1
|
Xie J, Zhang Z. Recent Advances and Therapeutic Implications of 2-Oxoglutarate-Dependent Dioxygenases in Ischemic Stroke. Mol Neurobiol 2024; 61:3949-3975. [PMID: 38041714 DOI: 10.1007/s12035-023-03790-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/08/2023] [Indexed: 12/03/2023]
Abstract
Ischemic stroke is a common disease with a high disability rate and mortality, which brings heavy pressure on families and medical insurance. Nowadays, the golden treatments for ischemic stroke in the acute phase mainly include endovascular therapy and intravenous thrombolysis. Some drugs are used to alleviate brain injury in patients with ischemic stroke, such as edaravone and 3-n-butylphthalide. However, no effective neuroprotective drug for ischemic stroke has been acknowledged. 2-Oxoglutarate-dependent dioxygenases (2OGDDs) are conserved and common dioxygenases whose activities depend on O2, Fe2+, and 2OG. Most 2OGDDs are expressed in the brain and are essential for the development and functions of the brain. Therefore, 2OGDDs likely play essential roles in ischemic brain injury. In this review, we briefly elucidate the functions of most 2OGDDs, particularly the effects of regulations of 2OGDDs on various cells in different phases after ischemic stroke. It would also provide promising potential therapeutic targets and directions of drug development for protecting the brain against ischemic injury and improving outcomes of ischemic stroke.
Collapse
Affiliation(s)
- Jian Xie
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
2
|
Yi XF, Gao RL, Sun L, Wu ZX, Zhang SL, Huang LT, Han CB, Ma JT. Dual antitumor immunomodulatory effects of PARP inhibitor on the tumor microenvironment: A counterbalance between anti-tumor and pro-tumor. Biomed Pharmacother 2023; 163:114770. [PMID: 37105074 DOI: 10.1016/j.biopha.2023.114770] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/10/2023] [Accepted: 04/22/2023] [Indexed: 04/29/2023] Open
Abstract
Poly (ADP-ribose)-polymerases (PARPs) play an essential role in the maintenance of genome integrity, DNA repair, and apoptosis. PARP inhibitors (PARPi) exert antitumor effects via synthetic lethality and PARP trapping. PARPi impact the antitumor immune response by modulating the tumor microenvironment, and their effect has dual properties of promoting and inhibiting the antitumor immune response. PARPi promote M1 macrophage polarization, antigen presentation by dendritic cells, infiltration of B and T cells and their killing capacity and inhibit tumor angiogenesis. PARPi can also inhibit the activation and function of immune cells by upregulating PD-L1. In this review, we summarize the dual immunomodulatory effects and possible underlying mechanisms of PARPi, providing a basis for the design of combination regimens for clinical treatment and the identification of populations who may benefit from these therapies.
Collapse
Affiliation(s)
- Xiao-Fang Yi
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ruo-Lin Gao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Li Sun
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhi-Xuan Wu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shu-Ling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Le-Tian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cheng-Bo Han
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Jie-Tao Ma
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Derby SJ, Chalmers AJ, Carruthers RD. Radiotherapy-Poly(ADP-ribose) Polymerase Inhibitor Combinations: Progress to Date. Semin Radiat Oncol 2022; 32:15-28. [PMID: 34861992 DOI: 10.1016/j.semradonc.2021.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Radiation resistance remains a huge clinical problem for cancer patients and oncologists in the 21st century. In recent years, the mammalian DNA damage response (DDR) has been extensively characterized and shown to play a key role in determining cellular survival following ionizing radiation exposure. Genomic instability due to altered DDR is a hallmark of cancer, with many tumors exhibiting abnormal DNA repair or lack of redundancy in DDR. Targeting the abnormal DDR phenotype of tumor cells could lead to substantial gains in radiotherapy efficacy, improving local control and survival for patients with cancers that are refractory to current therapies. Poly(ADP-ribose) polymerase inhibitors (PARPi) are the most clinically advanced DDR inhibitors under investigation as radiosensitisers. Preclinical evidence suggests that PARPi may provide tumor specific radiosensitisation in certain contexts. In addition to inhibition of DNA single strand break repair, PARPi may offer other benefits in combination treatment including radiosensitisation of hypoxic cells and targeting of alternative repair pathways such as microhomology mediated end joining which are increasingly recognized to be upregulated in cancer. Several early phase clinical trials of PARPi with radiation have completed or are in progress. Early reports have highlighted tumor specific challenges, with tolerability dependent upon anatomical location and use of concomitant systemic therapies; these challenges were largely predicted by preclinical data. This review discusses the role of PARP in the cellular response to ionizing radiation, summarizes preclinical studies of PARPi in combination with radiotherapy and explores current early phase clinical trials that are evaluating these combinations.
Collapse
Affiliation(s)
- Sarah J Derby
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland.
| | - Anthony J Chalmers
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland
| | - Ross D Carruthers
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland
| |
Collapse
|
4
|
Liu S, Luo W, Wang Y. Emerging role of PARP-1 and PARthanatos in ischemic stroke. J Neurochem 2021; 160:74-87. [PMID: 34241907 DOI: 10.1111/jnc.15464] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/10/2021] [Accepted: 07/06/2021] [Indexed: 01/01/2023]
Abstract
Cell death is a key feature of neurological diseases, including stroke and neurodegenerative disorders. Studies in a variety of ischemic/hypoxic mouse models demonstrate that poly(ADP-ribose) polymerase 1 (PARP-1)-dependent cell death, also named PARthanatos, plays a pivotal role in ischemic neuronal cell death and disease progress. PARthanatos has its unique triggers, processors, and executors that convey a highly orchestrated and programmed signaling cascade. In addition to its role in gene transcription, DNA damage repair, and energy homeostasis through PARylation of its various targets, PARP-1 activation in neuron and glia attributes to brain damage following ischemia/reperfusion. Pharmacological inhibition or genetic deletion of PARP-1 reduces infarct volume, eliminates inflammation, and improves recovery of neurological functions in stroke. Here, we reviewed the role of PARP-1 and PARthanatos in stroke and their therapeutic potential.
