scholarly journals Oxidative Stress and the Microbiota-Gut-Brain Axis

2018 ◽  
Vol 2018 ◽  
pp. 1-12 ◽  
Author(s):  
Laura Dumitrescu ◽  
Iulia Popescu-Olaru ◽  
Liviu Cozma ◽  
Delia Tulbă ◽  
Mihail Eugen Hinescu ◽  
...  

The gut-brain axis is increasingly recognized as an important pathway of communication and of physiological regulation, and gut microbiota seems to play a significant role in this mutual relationship. Oxidative stress is one of the most important pathogenic mechanisms for both neurodegenerative diseases, such as Alzheimer’s or Parkinson’s, and acute conditions, such as stroke or traumatic brain injury. A peculiar microbiota type might increase brain inflammation and reactive oxygen species levels and might favor abnormal aggregation of proteins. Reversely, brain lesions of various etiologies result in alteration of gut properties and microbiota. These recent hypotheses could open a door for new therapeutic approaches in various neurological diseases.

2012 ◽  
Vol 2012 ◽  
pp. 1-7 ◽  
Author(s):  
W. H. Fan ◽  
M. M. Cui ◽  
Z. W. Shi ◽  
C. Tan ◽  
X. P. Yang

This study examines the potential hazard of an individual nanomaterial on the Cu biotoxicity to aquatic organisms.Daphnia magnain the absence or presence of nano-TiO2was exposed to Cu. Maintaining nano-TiO2at a safe concentration cannot eliminate its potential hazard. The biomarkers superoxide dismutase, catalase, and Na+/K+-ATPase inD. magnawere measured. Cu in the presence of nano-TiO2induced higher levels of oxidative stress and physiological damage because of the sorption of Cu. Nano-TiO2also caused Na+/K+-ATPase inhibition possibly by impeding the Na+/K+transfer channel. The correlations among the biomarkers, mortality, and accumulation further showed that the overloading reactive oxygen species generation caused by nano-TiO2contributed to deeper oxidative stress and physiological regulation, thereby causing greater toxic injury.


Author(s):  
Joana Magalhaes ◽  
Emilie Tresse ◽  
Patrick Ejlerskov ◽  
Erling Hu ◽  
Yawei Liu ◽  
...  

AbstractFamilial Parkinson disease (PD) is associated with rare genetic mutations, but the etiology in most patients with sporadic (s)PD is largely unknown, and the basis for its progression to dementia (sPDD) is poorly characterized. We have identified that loss of IFNβ or IFNAR1, the receptor for IFNα/β, causes pathological and behavioral changes resembling PDD, prompting us to hypothesize that dysregulated genes in IFNβ-IFNAR signaling pathway predispose one to sPD. By transcriptomic analysis, we found defective neuronal IFNβ-IFNAR signaling, including particularly elevated PIAS2 associated with sPDD. With meta-analysis of GWASs, we identified sequence variants in IFNβ-IFNAR-related genes in sPD patients. Furthermore, sPDD patients expressed higher levels of PIAS2 mRNA and protein in neurons. To determine its function in brain, we overexpressed PIAS2 under a neuronal promoter, alone or with human α-synuclein, in the brains of mice, which caused motor and cognitive impairments and correlated with intraneuronal phosphorylated (p)α-synuclein accumulation and dopaminergic neuron loss. Ectopic expression of neuronal PIAS2 blocked mitophagy, increased the accumulation of senescent mitochondrial and oxidative stress, as evidenced by excessive oxDJ1 and 8OHdG, by inactivating ERK1/2-P53 signaling. Conversely, PIAS2 knockdown rescued the clinicopathological manifestations of PDD in Ifnb–/– mice on restoring mitochondrial homeostasis, oxidative stress, and pERK1/2-pP53 signaling. The regulation of JAK-STAT2-PIAS2 signaling was crucial for neurite outgrowth and neuronal survival and excitability and thus might prevent cognitive impairments. Our findings provide insights into the progression of sPD and dementia and have implications for new therapeutic approaches.


