appendino
- Department of Drug Science, University of Piemonte Orientale, Novara, Italy
| | - Marco A Calzado
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain.
- Department of Cellular Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.
- Hospital Universitario Hospital Reina Sofia, Cordoba, Spain.
| | - Eduardo Muñoz
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain.
- Department of Cellular Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.
- Hospital Universitario Hospital Reina Sofia, Cordoba, Spain.
| |
Collapse
|
6
|
Wang DX, Yang Y, Huang XS, Tang JY, Zhang X, Huang HX, Zhou B, Liu B, Xiao HQ, Li XH, Yang P, Zou SC, Liu K, Wang XY, Li XS. Pramipexole attenuates neuronal injury in Parkinson's disease by targeting miR-96 to activate BNIP3-mediated mitophagy. Neurochem Int 2021; 146:104972. [PMID: 33493581 DOI: 10.1016/j.neuint.2021.104972] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Parkinson's disease is a common neurodegenerative problem. Pramipexole (PPX) plays protective role in Parkinson's disease. Nevertheless, the mechanism of PPX in Parkinson's disease-like neuronal injury is largely uncertain. METHODS 1-methyl-4-phenylpyridinium (MPP+)-stimulated neuronal cells and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mice were used as the model of Parkinson's disease. MPP+-induced neuronal injury was assessed via cell viability, lactic dehydrogenase (LDH) release and apoptosis. microRNA-96 (miR-96) and BCL2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3) abundances were examined by quantitative reverse transcription polymerase chain reaction (qRT-PCR) or Western blotting. Mitophagy was tested by Western blotting and immunofluorescence staining. MPTP-induced neuronal injury in mice was investigated via behavioral tests and TUNEL. RESULTS PPX alleviated MPP+-induced neuronal injury via increasing cell viability and decreasing LDH release and apoptosis. PPX reversed MPP+-induced miR-96 expression and inhibition of mitophagy. miR-96 overexpression or BNIP3 interference weakened the suppressive role of PPX in MPP+-induced neuronal injury. miR-96 targeted BNIP3 to inhibit PTEN-induced putative kinase 1 (PINK1)/Parkin signals-mediated mitophagy. miR-96 overexpression promoted MPP+-induced neuronal injury via decreasing BNIP3. PPX weakened MPTP-induced neuronal injury in mice via regulating miR-96/BNIP3-mediated mitophagy. CONCLUSION PPX mitigated neuronal injury in MPP+-induced cells and MPTP-induced mice by activating BNIP3-mediated mitophagy via directly decreasing miR-96.
Collapse
Affiliation(s)
- Dong-Xin Wang
- The Research Institute of Mentality and Sanitation, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Ying Yang
- Medical Department of Neurology, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Xiao-Song Huang
- Medical Department of Neurology, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Jia-Yu Tang
- Medical Department of Neurology, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Xi Zhang
- Clinical Medical School, Hunan Traditional Chinese Medicine University, Changsha, 410006, Hunan Province, PR China
| | - Hong-Xing Huang
- Department of Neurosurgery, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Bin Zhou
- Department of Neurosurgery, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Bo Liu
- Department of Neurosurgery, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Hui-Qiong Xiao
- Department of Scientific Research, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Xiao-Hui Li
- Medical Department of Neurology, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Ping Yang
- Department of Psychology, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Shu-Cheng Zou
- Department of Neurosurgery, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Kun Liu
- Department of Neurosurgery, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Xiao-Ye Wang
- The Institution of Clinical Trials on Drugs, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China.
| | - Xiao-Song Li
- The Research Institute of Mentality and Sanitation, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China.