Collapse
Affiliation(s)
- Shuiqiao Liu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Weibo Luo
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yingfei Wang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
5
|
Demény MA, Virág L. The PARP Enzyme Family and the Hallmarks of Cancer Part 2: Hallmarks Related to Cancer Host Interactions. Cancers (Basel) 2021; 13:2057. [PMID: 33923319 PMCID: PMC8123211 DOI: 10.3390/cancers13092057] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/07/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Poly (ADP-ribose) polymerases (PARPs) modify target proteins with a single ADP-ribose unit or with a poly (ADP-ribose) (PAR) polymer. PARP inhibitors (PARPis) recently became clinically available for the treatment of BRCA1/2 deficient tumors via the synthetic lethality paradigm. This personalized treatment primarily targets DNA damage-responsive PARPs (PARP1-3). However, the biological roles of PARP family member enzymes are broad; therefore, the effects of PARPis should be viewed in a much wider context, which includes complex effects on all known hallmarks of cancer. In the companion paper (part 1) to this review, we presented the fundamental roles of PARPs in intrinsic cancer cell hallmarks, such as uncontrolled proliferation, evasion of growth suppressors, cell death resistance, genome instability, replicative immortality, and reprogrammed metabolism. In the second part of this review, we present evidence linking PARPs to cancer-associated inflammation, anti-cancer immune response, invasion, and metastasis. A comprehensive overview of the roles of PARPs can facilitate the identification of novel cancer treatment opportunities and barriers limiting the efficacy of PARPi compounds.
Collapse
Affiliation(s)
- Máté A. Demény
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, 4032 Debrecen, Hungary
| | - László Virág
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, 4032 Debrecen, Hungary
| |
Collapse
|
6
|
Barca C, Wiesmann M, Calahorra J, Wachsmuth L, Döring C, Foray C, Heiradi A, Hermann S, Peinado MÁ, Siles E, Faber C, Schäfers M, Kiliaan AJ, Jacobs AH, Zinnhardt B. Impact of hydroxytyrosol on stroke: tracking therapy response on neuroinflammation and cerebrovascular parameters using PET-MR imaging and on functional outcomes. Theranostics 2021; 11:4030-4049. [PMID: 33754046 PMCID: PMC7977466 DOI: 10.7150/thno.48110] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
Immune cells have been implicated in influencing stroke outcomes depending on their temporal dynamics, number, and spatial distribution after ischemia. Depending on their activation status, immune cells can have detrimental and beneficial properties on tissue outcome after stroke, highlighting the need to modulate inflammation towards beneficial and restorative immune responses. Novel dietary therapies may promote modulation of pro- and anti-inflammatory immune cell functions. Among the dietary interventions inspired by the Mediterranean diet, hydroxytyrosol (HT), the main phenolic component of the extra virgin olive oil (EVOO), has been suggested to have antioxidant and anti-inflammatory properties in vitro. However, immunomodulatory effects of HT have not yet been studied in vivo after stroke. The aim of this project is therefore to monitor the therapeutic effect of a HT-enriched diet in an experimental stroke model using non-invasive in vivo multimodal imaging, behavioural phenotyping and cross-correlation with ex vivo parameters. Methods: A total of N = 22 male C57BL/6 mice were fed with either a standard chow (n = 11) or a HT enriched diet (n = 11) for 35 days, following a 30 min transient middle cerebral artery occlusion (tMCAo). T2-weighted (lesion) and perfusion (cerebral blood flow)-/diffusion (cellular density)-weighted MR images were acquired at days 1, 3, 7, 14, 21 and 30 post ischemia. [18F]DPA-714 (TSPO, neuroinflammation marker) PET-CT scans were acquired at days 7, 14, 21 and 30 post ischemia. Infarct volume (mm3), cerebral blood flow (mL/100g/min), apparent diffusion coefficient (10-4·mm2/s) and percentage of injected tracer dose (%ID/mL) were assessed. Behavioural tests (grip test, rotarod, open field, pole test) were performed prior and after ischemia to access therapy effects on sensorimotor functions. Ex vivo analyses (IHC, IF, WB) were performed to quantify TSPO expression, immune cells including microglia/macrophages (Iba-1, F4/80), astrocytes (GFAP) and peripheral markers in serum such as thiobarbituric acid reactive substances (TBARS) and nitric oxide (NO) 35 days post ischemia. Additionally, gene expression of pro- and anti-inflammatory markers were assessed by rt-qPCR, including tspo, cd163, arg1, tnf and Il-1β. Results: No treatment effect was observed on temporal [18F]DPA-714 uptake within the ischemic and contralateral region (two-way RM ANOVA, p = 0.71). Quantification of the percentage of TSPO+ area by immunoreactivity indicated a slight 2-fold increase in TSPO expression within the infarct region in HT-fed mice at day 35 post ischemia (p = 0.011) correlating with a 2-3 fold increase in Iba-1+ cell population expressing CD163 as anti-inflammatory marker (R2 = 0.80). Most of the GFAP+ cells were TSPO-. Only few F4/80+ cells were observed at day 35 post ischemia in both groups. No significant treatment effect was observed on global ADC and CBF within the infarct and the contralateral region over time. Behavioural tests indicated improved strength of the forepaws at day 14 post ischemia (p = 0.031). Conclusion: An HT-enriched diet significantly increased the number of Iba-1+ microglia/macrophages in the post-ischemic area, inducing higher expression of anti-inflammatory markers while no clear-cut effect was observed. Also, HT did not affect recovery of the cerebrovascular parameters, including ADC and CBF. Altogether, our data indicated that a prolonged dietary intervention with HT, as a single component of the Mediterranean diet, induces molecular changes that may improve stroke outcomes. Therefore, we support the use of the Mediterranean diet as a multicomponent therapy approach after stroke.
Collapse
|
7
|
Calahorra J, Martínez-Lara E, Granadino-Roldán JM, Martí JM, Cañuelo A, Blanco S, Oliver FJ, Siles E. Crosstalk between hydroxytyrosol, a major olive oil phenol, and HIF-1 in MCF-7 breast cancer cells. Sci Rep 2020; 10:6361. [PMID: 32286485 PMCID: PMC7156391 DOI: 10.1038/s41598-020-63417-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 03/30/2020] [Indexed: 12/14/2022] Open
Abstract
Olive oil intake has been linked with a lower incidence of breast cancer. Hypoxic microenvironment in solid tumors, such as breast cancer, is known to play a crucial role in cancer progression and in the failure of anticancer treatments. HIF-1 is the foremost effector in hypoxic response, and given that hydroxytyrosol (HT) is one of the main bioactive compounds in olive oil, in this study we deepen into its modulatory role on HIF-1. Our results in MCF-7 breast cancer cells demonstrate that HT decreases HIF-1α protein, probably by downregulating oxidative stress and by inhibiting the PI3K/Akt/mTOR pathway. Strikingly, the expression of HIF-1 target genes does not show a parallel decrease. Particularly, adrenomedullin and vascular endothelial growth factor are up-regulated by high concentrations of HT even in HIF-1α silenced cells, pointing to HIF-1-independent mechanisms of regulation. In fact, we show, by in silico modelling and transcriptional analysis, that high doses of HT may act as an agonist of the aryl hydrocarbon receptor favoring the induction of these angiogenic genes. In conclusion, we suggest that the effect of HT in a hypoxic environment is largely affected by its concentration and involves both HIF-1 dependent and independent mechanisms.