Author(s):  
Felipe Salazar-Ramírez ◽  
Roberto Ramos-Mondragón ◽  
Gerardo García-Rivas

Ca2+ plays a pivotal role in mitochondrial energy production, contraction, and apoptosis. Mitochondrial Ca2+-targeted fluorescent probes have demonstrated that mitochondria Ca2+ transients are synchronized with Ca2+ fluxes occurring in the sarcoplasmic reticulum (SR). The presence of specialized proteins tethering SR to mitochondria ensures the local Ca2+ flux between these organelles. Furthermore, communication between SR and mitochondria impacts their functionality in a bidirectional manner. Mitochondrial Ca2+ uptake through the mitochondrial Ca2+ uniplex is essential for ATP production and controlled reactive oxygen species levels for proper cellular signaling. Conversely, mitochondrial ATP ensures the proper functioning of SR Ca2+-handling proteins, which ensures that mitochondria receive an adequate supply of Ca2+. Recent evidence suggests that altered SR Ca2+ proteins, such as ryanodine receptors and the sarco/endoplasmic reticulum Ca2+ ATPase pump, play an important role in maintaining proper cardiac membrane excitability, which may be initiated and potentiated when mitochondria are dysfunctional. This recognized mitochondrial role offers the opportunity to develop new therapeutic approaches aimed at preventing cardiac arrhythmias in cardiac disease.


2020 ◽  
Vol 26 (12) ◽  
pp. 1263-1276 ◽  
Author(s):  
Mónica E. Ureña-Guerrero ◽  
José L. Castañeda-Cabral ◽  
Martha C. Rivera-Cervantes ◽  
Rafael J. Macias-Velez ◽  
José J. Jarero-Basulto ◽  
...  

Background: Erythropoietin (Epo) and vascular endothelial growth factor (VEGF) are two vasoactive molecules with essential trophic effects for brain development. The expression and secretion of both molecules increase in response to neuronal damage and they exert protective and restorative effects, which may also be accompanied by adverse side effects. Objective: We review the most relevant evidence on the neuroprotective and neurorestorative effects of Epo and VEGF in three of the most frequent neurological disorders, namely, stroke, epilepsy and Alzheimer's disease, to develop new therapeutic approaches. Method: Several original scientific manuscripts and reviews that have discussed the evidence in critical way, considering both the beneficial and adverse effects of Epo and VEGF in the selected neurological disorders, were analysed. In addition, throughout this review, we propose several considerations to take into account in the design of therapeutic approaches based on Epo and VEGF signalling. Results: Although the three selected disorders are triggered by different mechanisms, they evolve through similar processes: excitotoxicity, oxidative stress, neuroinflammation, neuronal death, glial reactivity and vascular remodelling. Epo and VEGF exert neuroprotective and neurorestorative effects by acting on these processes due to their pleiotropism. In general, the evidence shows that both Epo and VEGF reduce neuronal death but that at the vascular level, their effects are contradictory. Conclusion: Because the Epo and VEGF signalling pathways are connected in several ways, we conclude that more experimental studies, primarily studies designed to thoroughly assess the functional interactions between Epo and VEGF in the brain under both physiological and pathophysiological conditions, are needed.


Biomolecules ◽  
2020 ◽  
Vol 10 (8) ◽  
pp. 1119 ◽  
Author(s):  
Amany Tawfik ◽  
Yara A. Samra ◽  
Nehal M. Elsherbiny ◽  
Mohamed Al-Shabrawey