| |
Collapse
|
7
|
Wang C, He C, Lu S, Wang X, Wang L, Liang S, Wang X, Piao M, Cui J, Chi G, Ge P. Autophagy activated by silibinin contributes to glioma cell death via induction of oxidative stress-mediated BNIP3-dependent nuclear translocation of AIF. Cell Death Dis 2020; 11:630. [PMID: 32801360 PMCID: PMC7429844 DOI: 10.1038/s41419-020-02866-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023]
Abstract
Induction of lethal autophagy has become a strategy to eliminate glioma cells, but it remains elusive whether autophagy contributes to cell death via causing mitochondria damage and nuclear translocation of apoptosis inducing factor (AIF). In this study, we find that silibinin induces AIF translocation from mitochondria to nuclei in glioma cells in vitro and in vivo, which is accompanied with autophagy activation. In vitro studies reveal that blocking autophagy with 3MA, bafilomycin A1 or by knocking down ATG5 with SiRNA inhibits silibinin-induced mitochondrial accumulation of superoxide, AIF translocation from mitochondria to nuclei and glioma cell death. Mechanistically, silibinin activates autophagy through depleting ATP by suppressing glycolysis. Then, autophagy improves intracellular H2O2 via promoting p53-mediated depletion of GSH and cysteine and downregulation of xCT. The increased H2O2 promotes silibinin-induced BNIP3 upregulation and translocation to mitochondria. Knockdown of BNIP3 with SiRNA inhibits silibinin-induced mitochondrial depolarization, accumulation of mitochondrial superoxide, and AIF translocation from mitochondria to nuclei, as well as prevents glioma cell death. Furthermore, we find that the improved H2O2 reinforces silibinin-induced glycolysis dysfunction. Collectively, autophagy contributes to silibinin-induced glioma cell death via promotion of oxidative stress-mediated BNIP3-dependent nuclear translocation of AIF.
Collapse
Affiliation(s)
- Chongcheng Wang
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Chuan He
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Shan Lu
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Xuanzhong Wang
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Lei Wang
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Shipeng Liang
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China
| | - Xinyu Wang
- Department of Radiotherapy, Second Hospital of Jilin University, 130021, Changchun, China
| | - Meihua Piao
- Department of Anesthesiology, First Hospital of Jilin University, 130021, Changchun, China
| | - Jiayue Cui
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, 130021, Changchun, China
| | - Guangfan Chi
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 130021, Changchun, China
| | - Pengfei Ge
- Department of Neurosurgery, First Hospital of Jilin University, 130021, Changchun, China.
- Research Center of Neuroscience, First Hospital of Jilin University, 130021, Changchun, China.
| |
Collapse
|
8
|
Overbey EG, da Silveira WA, Stanbouly S, Nishiyama NC, Roque-Torres GD, Pecaut MJ, Zawieja DC, Wang C, Willey JS, Delp MD, Hardiman G, Mao XW. Spaceflight influences gene expression, photoreceptor integrity, and oxidative stress-related damage in the murine retina. Sci Rep 2019; 9:13304. [PMID: 31527661 PMCID: PMC6746706 DOI: 10.1038/s41598-019-49453-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/19/2019] [Indexed: 11/08/2022] Open
Abstract
Extended spaceflight has been shown to adversely affect astronaut visual acuity. The purpose of this study was to determine whether spaceflight alters gene expression profiles and induces oxidative damage in the retina. Ten week old adult C57BL/6 male mice were flown aboard the ISS for 35 days and returned to Earth alive. Ground control mice were maintained on Earth under identical environmental conditions. Within 38 (+/-4) hours after splashdown, mice ocular tissues were collected for analysis. RNA sequencing detected 600 differentially expressed genes (DEGs) in murine spaceflight retinas, which were enriched for genes related to visual perception, the phototransduction pathway, and numerous retina and photoreceptor phenotype categories. Twelve DEGs were associated with retinitis pigmentosa, characterized by dystrophy of the photoreceptor layer rods and cones. Differentially expressed transcription factors indicated changes in chromatin structure, offering clues to the observed phenotypic changes. Immunofluorescence assays showed degradation of cone photoreceptors and increased retinal oxidative stress. Total retinal, retinal pigment epithelium, and choroid layer thickness were significantly lower after spaceflight. These results indicate that retinal performance may decrease over extended periods of spaceflight and cause visual impairment.
Collapse
Affiliation(s)
- Eliah G Overbey
- University of Washington, Department of Genome Sciences, Seattle, WA, USA.