Collapse
Affiliation(s)
- Jesús Calahorra
- Departamento de Biología Experimental, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain
| | - Esther Martínez-Lara
- Departamento de Biología Experimental, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain
| | - José M Granadino-Roldán
- Departamento de Química Física y Analítica, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain
| | - Juan M Martí
- Instituto López Neyra de Parasitología y Biomedicina, IPBLN, CSIC PTS-Granada, Armilla, 18016, Spain
| | - Ana Cañuelo
- Departamento de Biología Experimental, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain
| | - Santos Blanco
- Departamento de Biología Experimental, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain
| | - F Javier Oliver
- Instituto López Neyra de Parasitología y Biomedicina, IPBLN, CSIC PTS-Granada, Armilla, 18016, Spain
| | - Eva Siles
- Departamento de Biología Experimental, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain.
| |
Collapse
|
8
|
Martí JM, Fernández-Cortés M, Serrano-Sáenz S, Zamudio-Martinez E, Delgado-Bellido D, Garcia-Diaz A, Oliver FJ. The Multifactorial Role of PARP-1 in Tumor Microenvironment. Cancers (Basel) 2020; 12:cancers12030739. [PMID: 32245040 PMCID: PMC7140056 DOI: 10.3390/cancers12030739] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/03/2020] [Accepted: 03/15/2020] [Indexed: 02/08/2023] Open
Abstract
Poly(ADP-ribose) polymerases (PARPs), represent a family of 17 proteins implicated in a variety of cell functions; some of them possess the enzymatic ability to synthesize and attach poly (ADP-ribose) (also known as PAR) to different protein substrates by a post-translational modification; PARPs are key components in the cellular response to stress with consequences for different physiological and pathological events, especially during neoplasia. In recent years, using PARP inhibitors as antitumor agents has raised new challenges in understanding their role in tumor biology. Notably, the function of PARPs and PAR in the dynamic of tumor microenvironment is only starting to be understood. In this review, we summarized the conclusions arising from recent studies on the interaction between PARPs, PAR and key features of tumor microenvironment such as hypoxia, autophagy, tumor initiating cells, angiogenesis and cancer-associated immune response.
Collapse
|
9
|
Calahorra J, Shenk J, Wielenga VH, Verweij V, Geenen B, Dederen PJ, Peinado MÁ, Siles E, Wiesmann M, Kiliaan AJ. Hydroxytyrosol, the Major Phenolic Compound of Olive Oil, as an Acute Therapeutic Strategy after Ischemic Stroke. Nutrients 2019; 11:E2430. [PMID: 31614692 PMCID: PMC6836045 DOI: 10.3390/nu11102430] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 12/17/2022] Open
Abstract
Stroke is one of the leading causes of adult disability worldwide. After ischemic stroke, damaged tissue surrounding the irreversibly damaged core of the infarct, the penumbra, is still salvageable and is therefore a target for acute therapeutic strategies. The Mediterranean diet (MD) has been shown to lower stroke risk. MD is characterized by increased intake of extra-virgin olive oil, of which hydroxytyrosol (HT) is the foremost phenolic component. This study investigates the effect of an HT-enriched diet directly after stroke on regaining motor and cognitive functioning, MRI parameters, neuroinflammation, and neurogenesis. Stroke mice on an HT diet showed increased strength in the forepaws, as well as improved short-term recognition memory probably due to improvement in functional connectivity (FC). Moreover, mice on an HT diet showed increased cerebral blood flow (CBF) and also heightened expression of brain derived neurotrophic factor (Bdnf), indicating a novel neurogenic potential of HT. This result was additionally accompanied by an enhanced transcription of the postsynaptic marker postsynaptic density protein 95 (Psd-95) and by a decreased ionized calcium-binding adapter molecule 1 (IBA-1) level indicative of lower neuroinflammation. These results suggest that an HT-enriched diet could serve as a beneficial therapeutic approach to attenuate ischemic stroke-associated damage.
Collapse
Affiliation(s)
- Jesús Calahorra
- Department of Experimental Biology, University of Jaén, Campus Las Lagunillas s/n, 23071 Jaén, Spain.
| | - Justin Shenk
- Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Radboud Alzheimer Center, Department of Anatomy, Preclinical Imaging Centre PRIME, 6500 HB Nijmegen, The Netherlands.
| | - Vera H Wielenga
- Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Radboud Alzheimer Center, Department of Anatomy, Preclinical Imaging Centre PRIME, 6500 HB Nijmegen, The Netherlands.
| | - Vivienne Verweij
- Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Radboud Alzheimer Center, Department of Anatomy, Preclinical Imaging Centre PRIME, 6500 HB Nijmegen, The Netherlands.
| | - Bram Geenen
- Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Radboud Alzheimer Center, Department of Anatomy, Preclinical Imaging Centre PRIME, 6500 HB Nijmegen, The Netherlands.
| | - Pieter J Dederen
- Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Radboud Alzheimer Center, Department of Anatomy, Preclinical Imaging Centre PRIME, 6500 HB Nijmegen, The Netherlands.
| | - M Ángeles Peinado
- Department of Experimental Biology, University of Jaén, Campus Las Lagunillas s/n, 23071 Jaén, Spain.
| | - Eva Siles
- Department of Experimental Biology, University of Jaén, Campus Las Lagunillas s/n, 23071 Jaén, Spain.
| | - Maximilian Wiesmann
- Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Radboud Alzheimer Center, Department of Anatomy, Preclinical Imaging Centre PRIME, 6500 HB Nijmegen, The Netherlands.