Elevated plasma homocysteine (Hcy) level, known as hyperhomocysteinemia (HHcy) has been linked to different systemic and neurological diseases, well-known as a risk factor for systemic atherosclerosis and cardiovascular disease (CVD) and has been identified as a risk factor for several ocular disorders, such as diabetic retinopathy (DR) and age-related macular degeneration (AMD). Different mechanisms have been proposed to explain HHcy-induced visual dysfunction, including oxidative stress, upregulation of inflammatory mediators, retinal ganglion cell apoptosis, and extracellular matrix remodeling. Our previous studies using in vivo and in vitro models of HHcy have demonstrated that Hcy impairs the function of both inner and outer blood retinal barrier (BRB). Dysfunction of BRB is a hallmark of vision loss in DR and AMD. Our findings highlighted oxidative stress, ER stress, inflammation, and epigenetic modifications as possible mechanisms of HHcy-induced BRB dysfunction. In addition, we recently reported HHcy-induced brain inflammation as a mechanism of blood–brain barrier (BBB) dysfunction and pathogenesis of Alzheimer’s disease (AD). Moreover, we are currently investigating the activation of glutamate receptor N-methyl-d-aspartate receptor (NMDAR) as the molecular mechanism for HHcy-induced BRB dysfunction. This review focuses on the studied effects of HHcy on BRB and the controversial role of HHcy in the pathogenesis of aging neurological diseases such as DR, AMD, and AD. We also highlight the possible mechanisms for such deleterious effects of HHcy.


Processes ◽  
2020 ◽  
Vol 8 (7) ◽  
pp. 802 ◽  
Author(s):  
Andrea Cucchi ◽  
Roberto Ramoni ◽  
Giuseppina Basini ◽  
Simona Bussolati ◽  
Fausto Quintavalla

Oxidative stress is a prominent event in several acute and chronic diseases including neoplasia. Although its direct involvement in carcinogenesis still remains to be clearly defined, a deeper knowledge of oxidative stress in oncologic patients could help to monitor their clinical outcome and to develop new therapeutic approaches. Therefore, the present study was undertaken to explore redox status in blood of neoplastic dogs affected either by multicentric lymphoma or by primary cutaneous mastocytoma. Superoxide anion (O2 •−), nitric oxide (NO) and hydroperoxides (ROOH) were measured. Detoxifying enzyme superoxide dismutase (SOD) and total non-enzymatic antioxidant capacity (ferric reducing-antioxidant power (FRAP)) were assessed. The oxidative stress index (OSi) both for enzymatic (OSiE) and non-enzymatic (OSiNE) scavengers were evaluated. Both pathologies, showed a reduced NO generation, while O2 •− levels were decreased only in mastocytoma. The oxidative stress indexes showed a significant decrease in mastocytoma patients, only for OSiE.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3639-3639 ◽  
Author(s):  
Min Hui Cui ◽  
Henny H Billett ◽  
Sandra Suzuka ◽  
Kamalakar Ambadipudi ◽  
Caterina P. Minniti ◽  
...  