| | - Willian Abraham da Silveira
- Queen's University Belfast, Faculty of Medicine, Health and Life Sciences, School of Biological Sciences, Institute for Global Food Security (IGFS), 19 Chlorine Gardens, Belfast, Northern Ireland, BT9 5DL, UK
| | - Seta Stanbouly
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, 92350, USA
- Center for Genomics, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Nina C Nishiyama
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, 92350, USA
| | | | - Michael J Pecaut
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, 92350, USA
| | - David Carl Zawieja
- Department of Medical Physiology, Texas A&M University, College Station, Texas, USA
| | - Charles Wang
- Center for Genomics, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Jeffrey S Willey
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Michael D Delp
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, 32306, USA
| | - Gary Hardiman
- Queen's University Belfast, Faculty of Medicine, Health and Life Sciences, School of Biological Sciences, Institute for Global Food Security (IGFS), 19 Chlorine Gardens, Belfast, Northern Ireland, BT9 5DL, UK
| | - Xiao Wen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, 92350, USA
| |
Collapse
|
9
|
Xu WL, Wang SH, Sun WB, Gao J, Ding XM, Kong J, Xu L, Ke S. Insufficient radiofrequency ablation-induced autophagy contributes to the rapid progression of residual hepatocellular carcinoma through the HIF-1α/BNIP3 signaling pathway. BMB Rep 2019. [PMID: 30940322 PMCID: PMC6507849 DOI: 10.5483/bmbrep.2019.52.4.263] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Currently speaking, it is noted that radiofrequency ablation (RFA) has been the most widely used treatment for hepatocellular carcinoma (HCC) occurring in patients. However, accumulating evidence has demonstrated that the incidence of insufficient RFA (IRFA) may result in the identified rapid progression of residual HCC in the patient, which can greatly hinder the effectiveness and patient reported benefits of utilizing this technique. Although many efforts have been proposed, the underlying mechanisms triggering the rapid progression of residual HCC after IRFA have not yet been fully clarified through current research literature reviews. It was shown in this study that cell proliferation, migration and invasion of residual HepG2 and SMMC7721 cells were significantly increased after the IRFA was simulated in vitro. In other words, it is noted that IRFA could do this by enhancing the image of autophagy of the residual HCC cell via the HIF-1α/BNIP3 pathway. Consequently, the down-regulation of BNIP3 may result in the inhibition of the residual HCC cell progression and autophagy after IRFA. Our present study results suggest that IRFA could promote residual HCC cell progression in vitro by enhancing autophagy via the HIF-1α/BNIP3 pathway. For this reason, it is noted that the targeting of the BNIP3 may be useful in preventing the rapid growth and metastasis of residual HCC after IRFA.
Collapse
Affiliation(s)
- Wen-Lei Xu
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Shao-Hong Wang
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Wen-Bing Sun
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Jun Gao
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Xue-Mei Ding
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Jian Kong
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Li Xu
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Shan Ke
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| |
Collapse
|
10
|
Yashiro M, Ohya M, Mima T, Ueda Y, Nakashima Y, Kawakami K, Ishizawa Y, Yamamoto S, Kobayashi S, Yano T, Tanaka Y, Okuda K, Sonou T, Shoshihara T, Iwashita Y, Iwashita Y, Tatsuta K, Matoba R, Negi S, Shigematsu T. FGF23 modulates the effects of erythropoietin on gene expression in renal epithelial cells. Int J Nephrol Renovasc Dis 2018; 11:125-136. [PMID: 29670389 PMCID: PMC5894721 DOI: 10.2147/ijnrd.s158422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background FGF23 plays an important role in calcium–phosphorus metabolism. Other roles of FGF23 have recently been reported, such as commitment to myocardium enlargement and immunological roles in the spleen. In this study, we aimed to identify the roles of FGF23 in the kidneys other than calcium–phosphorus metabolism. Methods DNA microarrays and bioinformatics tools were used to analyze gene expression in mIMCD3 mouse renal tubule cells following treatment with FGF23, erythropoietin and/or an inhibitor of ERK. Results Three protein-coding genes were upregulated and 12 were downregulated in response to FGF23. Following bioinformatics analysis of these genes, PPARγ and STAT3 were identified as candidate transcript factors for mediating their upregulation, and STAT1 as a candidate for mediating their downregulation. Because STAT1 and STAT3 also mediate erythropoietin signaling, we investigated whether FGF23 and erythropoietin might show interactive effects in these cells. Of the 15 genes regulated by FGF23, 11 were upregulated by erythropoietin; 10 of these were downregulated following cotreatment with FGF23. Inhibition of ERK, an intracellular mediator of FGF23, reversed the effects of FGF23. However, FGF23 did not influence STAT1 phosphorylation, suggesting that it impinges on erythropoietin signaling through other mechanisms. Conclusion Our results suggest cross talk between erythropoietin and FGF23 signaling in the regulation of renal epithelial cells.