| | - Amanda J Kiliaan
- Radboud University Medical Center, Donders Institute for Brain, Cognition & Behaviour, Radboud Alzheimer Center, Department of Anatomy, Preclinical Imaging Centre PRIME, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
10
|
Herrera-Marschitz M, Perez-Lobos R, Lespay-Rebolledo C, Tapia-Bustos A, Casanova-Ortiz E, Morales P, Valdes JL, Bustamante D, Cassels BK. Targeting Sentinel Proteins and Extrasynaptic Glutamate Receptors: a Therapeutic Strategy for Preventing the Effects Elicited by Perinatal Asphyxia? Neurotox Res 2018; 33:461-473. [PMID: 28844085 PMCID: PMC5766721 DOI: 10.1007/s12640-017-9795-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/29/2022]
Abstract
Perinatal asphyxia (PA) is a relevant cause of death at the time of labour, and when survival is stabilised, associated with short- and long-term developmental disabilities, requiring inordinate care by health systems and families. Its prevalence is high (1 to 10/1000 live births) worldwide. At present, there are few therapeutic options, apart from hypothermia, that regrettably provides only limited protection if applied shortly after the insult.PA implies a primary and a secondary insult. The primary insult relates to the lack of oxygen, and the secondary one to the oxidative stress triggered by re-oxygenation, formation of reactive oxygen (ROS) and reactive nitrogen (RNS) species, and overactivation of glutamate receptors and mitochondrial deficiencies. PA induces overactivation of a number of sentinel proteins, including hypoxia-induced factor-1α (HIF-1α) and the genome-protecting poly(ADP-ribose) polymerase-1 (PARP-1). Upon activation, PARP-1 consumes high amounts of ATP at a time when this metabolite is scarce, worsening in turn the energy crisis elicited by asphyxia. The energy crisis also impairs ATP-dependent transport, including glutamate re-uptake by astroglia. Nicotinamide, a PARP-1 inhibitor, protects against the metabolic cascade elicited by the primary stage, avoiding NAD+ exhaustion and the energetic crisis. Upon re-oxygenation, however, oxidative stress leads to nuclear translocation of the NF-κB subunit p65, overexpression of the pro-inflammatory cytokines IL-1β and TNF-α, and glutamate-excitotoxicity, due to impairment of glial-glutamate transport, extracellular glutamate overflow, and overactivation of NMDA receptors, mainly of the extrasynaptic type. This leads to calcium influx, mitochondrial impairment, and inactivation of antioxidant enzymes, increasing further the activity of pro-oxidant enzymes, thereby making the surviving neonate vulnerable to recurrent metabolic insults whenever oxidative stress is involved. Here, we discuss evidence showing that (i) inhibition of PARP-1 overactivation by nicotinamide and (ii) inhibition of extrasynaptic NMDA receptor overactivation by memantine can prevent the short- and long-term consequences of PA. These hypotheses have been evaluated in a rat preclinical model of PA, aiming to identify the metabolic cascades responsible for the long-term consequences induced by the insult, also assessing postnatal vulnerability to recurrent oxidative insults. Thus, we present and discuss evidence demonstrating that PA induces long-term changes in metabolic pathways related to energy and oxidative stress, priming vulnerability of cells with both the neuronal and the glial phenotype. The effects induced by PA are region dependent, the substantia nigra being particularly prone to cell death. The issue of short- and long-term consequences of PA provides a framework for addressing a fundamental issue referred to plasticity of the CNS, since the perinatal insult triggers a domino-like sequence of events making the developing individual vulnerable to recurrent adverse conditions, decreasing his/her coping repertoire because of a relevant insult occurring at birth.
Collapse
Affiliation(s)
- Mario Herrera-Marschitz
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Ronald Perez-Lobos
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
- Escuela de Tecnologia Medica, Facultad de Medicina, Universidad Andres Bello, PO Box 8370146, Santiago, Chile
| | - Carolyne Lespay-Rebolledo
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Andrea Tapia-Bustos
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Emmanuel Casanova-Ortiz
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Paola Morales
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
- Faculty of Sciences, University of Chile, Santiago, Chile
| | | | - Diego Bustamante
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Bruce K. Cassels
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
11
|
Zhang G, Zhao C, Wang Q, Gu Y, Li Z, Tao P, Chen J, Yin S. Identification of HIF-1 signaling pathway in Pelteobagrus vachelli using RNA-Seq: effects of acute hypoxia and reoxygenation on oxygen sensors, respiratory metabolism, and hematology indices. J Comp Physiol B 2017; 187:931-943. [PMID: 28353178 DOI: 10.1007/s00360-017-1083-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/26/2017] [Accepted: 03/06/2017] [Indexed: 12/19/2022]
Abstract
Oxygen is a vital element in aquatic environments. The concentration of oxygen to which aquatic organisms are exposed is influenced by salinity, water temperature, weather, and surface water runoff. Hypoxia has a serious effect on fish populations, and can lead to the loss of habitat and die-offs. Therefore, in the present study we used next-generation sequencing technology to characterize the transcriptomes of Pelteobagrus vachelli and identified 70 candidate genes in the HIF-1 signaling pathway that are important for the hypoxic response in all metazoan species. For the first time, the present study reported the effects of acute hypoxia and reoxygenation on oxygen sensors, respiratory metabolism, and hematology indices in P. vachelli. The predicted physiological adjustments show that P. vachelli's blood oxygen-carrying capacity was increased through increased RBC, HB, and SI after hypoxia exposure. Glycolysis-related enzyme activities (PFK, HK, and PK) and LDH in the brain and liver also increased, indicating a rise in anaerobic metabolism. The observed reduction in oxidative enzyme level (CS) in the liver during hypoxia suggests a concomitant depression in aerobic metabolism. There were significant increases in oxygen sensor mRNA expression and HIF-1α protein expression during hypoxia and reoxygenation exposure, suggesting that the HIF-1 signaling pathway was activated in the liver and brain of P. vachelli in response to acute hypoxia and reoxygenation. Our findings suggest that oxygen sensors (e.g., HIF-1α) of P. vachelli are potentially useful biomarkers of environmental hypoxic exposure. These data contribute to a better understanding of the molecular mechanisms of the hypoxia signaling pathway in fish under hypoxia and reoxygenation.
Collapse
Affiliation(s)
- Guosong Zhang
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, Jiangsu, China.,Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, 222005, Jiangsu, China
| | - Cheng Zhao
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, Jiangsu, China.,Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, 222005, Jiangsu, China
| | - Qintao Wang
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, Jiangsu, China
| | - Yichun Gu
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, Jiangsu, China
| | - Zecheng Li
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, Jiangsu, China
| | - Panfeng Tao
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, Jiangsu, China
| | - Jiawei Chen
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, Jiangsu, China
| | - Shaowu Yin
- College of Life Sciences, Key Laboratory of Biodiversity and Biotechnology of Jiangsu Province, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, Jiangsu, China. .,Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, 222005, Jiangsu, China.
| |
Collapse
|
12
|
Martínez-Lara E, Peña A, Calahorra J, Cañuelo A, Siles E. Hydroxytyrosol decreases the oxidative and nitrosative stress levels and promotes angiogenesis through HIF-1 independent mechanisms in renal hypoxic cells. Food Funct 2016; 7:540-8. [PMID: 26608793 DOI: 10.1039/c5fo00928f] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In the kidney, tissue oxygen tension is comparatively low and this renders this organ more prone to hypoxic injury. In fact, hypoxia has a central role in the development and progression of renal disease. The recovery from this situation is dependent on the degree to which sublethally damaged cells restore normal function. The master regulator of the hypoxic response is hypoxia-inducible factor-1 (HIF-1). HIF-1 activity depends on the HIF-1α subunit level which is regulated by oxygen, nitric oxide (NO), reactive oxygen species and mTOR. Given the antioxidant and antinitrosative properties ascribed to hydroxytyrosol (HT), this study evaluates the impact of this olive oil polyphenol on the response to hypoxia in kidney cells. For this purpose, the human embryonic kidney HEK293T cell line was treated with HT and cultured under sublethal hypoxic conditions. Our results demonstrate that HT treatment decreases both, post-hypoxic reactive oxygen species and NO levels and, consequently, HIF-1α accumulation. However, HT does not affect mTOR activation or the factor inhibiting HIF level but promotes the expression of angiogenic proteins, suggesting that HT activates an adaptive response to hypoxia in a HIF-1α-independent pathway. In fact, this effect could be ascribed to the up-regulation of estrogen-related receptor α. In conclusion, our results suggest that in renal hypoxia, HT treatment might act as an effective preventive therapeutic approach to decrease stress and to improve the adaptive response to this pathological situation.