Abstract Introduction: Fetal hemoglobin (HbF) inhibits the deoxygenation-induced polymerization of sickle hemoglobin (HbS), the underlying pathophysiology of sickle cell disease (SCD). Higher HbF levels are associated with reduced sickle related pathology (Lettre G, Bauer DE. The Lancet 2016; 387(10037): 2554-64.). A major therapeutic approach to SCD has been to try to increase HbF levels, primarily by hydroxyurea administration. While hydroxyurea improves HbF and reduces sickling, its mechanism of action remains controversial, and even less is known regarding its effects on sickle-related cerebral pathology. We studied transgenic Berkeley sickle mice (BERK) (Paszty C et. al., Science 1997; 278(5339): 876-8.) with different HbF levels using in vivo MRI in an effort to estimate the ameliorative effects of HbF on cerebral blood flow and brain inflammation. Methods: BERK mice, expressing exclusively human a- and bS-globins with low (LF, n=7), medium (MF, n=8) and high (HF, n=6) HbF levels were studied at 9.4 T MRI system under isoflurane anesthesia. C57BL/6J mice were used as controls (n=9). We measured whole-brain cerebral blood flow (CBF-WB) using MRI. Diffusion tensor imaging was used to measure whole-brain mean diffusivity (MD-WB), a measure of brain edema and inflammation, and white matter fractional anisotropy (FA-WM) a biomarker of neuropathology. Tail vein blood was employed to obtain hematological parameters prior to MRI. Results: Select MRI and related data are shown in Table 1. BERK-LF mice (~3% HbF) exhibited lower mean cell hemoglobin concentration (MCHC) and hematocrit (Hct) compared with C57BL mice. BERK-MF mice (~20% HbF) showed increased MCHC compared with BERK-LF mice. BERK-HF mice (~30% HbF) had the highest MCHC and Hct of all BERK mice, but MCHC was still lower than levels seen in C57BL mice. The low MCHC and Hct levels in BERK-LF mice were associated with high CBF and MD . CBF was significantly elevated in BERK-LF and BERK-MF mice by 130% and 120%, respectively, compared to C57BL mice. BERK-HF mice, on the other hand, exhibited normalized CBF (187.0±27.5 vs. 180.0±30.5 ml/100g/min, BERK-HF vs. C57BL respectively), despite lower MCHC and Hct levels compared to C57BL mice. Brain inflammation (elevated MD levels) were also observed in BERK-LF mice compared to controls. In contrast, BERK-MF and BERK-HF mice showed reduced MD; both were significantly lower than BERK-LF mice and not different from control. Only the BERK-LF demonstrated significantly lower FA values (reduced microstructuraly complexity) compared to controls. CBF in all mice (BERK and C57BL) was inversely related to Hct (r=-0.433, p=0.019) and MCHC (r=-0.527, p=0.003). However, when controls were excluded from this analysis, only HbF level significantly predicted CBF (r=-0.485, p=0.030) and MD (r=-0.636, p=0.003), shown in Figure 1. HbF level also positively predicted MCHC (r=0.832, p<0.0001) and Hct (r=0.448, p=0.042) in BERK mice. Conclusions: Cerebral perfusion is markedly elevated in BERK mice compared to C57BL control mice and is inversely correlated with Hct, MCHC and total fetal Hb level. These data suggest that increased HbF can significantly reduce CBF, and in our study CBF levels in high-gamma BERK (~30%) were indistinguishable from control CBF, despite the lower Hct levels. We also demonstrate that brain edema, as reflected in lower MD, is improved when HbF levels reach ~20%. The improved CBF and reduced edema with higher HbF may be due to several factors: an antisickling effect on HbS, reduced oxidative stress, improved NO activity, and improved blood oxygen carrying capacity. HbF levels approaching 30% improved CBF, improved FA and reduced MD, suggesting that increased HbF levels may reduce cerebral insults stemming from cerebral hyperemia. These data suggest that new therapeutic approaches to further increase HbF to even higher levels than that achieved with hydroxyurea may provide greater hematologic and neuropathologic improvements in patients with SCD. If hydroxyuyrea induced HbF increases cannot be further improved, additional or combined therapeutic approaches (such as Pegylated Hb) may be combined with HbF therapies to improve oxygen delivery, reduce oxidative stress and reduce CBF toward normal levels. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 114 (15) ◽  
pp. E3041-E3050 ◽  
Author(s):  
Blas Moreno-Beltrán ◽  
Alejandra Guerra-Castellano ◽  
Antonio Díaz-Quintana ◽  
Rebecca Del Conte ◽  
Sofía M. García-Mauriño ◽  
...  

Regulation of mitochondrial activity allows cells to adapt to changing conditions and to control oxidative stress, and its dysfunction can lead to hypoxia-dependent pathologies such as ischemia and cancer. Although cytochrome c phosphorylation—in particular, at tyrosine 48—is a key modulator of mitochondrial signaling, its action and molecular basis remain unknown. Here we mimic phosphorylation of cytochrome c by replacing tyrosine 48 with p-carboxy-methyl-l-phenylalanine (pCMF). The NMR structure of the resulting mutant reveals significant conformational shifts and enhanced dynamics around pCMF that could explain changes observed in its functionality: The phosphomimetic mutation impairs cytochrome c diffusion between respiratory complexes, enhances hemeprotein peroxidase and reactive oxygen species scavenging activities, and hinders caspase-dependent apoptosis. Our findings provide a framework to further investigate the modulation of mitochondrial activity by phosphorylated cytochrome c and to develop novel therapeutic approaches based on its prosurvival effects.


Sign in / Sign up

Export Citation Format

Share Document