Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
Add more filters










Publication year range
2.
Res Sq ; 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38645104

ABSTRACT

Background: Recent studies have shed light on the potential role of gut dysbiosis in shaping traumatic brain injury (TBI) outcomes. Changes in the levels and types of Lactobacillus bacteria present might impact the immune system disturbances, neuroinflammatory responses, anxiety and depressive-like behaviors, and compromised neuroprotection mechanisms triggered by TBI. Objective: This study aimed to investigate the effects of a daily pan-probiotic (PP) mixture in drinking water containing strains of Lactobacillus plantarum, L. reuteri, L. helveticus, L. fermentum, L. rhamnosus, L. gasseri, and L. casei, administered for either two or seven weeks before inducing TBI on both male and female mice. Methods: Mice were subjected to controlled cortical impact (CCI) injury. Short-chain fatty acids (SCFAs) analysis was performed for metabolite measurements. The taxonomic profiles of murine fecal samples were evaluated using 16S rRNA V1-V3 sequencing analysis. Histological analyses were used to assess neuroinflammation and gut changes post-TBI, while behavioral tests were conducted to evaluate sensorimotor and cognitive functions. Results: Our findings suggest that PP administration modulates the diversity and composition of the microbiome and increases the levels of SCFAs in a sex-dependent manner. We also observed a reduction of lesion volume, cell death, and microglial and macrophage activation after PP treatment following TBI in male mice. Furthermore, PP-treated mice show motor function improvements and decreases in anxiety and depressive-like behaviors. Conclusion: Our findings suggest that PP administration can mitigate neuroinflammation and ameliorate motor and anxiety and depressive-like behavior deficits following TBI. These results underscore the potential of probiotic interventions as a viable therapeutic strategy to address TBI-induced impairments, emphasizing the need for gender-specific treatment approaches.

3.
Free Radic Biol Med ; 220: 43-55, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38677488

ABSTRACT

Exercise has the unique aptitude to benefit overall health of body and brain. Evidence indicates that the effects of exercise can be saved in the epigenome for considerable time to elevate the threshold for various diseases. The action of exercise on epigenetic regulation seems central to building an "epigenetic memory" to influence long-term brain function and behavior. As an intrinsic bioenergetic process, exercise engages the function of the mitochondria and redox pathways to impinge upon molecular mechanisms that regulate synaptic plasticity and learning and memory. We discuss how the action of exercise uses mechanisms of bioenergetics to support a "epigenetic memory" with long-term implications for neural and behavioral plasticity. This information is crucial for directing the power of exercise to reduce the burden of neurological and psychiatric disorders.


Subject(s)
Brain , Cognition , Energy Metabolism , Epigenesis, Genetic , Exercise , Neuronal Plasticity , Humans , Neuronal Plasticity/genetics , Energy Metabolism/genetics , Exercise/physiology , Cognition/physiology , Brain/metabolism , Brain/physiology , Animals , Mitochondria/metabolism , Mitochondria/genetics
5.
Biochim Biophys Acta Mol Basis Dis ; 1870(2): 166937, 2024 02.
Article in English | MEDLINE | ID: mdl-37926362

ABSTRACT

Traumatic brain injury (TBI) often results in a reduction of the capacity of cells to sustain energy demands, thus, compromising neuronal function and plasticity. Here we show that the mitochondrial activator humanin (HN) counteracts a TBI-related reduction in mitochondrial bioenergetics, including oxygen consumption rate. HN normalized the disruptive action of TBI on memory function, and restored levels of synaptic proteins (synapsin 1 and p-CREB). HN also counteracted TBI-related elevations of pro-inflammatory cytokines in plasma (TNF-α, INF-y, IL 17, IL 5, MCP 5, GCSF, RANNETS, sTNFRI) as well as in the hippocampus (gp-130 and p-STAT3). Gp-130 is an integral part of cytokine receptor impinging on STAT3 (Tyr-705) signaling. Furthermore, HN reduced astrocyte proliferation in TBI. The overall evidence suggests that HN plays an integral role in normalizing fundamental aspects of TBI pathology which are central to energy balance, brain function, and plasticity.