Collapse
Affiliation(s)
- Esther Martínez-Lara
- Department of Experimental Biology, University of Jaén, Paraje Las Lagunillas s/n, 23071-Jaén, Spain.
| | - Ana Peña
- Department of Experimental Biology, University of Jaén, Paraje Las Lagunillas s/n, 23071-Jaén, Spain.
| | - Jesús Calahorra
- Department of Experimental Biology, University of Jaén, Paraje Las Lagunillas s/n, 23071-Jaén, Spain.
| | - Ana Cañuelo
- Department of Experimental Biology, University of Jaén, Paraje Las Lagunillas s/n, 23071-Jaén, Spain.
| | - Eva Siles
- Department of Experimental Biology, University of Jaén, Paraje Las Lagunillas s/n, 23071-Jaén, Spain.
| |
Collapse
|
13
|
Yao Y, Zhang M, Liu T, Zhou J, Gao Y, Wen Z, Guan J, Zhu J, Lin Z, He D. Perfluorocarbon-Encapsulated PLGA-PEG Emulsions as Enhancement Agents for Highly Efficient Reoxygenation to Cell and Organism. ACS APPLIED MATERIALS & INTERFACES 2015; 7:18369-18378. [PMID: 26222132 DOI: 10.1021/acsami.5b04226] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Perfluorocarbon (PFC), a kind of oxygen carrier, is encapsulated in PLGA-PEG to prepare a PLGA-PEG/PFC emulsion for highly efficient reoxygenation to cell and organism. HCT 116 cells are used as a model cell, whose viability has a significant enhancement after reoxygenation with PLGA-PEG/PFC emulsion because of the sufficient and timely oxygen supply. Meanwhile, hypoxia-reoxygenation injury will happen along with cell hypoxia-reoxygenation treatment, which is reflected by increasing reactive oxygen species (ROS) in cells. However, the integration of intracellular ROS and cell viability implies that the degree of hypoxia-reoxygenation injury is sublethal to HCT116 cells when the concentration of PLGA-PEG/PFC emulsion is lower than 0.2 mg/mL. Furthermore, the change of the expression level of hypoxia-inducible factor-1α (HIF-1α) is similar to that of cell viability during reoxygenation, which suggests that HIF-1α or its downstream proteins may make a significant contribution to cell viability. In vivo oxygen supply is assessed in rats through pulmonary delivery, which shows that PLGA-PEG/PFC emulsion can supply oxygen to rats and improve rats' lung ventilation.
Collapse
Affiliation(s)
- Yanjie Yao
- School of Materials Science and Engineering, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Minmin Zhang
- Department of Gastroenterology, ChangHai Hospital, Second Military Medical University , Shanghai 200433, P. R. China
| | - Tian Liu
- School of Materials Science and Engineering, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Juan Zhou
- National Engineering Research Center for Nanotechnology , 28 East Jiang Chuan Road, Shanghai 200241, P. R. China
| | - Yuan Gao
- Department of Gastroenterology, ChangHai Hospital, Second Military Medical University , Shanghai 200433, P. R. China
| | - Zhengfeng Wen
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai 200003, P. R. China
| | - Jun Guan
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai 200003, P. R. China
| | - Jun Zhu
- National Engineering Research Center for Nanotechnology , 28 East Jiang Chuan Road, Shanghai 200241, P. R. China
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences , Shanghai 200032, P. R. China
| | - Zhaofen Lin
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai 200003, P. R. China
| | - Dannong He
- School of Materials Science and Engineering, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, P. R. China
- National Engineering Research Center for Nanotechnology , 28 East Jiang Chuan Road, Shanghai 200241, P. R. China
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences , Shanghai 200032, P. R. China
| |
Collapse
|
14
|
Bai P, Nagy L, Fodor T, Liaudet L, Pacher P. Poly(ADP-ribose) polymerases as modulators of mitochondrial activity. Trends Endocrinol Metab 2015; 26:75-83. [PMID: 25497347 DOI: 10.1016/j.tem.2014.11.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 12/21/2022]
Abstract
Mitochondria are essential in cellular stress responses. Mitochondrial output to environmental stress is a major factor in metabolic adaptation and is regulated by a complex network of energy and nutrient sensing proteins. Activation of poly(ADP-ribose) polymerases (PARPs) has been known to impair mitochondrial function; however, our view of PARP-mediated mitochondrial dysfunction and injury has only recently fundamentally evolved. In this review, we examine our current understanding of PARP-elicited mitochondrial damage, PARP-mediated signal transduction pathways, transcription factors that interact with PARPs and govern mitochondrial biogenesis, as well as mitochondrial diseases that are mediated by PARPs. With PARP activation emerging as a common underlying mechanism in numerous pathologies, a better understanding the role of various PARPs in mitochondrial regulation may help open new therapeutic avenues.
Collapse
Affiliation(s)
- Peter Bai
- Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE Lendület Laboratory of Cellular Metabolism Research Group, Debrecen, Hungary; Department of Medical Chemistry, University of Debrecen, Debrecen, Hungary.
| | - Lilla Nagy
- MTA-DE Lendület Laboratory of Cellular Metabolism Research Group, Debrecen, Hungary; Department of Medical Chemistry, University of Debrecen, Debrecen, Hungary
| | - Tamás Fodor
- MTA-DE Lendület Laboratory of Cellular Metabolism Research Group, Debrecen, Hungary; Department of Medical Chemistry, University of Debrecen, Debrecen, Hungary
| | - Lucas Liaudet
- Department of Intensive Care Medicine and Burn Center, Lausanne University Hospital Medical Center, Lausanne, Switzerland
| | - Pal Pacher
- Laboratory Physiological Studies, Section on Oxidative Stress and Tissue Injury, NIH/NIAAA/DICBR, Bethesda, MD, USA
| |
Collapse
|
15
|
Rodríguez MI, Majuelos-Melguizo J, Martí Martín-Consuegra JM, Ruiz de Almodóvar M, López-Rivas A, Javier Oliver F. Deciphering the insights of poly(ADP-ribosylation) in tumor progression. Med Res Rev 2015; 35:678-97. [PMID: 25604534 DOI: 10.1002/med.21339] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors are particularly efficient against tumors with defects in the homologous recombination repair pathway. Nonetheless poly(ADP-ribosylation) (PARylation) modulates prometastasic activities and adaptation of tumor to a hostile microenvironment. Modulation of metastasis-promoting traits is possible through the alteration of key transcription factors involved in the regulation of the hypoxic response, the recruitment of new vessels (or angiogenesis), and the stimulation of epithelial to mesenchymal transition (EMT). In this review, we summarized some of the findings that focalize on PARP-1's action on tumor aggressiveness, suggesting new therapeutic opportunities against an assembly of tumors not necessarily bearing DNA repair defects. Metastasis accounts for the vast majority of mortality derived from solid cancer. PARP-1 is an active player in tumor adaptation to metastasis and PARP inhibitors, recognized as promising therapeutic agents against homologous recombination deficient tumors, has novel properties responsible for the antimetastatic actions in different tumor settings.