Collapse
Affiliation(s)
- Mitsuru Yashiro
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Masaki Ohya
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Toru Mima
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Yumi Ueda
- DNA Chip Research Inc., Minato, Japan
| | - Yuri Nakashima
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Kazuki Kawakami
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | | | - Shuto Yamamoto
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Sou Kobayashi
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Takurou Yano
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Yusuke Tanaka
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Kouji Okuda
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Tomohiro Sonou
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | | | - Yuko Iwashita
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Yu Iwashita
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Kouichi Tatsuta
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | | | - Shigeo Negi
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | | |
Collapse
|
11
|
Moulis MF, Millet AM, Daloyau M, Miquel MC, Ronsin B, Wissinger B, Arnauné-Pelloquin L, Belenguer P. OPA1 haploinsufficiency induces a BNIP3-dependent decrease in mitophagy in neurons: relevance to Dominant Optic Atrophy. J Neurochem 2016; 140:485-494. [DOI: 10.1111/jnc.13894] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/28/2016] [Accepted: 10/24/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Manon F Moulis
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Aurélie M Millet
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Marlène Daloyau
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Marie-Christine Miquel
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Brice Ronsin
- Center of Developmental Biology (CBD); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Bernd Wissinger
- Center for Ophthalmology; University of Tübingen; Tübingen Germany
| | - Laetitia Arnauné-Pelloquin
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Pascale Belenguer
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| |
Collapse
|
12
|
Chen JL, David J, Cook-Spaeth D, Casey S, Cohen D, Selvendiran K, Bekaii-Saab T, Hays JL. Autophagy Induction Results in Enhanced Anoikis Resistance in Models of Peritoneal Disease. Mol Cancer Res 2016; 15:26-34. [PMID: 27807188 DOI: 10.1158/1541-7786.mcr-16-0200-t] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/13/2016] [Accepted: 10/06/2016] [Indexed: 12/27/2022]
Abstract
Peritoneal carcinomatosis and peritoneal sarcomatosis is a potential complication of nearly all solid tumors and results in profoundly increased morbidity and mortality. Despite the ubiquity of peritoneal carcinomatosis/peritoneal sarcomatosis, there are no clinically relevant targeted therapies for either its treatment or prevention. To identify potential therapies, we developed in vitro models of peritoneal carcinomatosis/peritoneal sarcomatosis using tumor cell lines and patient-derived spheroids (PDS) that recapitulate anoikis resistance and spheroid proliferation across multiple cancer types. Epithelial- and mesenchymal-derived cancer cell lines (YOU, PANC1, HEYA8, CHLA10, and TC71) were used to generate spheroids and establish growth characteristics. Differential gene expression analyses of these spheroids to matched adherent cells revealed a consensus spheroid signature. This spheroid signature discriminates primary tumor specimens from tumor cells found in ascites of ovarian cancer patients and in our PDS models. Key in this gene expression signature is BNIP3 and BNIP3L, known regulators of autophagy and apoptosis. Elevated BNIP3 mRNA expression is associated with poor survival in ovarian cancer patients and elevated BNIP3 protein, as measured by IHC, and is also associated with higher grade tumors and shorter survival. Pharmacologic induction of autophagy with rapamycin significantly increased spheroid formation and survival while decreasing the induction of apoptosis. In contrast, the autophagy inhibitor hydroxychloroquine abrogated spheroid formation with a clear increase in apoptosis. Modulation of BNIP3 and the critical autophagy gene Beclin-1 (BECN1) also caused a significant decrease in spheroid formation. Combined, these data demonstrate how modulation of BNIP3-related autophagy, in PDS and in vitro spheroid models, alters the survival and morphology of spheroids. IMPLICATIONS Development of BNIP3/BNIP3L-targeting agents or autophagy-targeting agents may reduce morbidity and mortality associated with peritoneal carcinomatosis and sarcomatosis. Mol Cancer Res; 15(1); 26-34. ©2016 AACR.