Collapse
Affiliation(s)
- María Isabel Rodríguez
- Instituto de Parasitología y Biomedicina López Neyra (IPBLN), CSIC, Granada, Spain, 18016
| | - Jara Majuelos-Melguizo
- Instituto de Parasitología y Biomedicina López Neyra (IPBLN), CSIC, Granada, Spain, 18016
| | | | | | - Abelardo López-Rivas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Consejo Superior de Investigaciones Científicas, Sevilla, Spain, 41092
| | | |
Collapse
|
16
|
Abstract
Cerebral ischemia occurs when blood flow to the brain is insufficient to meet metabolic demand. This can result from cerebral artery occlusion that interrupts blood flow, limits CNS supply of oxygen and glucose, and causes an infarction/ischemic stroke. Ischemia initiates a cascade of molecular events in neurons and cerebrovascular endothelial cells including energy depletion, dissipation of ion gradients, calcium overload, excitotoxicity, oxidative stress, and accumulation of ions and fluid. Blood-brain barrier (BBB) disruption is associated with cerebral ischemia and leads to vasogenic edema, a primary cause of stroke-associated mortality. To date, only a single drug has received US Food and Drug Administration (FDA) approval for acute ischemic stroke treatment, recombinant tissue plasminogen activator (rt-PA). While rt-PA therapy restores perfusion to ischemic brain, considerable tissue damage occurs when cerebral blood flow is reestablished. Therefore, there is a critical need for novel therapeutic approaches that can "rescue" salvageable brain tissue and/or protect BBB integrity during ischemic stroke. One class of drugs that may enable neural cell rescue following cerebral ischemia/reperfusion injury is the HMG-CoA reductase inhibitors (i.e., statins). Understanding potential CNS drug delivery pathways for statins is critical to their utility in ischemic stroke. Here, we review molecular pathways associated with cerebral ischemia and novel approaches for delivering drugs to treat ischemic disease. Specifically, we discuss utility of endogenous BBB drug uptake transporters such as organic anion transporting polypeptides and nanotechnology-based carriers for optimization of CNS drug delivery. Overall, this chapter highlights state-of-the-art technologies that may improve pharmacotherapy of cerebral ischemia.
Collapse
|
17
|
Catrina SB. Impaired hypoxia-inducible factor (HIF) regulation by hyperglycemia. J Mol Med (Berl) 2014; 92:1025-34. [PMID: 25027070 DOI: 10.1007/s00109-014-1166-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 05/06/2014] [Accepted: 05/08/2014] [Indexed: 12/13/2022]
Abstract
The mechanisms that contribute to the development of diabetes complications remain unclear. A defective reaction of tissues to hypoxia has recently emerged as a new pathogenic mechanism and consists of a complex repression of hypoxia-inducible factor (HIF), which is the main regulator of the adaptive response to hypoxia. This paper discusses the mechanisms by which hyperglycaemia contributes to HIF repression in diabetes. Furthermore, a comprehensive analysis of the functional relevance of these new findings to the development of chronic diabetes complications is provided, along with examples from animal models and clinics.
Collapse
Affiliation(s)
- Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska University Hospital, L1:01, 17176, Stockholm, Sweden,
| |
Collapse
|
18
|
Thompson BJ, Sanchez-Covarrubias L, Slosky LM, Zhang Y, Laracuente ML, Ronaldson PT. Hypoxia/reoxygenation stress signals an increase in organic anion transporting polypeptide 1a4 (Oatp1a4) at the blood-brain barrier: relevance to CNS drug delivery. J Cereb Blood Flow Metab 2014; 34:699-707. [PMID: 24473481 PMCID: PMC3982098 DOI: 10.1038/jcbfm.2014.4] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 12/27/2013] [Accepted: 12/30/2013] [Indexed: 11/09/2022]
Abstract
Cerebral hypoxia and subsequent reoxygenation stress (H/R) is a component of several diseases. One approach that may enable neural tissue rescue after H/R is central nervous system (CNS) delivery of drugs with brain protective effects such as 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (i.e., statins). Our present in vivo data show that atorvastatin, a commonly prescribed statin, attenuates poly (ADP-ribose) polymerase (PARP) cleavage in the brain after H/R, suggesting neuroprotective efficacy. However, atorvastatin use as a CNS therapeutic is limited by poor blood-brain barrier (BBB) penetration. Therefore, we examined regulation and functional expression of the known statin transporter organic anion transporting polypeptide 1a4 (Oatp1a4) at the BBB under H/R conditions. In rat brain microvessels, H/R (6% O2, 60 minutes followed by 21% O2, 10 minutes) increased Oatp1a4 expression. Brain uptake of taurocholate (i.e., Oap1a4 probe substrate) and atorvastatin were reduced by Oatp inhibitors (i.e., estrone-3-sulfate and fexofenadine), suggesting involvement of Oatp1a4 in brain drug delivery. Pharmacological inhibition of transforming growth factor-β (TGF-β)/activin receptor-like kinase 5 (ALK5) signaling with the selective inhibitor SB431542 increased Oatp1a4 functional expression, suggesting a role for TGF-β/ALK5 signaling in Oatp1a4 regulation. Taken together, our novel data show that targeting an endogenous BBB drug uptake transporter (i.e., Oatp1a4) may be a viable approach for optimizing CNS drug delivery for treatment of diseases with an H/R component.
Collapse
Affiliation(s)
- Brandon J Thompson
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | | | - Lauren M Slosky
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Yifeng Zhang
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Mei-li Laracuente
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Patrick T Ronaldson
- 1] Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA [2] Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona, USA
| |
Collapse
|
19
|
Wang F, Zhou X, Liu W, Sun X, Chen C, Hudson LG, Jian Liu K. Arsenite-induced ROS/RNS generation causes zinc loss and inhibits the activity of poly(ADP-ribose) polymerase-1. Free Radic Biol Med 2013; 61:249-56. [PMID: 23602911 PMCID: PMC3766412 DOI: 10.1016/j.freeradbiomed.2013.04.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 03/19/2013] [Accepted: 04/12/2013] [Indexed: 12/16/2022]
Abstract
Arsenic enhances the genotoxicity of other carcinogenic agents such as ultraviolet radiation and benzo[a]pyrene. Recent reports suggest that inhibition of DNA repair is an important aspect of arsenic cocarcinogenesis, and DNA repair proteins such as poly(ADP ribose) polymerase (PARP)-1 are direct molecular targets of arsenic. Although arsenic has been shown to generate reactive oxygen/nitrogen species (ROS/RNS), little is known about the role of arsenic-induced ROS/RNS in the mechanism underlying arsenic inhibition of DNA repair. We report herein that arsenite-generated ROS/RNS inhibits PARP-1 activity in cells. Cellular exposure to arsenite, as well as hydrogen peroxide and NONOate (nitric oxide donor), decreased PARP-1 zinc content, enzymatic activity, and PARP-1 DNA binding. Furthermore, the effects of arsenite on PARP-1 activity, DNA binding, and zinc content were partially reversed by the antioxidant ascorbic acid, catalase, and the NOS inhibitor, aminoguanidine. Most importantly, arsenite incubation with purified PARP-1 protein in vitro did not alter PARP-1 activity or DNA-binding ability, whereas hydrogen peroxide or NONOate retained PARP-1 inhibitory activity. These results strongly suggest that cellular generation of ROS/RNS plays an important role in arsenite inhibition of PARP-1 activity, leading to the loss of PARP-1 DNA-binding ability and enzymatic activity.
Collapse
Affiliation(s)
- Feng Wang
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131, USA; Department of Nutrition and Food Hygiene, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xixi Zhou
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131, USA
| | - Wenlan Liu
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131, USA
| | - Xi Sun
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131, USA
| | - Chen Chen
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131, USA
| | - Laurie G Hudson
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131, USA.
| | - Ke Jian Liu
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131, USA.
| |
Collapse
|
20
|
Hypobaric Hypoxia and Reoxygenation Induce Proteomic Profile Changes in the Rat Brain Cortex. Neuromolecular Med 2012; 15:82-94. [DOI: 10.1007/s12017-012-8197-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 08/24/2012] [Indexed: 10/27/2022]
|
21
|
Martínez-Romero R, Cañuelo A, Siles E, Oliver FJ, Martínez-Lara E. Nitric oxide modulates hypoxia-inducible factor-1 and poly(ADP-ribose) polymerase-1 cross talk in response to hypobaric hypoxia. J Appl Physiol (1985) 2012; 112:816-23. [DOI: 10.1152/japplphysiol.00898.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The physiological response to hypobaric hypoxia represents a complex network of biochemical pathways in which the nitrergic system plays an important role. Previous studies have provided evidence for an interplay between the hypoxia-inducible factor-1 (HIF-1) and poly(ADP-ribose) polymerase-1 (PARP-1) under hypoxia. Here, we evaluate the potential involvement of nitric oxide (NO) in the cross talk between these two proteins. With this aim, we studied comparatively the effect of pharmacological inhibitors of NO production or PARP activity in the response of the mouse cerebral cortex to 4 h of exposure to a simulated altitude of 31,000 ft. Particularly, we analyzed the NO and reactive oxygen species production, the expression of NO synthase (NOS) isoforms, PARP-1 activity, HIF-1α expression and HIF-1 transcriptional activity, the protein level of the factor inhibiting HIF, and, finally, beclin-1 and fractin expression, as markers of cellular damage. Our results demonstrate that the reduction of NO level did not affect reactive oxygen species production but significantly 1) dampened the posthypoxic increase in neuronal NOS and inducible NOS expression without altering endothelial NOS protein level; 2) prevented PARP activation; 3) decreased HIF-1α response to hypoxia; 4) achieved a higher long-term HIF-1 transcriptional activity by reducing factor inhibiting HIF expression; and 5) reduced hypoxic damage. The pharmacological inhibition of PARP reproduced the NOS expression pattern and the HIF-1α response observed in NOS-inhibited mice, supporting its involvement in the NO-dependent regulation of hypoxia. As a whole, these results provide new data about the molecular mechanism underlying the beneficial effects of controlling NO production under hypobaric hypoxic conditions.
Collapse
Affiliation(s)
| | - Ana Cañuelo
- Department of Experimental Biology, University of Jaén, Jaén; and
| | - Eva Siles
- Department of Experimental Biology, University of Jaén, Jaén; and
| | - F. Javier Oliver
- Institute of Parasitology and Biomedicine, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | | |
Collapse
|
22
|
Rus A, Castro L, Del Moral ML, Peinado A. Inducible NOS inhibitor 1400W reduces hypoxia/re-oxygenation injury in rat lung. Redox Rep 2010; 15:169-78. [PMID: 20663293 DOI: 10.1179/174329210x12650506623609] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Nitric oxide (NO(*)) from inducible NO(*) synthase (iNOS) has been reported to either protect against, or contribute to, hypoxia/re-oxygenation lung injury. The present work aimed to clarify this double role in the hypoxic lung. With this objective, a follow-up study was made in Wistar rats submitted to hypoxia/re-oxygenation (hypoxia for 30 min; re-oxygenation of 0 h, 48 h, and 5 days), with or without prior treatment with the selective iNOS inhibitor 1400W (10 mg/kg). NO(*) levels (NOx), lipid peroxidation, apoptosis, and protein nitration were analysed. This is the first time-course study which investigates the effects of 1400W during hypoxia/re-oxygenation in the rat lung. The results showed that the administration of 1400W lowered NOx levels in all the experimental groups. In addition, lipid peroxidation, the percentage of apoptotic cells, and nitrated protein expression fell in the late post-hypoxia period (48 h and 5 days). Our results reveal that the inhibition of iNOS in the hypoxic lung reduced the damage observed before the treatment with 1400W, suggesting that iNOS-derived NO(*) may exert a negative effect on this organ during hypoxia/re-oxygenation. These findings are notable, since they indicate that any therapeutic strategy aimed at controlling excess generation of NO(*) from iNOS may be useful in alleviating NO(*)-mediated adverse effects in hypoxic lungs.