Collapse
Affiliation(s)
- James L Chen
- Department of Biomedical Informatics, Division of Bioinformatics, The Ohio State University, Columbus, Ohio.,Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, Ohio
| | - Jason David
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, Ohio
| | - Douglas Cook-Spaeth
- Department of Biomedical Informatics, Division of Bioinformatics, The Ohio State University, Columbus, Ohio
| | - Sydney Casey
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, Ohio
| | - David Cohen
- Department of Pathology, Anatomic Pathology Branch, The Ohio State University, Columbus, Ohio
| | - Karuppaiyah Selvendiran
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, The Ohio State University, Columbus, Ohio
| | - Tanios Bekaii-Saab
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, Ohio
| | - John L Hays
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, Ohio. .,Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
13
|
Zhang T, Xue L, Li L, Tang C, Wan Z, Wang R, Tan J, Tan Y, Han H, Tian R, Billiar TR, Tao WA, Zhang Z. BNIP3 Protein Suppresses PINK1 Kinase Proteolytic Cleavage to Promote Mitophagy. J Biol Chem 2016; 291:21616-21629. [PMID: 27528605 PMCID: PMC5076832 DOI: 10.1074/jbc.m116.733410] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/26/2016] [Indexed: 01/11/2023] Open
Abstract
Mutations in PINK1 (PTEN-induced putative kinase 1) cause early onset familial Parkinson's disease (PD). PINK1 accumulates on the outer membrane of damaged mitochondria followed by recruiting parkin to promote mitophagy. Here, we demonstrate that BCL2/adenovirus E1B 19-kDa interacting protein 3 (BNIP3), a mitochondrial BH3-only protein, interacts with PINK1 to promote the accumulation of full-length PINK1 on the outer membrane of mitochondria, which facilitates parkin recruitment and PINK1/parkin-mediated mitophagy. Inactivation of BNIP3 in mammalian cells promotes PINK1 proteolytic processing and suppresses PINK1/parkin-mediated mitophagy. Hypoxia-induced BNIP3 expression results in increased expression of full-length PINK1 and mitophagy. Consistently, expression of BNIP3 in Drosophila suppresses muscle degeneration and the mitochondrial abnormality caused by PINK1 inactivation. Together, the results suggest that BNIP3 plays a vital role in regulating PINK1 mitochondrial outer membrane localization, the proteolytic process of PINK1 and PINK1/parkin-mediated mitophagy under physiological conditions. Functional up-regulation of BNIP3 may represent a novel therapeutic strategy to suppress the progression of PD.
Collapse
Affiliation(s)
- Tongmei Zhang
- From the Institute of Precision Medicine, the Xiangya Hospital and State Key Laboratory of Medical Genetics, the Xiangya Medical School, Central South University, Changsha, Hunan 410078, China
| | - Liang Xue
- the Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907
| | - Li Li
- From the Institute of Precision Medicine, the Xiangya Hospital and State Key Laboratory of Medical Genetics, the Xiangya Medical School, Central South University, Changsha, Hunan 410078, China
| | - Chengyuan Tang
- From the Institute of Precision Medicine, the Xiangya Hospital and State Key Laboratory of Medical Genetics, the Xiangya Medical School, Central South University, Changsha, Hunan 410078, China
| | - Zhengqing Wan
- From the Institute of Precision Medicine, the Xiangya Hospital and State Key Laboratory of Medical Genetics, the Xiangya Medical School, Central South University, Changsha, Hunan 410078, China
| | - Ruoxi Wang
- From the Institute of Precision Medicine, the Xiangya Hospital and State Key Laboratory of Medical Genetics, the Xiangya Medical School, Central South University, Changsha, Hunan 410078, China
| | - Jieqiong Tan
- From the Institute of Precision Medicine, the Xiangya Hospital and State Key Laboratory of Medical Genetics, the Xiangya Medical School, Central South University, Changsha, Hunan 410078, China
| | - Ya Tan
- From the Institute of Precision Medicine, the Xiangya Hospital and State Key Laboratory of Medical Genetics, the Xiangya Medical School, Central South University, Changsha, Hunan 410078, China
| | - Hailong Han
- From the Institute of Precision Medicine, the Xiangya Hospital and State Key Laboratory of Medical Genetics, the Xiangya Medical School, Central South University, Changsha, Hunan 410078, China
| | - Runyi Tian
- From the Institute of Precision Medicine, the Xiangya Hospital and State Key Laboratory of Medical Genetics, the Xiangya Medical School, Central South University, Changsha, Hunan 410078, China
| | - Timothy R Billiar
- From the Institute of Precision Medicine, the Xiangya Hospital and State Key Laboratory of Medical Genetics, the Xiangya Medical School, Central South University, Changsha, Hunan 410078, China
- the Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, and
| | - W Andy Tao
- the Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907
| | - Zhuohua Zhang
- From the Institute of Precision Medicine, the Xiangya Hospital and State Key Laboratory of Medical Genetics, the Xiangya Medical School, Central South University, Changsha, Hunan 410078, China
- the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|