Collapse
Affiliation(s)
- Alma Rus
- Department of Experimental Biology, University of Jaén, Jaén, Spain
| | | | | | | |
Collapse
|
23
|
Mangerich A, Bürkle A. How to kill tumor cells with inhibitors of poly(ADP-ribosyl)ation. Int J Cancer 2010; 128:251-65. [DOI: 10.1002/ijc.25683] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2010] [Accepted: 08/19/2010] [Indexed: 02/07/2023]
|
24
|
Lee KH. CaMKII Inhibitor KN-62 Blunts Tumor Response to Hypoxia by Inhibiting HIF-1α in Hepatoma Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2010; 14:331-6. [PMID: 21165333 DOI: 10.4196/kjpp.2010.14.5.331] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 10/08/2010] [Accepted: 10/18/2010] [Indexed: 12/22/2022]
Abstract
In rapidly growing tumors, hypoxia commonly develops due to the imbalance between O(2) consumption and supply. Hypoxia Inducible Factor (HIF)-1α is a transcription factor responsible for tumor growth and angiogenesis in the hypoxic microenvironment; thus, its inhibition is regarded as a promising strategy for cancer therapy. Given that CamKII or PARP inhibitors are emerging anticancer agents, we investigated if they have the potential to be developed as new HIF-1α-targeting drugs. When treating various cancer cells with the inhibitors, we found that a CamKII inhibitor, KN-62, effectively suppressed HIF-1α specifically in hepatoma cells. To examine the effect of KN-62 on HIF-1α-driven gene expression, we analyzed the EPO-enhancer reporter activity and mRNA levels of HIF-1α downstream genes, such as EPO, LOX and CA9. Both the reporter activity and the mRNA expression were repressed by KN-62. We also found that KN-62 suppressed HIF-1α by impairing synthesis of HIF-1α protein. Based on these results, we propose that KN-62 is a candidate as a HIF-1α-targeting anticancer agent.
Collapse
Affiliation(s)
- Kyoung-Hwa Lee
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-799, Korea
| |
Collapse
|
25
|
Chan N, Pires IM, Bencokova Z, Coackley C, Luoto KR, Bhogal N, Lakshman M, Gottipati P, Oliver FJ, Helleday T, Hammond EM, Bristow RG. Contextual synthetic lethality of cancer cell kill based on the tumor microenvironment. Cancer Res 2010; 70:8045-54. [PMID: 20924112 DOI: 10.1158/0008-5472.can-10-2352] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Acute and chronic hypoxia exists within the three-dimensional microenvironment of solid tumors and drives therapy resistance, genetic instability, and metastasis. Replicating cells exposed to either severe acute hypoxia (16 hours with 0.02% O(2)) followed by reoxygenation or moderate chronic hypoxia (72 hours with 0.2% O(2)) treatments have decreased homologous recombination (HR) protein expression and function. As HR defects are synthetically lethal with poly(ADP-ribose) polymerase 1 (PARP1) inhibition, we evaluated the sensitivity of repair-defective hypoxic cells to PARP inhibition. Although PARP inhibition itself did not affect HR expression or function, we observed increased clonogenic killing in HR-deficient hypoxic cells following chemical inhibition of PARP1. This effect was partially reversible by RAD51 overexpression. PARP1(-/-) murine embryonic fibroblasts (MEF) showed a proliferative disadvantage under hypoxic gassing when compared with PARP1(+/+) MEFs. PARP-inhibited hypoxic cells accumulated γH2AX and 53BP1 foci as a consequence of altered DNA replication firing during S phase-specific cell killing. In support of this proposed mode of action, PARP inhibitor-treated xenografts displayed increased γH2AX and cleaved caspase-3 expression in RAD51-deficient hypoxic subregions in vivo, which was associated with decreased ex vivo clonogenic survival following experimental radiotherapy. This is the first report of selective cell killing of HR-defective hypoxic cells in vivo as a consequence of microenvironment-mediated "contextual synthetic lethality." As all solid tumors contain aggressive hypoxic cells, this may broaden the clinical utility of PARP and DNA repair inhibition, either alone or in combination with radiotherapy and chemotherapy, even in tumor cells lacking synthetically lethal, genetic mutations.
Collapse
Affiliation(s)
- Norman Chan
- Princess Margaret Hospital (University Health Network) and Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Strosznajder RP, Czubowicz K, Jesko H, Strosznajder JB. Poly(ADP-ribose) metabolism in brain and its role in ischemia pathology. Mol Neurobiol 2010; 41:187-96. [PMID: 20411356 DOI: 10.1007/s12035-010-8124-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 03/23/2010] [Indexed: 11/24/2022]
Abstract
The biological roles of poly(ADP-ribose) polymers (PAR) and poly(ADP-ribosyl)ation of proteins in the central nervous system are diverse. The homeostasis of PAR orchestrated by poly(ADP-ribose) polymerase-1 (PARP-1) and poly(ADP-ribose) glycohydrolase (PARG) is crucial for cell physiology and pathology. Both enzymes are ubiquitously distributed in neurons and glia; however, they are segregated at the subcellular level. PARP-1 serves as a "nick sensor" for single- or double-stranded breaks in DNA and is involved in long and short patch base-excision repair, while PARG breaks down PAR. The stimulation of PARP-1 and PAR formation can activate proinflammatory transcription factors, including nuclear factor kappa B. However, hyperactivation of PARP-1 can result in depletion of NAD/ATP, and in PAR-dependent mitochondrial pore formation leading to release of apoptosis inducing factor and cell death. The role of PAR as a death signaling molecule in brain ischemia-reperfusion and inflammation as well as the effect of gender and aging is presented in this review. Modulating the PAR level through pharmacological or genetic intervention on PARP-1/PARG activity and gene expression should be a valuable way for neuroprotective strategy.
Collapse
Affiliation(s)
- Robert Piotr Strosznajder
- Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego St., 02-106, Warsaw, Poland.
| | | | | | | |
Collapse
|
27
|
Giansanti V, Donà F, Tillhon M, Scovassi AI. PARP inhibitors: new tools to protect from inflammation. Biochem Pharmacol 2010; 80:1869-77. [PMID: 20417190 DOI: 10.1016/j.bcp.2010.04.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 04/12/2010] [Accepted: 04/13/2010] [Indexed: 10/19/2022]
Abstract
Poly(ADP-ribosylation) consists in the conversion of β-NAD(+) into ADP-ribose, which is then bound to acceptor proteins and further used to form polymers of variable length and structure. The correct turnover of poly(ADP-ribose) is ensured by the concerted action of poly(ADP-ribose) polymerase (PARP) and poly(ADP-ribose) glycohydrolase (PARG) enzymes, which are responsible for polymer synthesis and degradation, respectively. Despite the positive role of poly(ADP-ribosylation) in sensing and repairing DNA damage, generated also by ROS, PARP over-activation could allow NAD depletion and consequent necrosis, thus leading to an inflammatory condition in many diseases. In this respect, inhibition of PARP enzymes could exert a protective role towards a number of pathological conditions; i.e. the combined treatment of tumors with PARP inhibitors/anticancer agents proved to have a beneficial effect in cancer therapy. Thus, pharmacological inactivation of poly(ADP-ribosylation) could represent a novel therapeutic strategy to limit cellular injury and to attenuate the inflammatory processes that characterize many disorders.
Collapse
Affiliation(s)
- Vincenzo Giansanti
- Istituto di Genetica Molecolare CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy
| | | | | | | |
Collapse
|