Subject(s)
Brain Injuries, Traumatic , Cognitive Dysfunction , Mitochondrial Diseases , Rats , Animals , Rats, Sprague-Dawley , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Apoptosis Regulatory Proteins , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Inflammation/drug therapy
6.
Article in English | MEDLINE | ID: mdl-38037566

ABSTRACT

Traumatic brain injury (TBI) results in metabolic deficits and functionally compromised tissue. The BDNF mimetic R13 has a significant positive effect on both tissue metabolism and behavioral outcome after TBI, indicating a promising therapeutic. To understand the mechanism of action for this intervention, we determined whether there was any association between the underlying metabolic insult and any improvement in resting state functional connectivity (FC) with MRI, or whether R13 acts through mechanisms unrelated to metabolic recovery. We found perfusion deficits could be reasonably approximated by reductions in mean diffusivity (MD) acutely after injury, because a majority of regions with low perfusion matched to regions of low MD, indicative of cell swelling. Injury alone resulted in reduced cross-brain FC and contralateral hyperconnectivity at 1d compared to sham and these were spatially coincident with regions of low MD. R13 intervention at 1-7d altered the tissue trajectory of MD pathology away from pseudo-normalization so that a greater volume of tissue remained with low MD at 7d. These same regions were associated with significant changes in cross-brain and contralateral FC in R13 treated rats compared to injured vehicle-treated rats. These data indicate a likely metabolic effect of R13 acutely after injury.

7.
Biochim Biophys Acta Mol Basis Dis ; 1869(7): 166781, 2023 10.
Article in English | MEDLINE | ID: mdl-37286142

ABSTRACT

Traumatic brain injury (TBI) is major neurological burden globally, and effective treatments are urgently needed. TBI is characterized by a reduction in energy metabolism and synaptic function that seems a primary cause of neuronal dysfunction. R13, a small drug and BDNF mimetic showed promising results in improving spatial memory and anxiety-like behavior after TBI. Additionally, R13 was found to counteract reductions in molecules associated with BDNF signaling (p-TrkB, p-PI3K, p-AKT), synaptic plasticity (GluR2, PSD95, Synapsin I) as well as bioenergetic components such as mitophagy (SOD, PGC-1α, PINK1, Parkin, BNIP3, and LC3) and real-time mitochondrial respiratory capacity. Behavioral and molecular changes were accompanied by adaptations in functional connectivity assessed using MRI. Results highlight the potential of R13 as a therapeutic agent for TBI and provide valuable insights into the molecular and functional changes associated with this condition.


Subject(s)
Brain Injuries, Traumatic , Brain-Derived Neurotrophic Factor , Humans , Brain-Derived Neurotrophic Factor/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Signal Transduction , Mitochondria/metabolism , Energy Metabolism
8.
Biochim Biophys Acta Mol Basis Dis ; 1869(6): 166728, 2023 08.
Article in English | MEDLINE | ID: mdl-37137432

ABSTRACT

Clinical evidence indicates that injury to the brain elicits systemic metabolic disturbances that contributes to the brain pathology. Since dietary fructose is metabolized in the liver, we explored mechanisms by which traumatic brain injury (TBI) and dietary fructose influence liver function and their possible repercussions to brain. Consumption of fructose contributed to the detrimental effects of TBI on liver operation, in terms of glucose and lipid metabolism, de novo lipogenesis, lipid peroxidation. Thyroid hormone (T4) is metabolized in the liver and found that T4 supply improved lipid metabolism by reducing de novo lipogenesis, lipid accumulation, lipogenic enzymes (ACC, AceCS1, FAS), lipid peroxidation in liver in response to fructose and fructose-TBI. T4 supply also helped to normalize glucose metabolism and improve insulin sensitivity. Furthermore, T4 counteracted elevations of the pro-inflammatory cytokines, Tnfα and Mcp-1 after TBI and/or fructose intake in liver and circulation. T4 also exerted an effect on isolated primary hepatocytes by potentiating phosphorylation of AMPKα and AKT substrate, AS160, leading to increased glucose uptake. In addition, T4 restored the metabolism of DHA in the liver disrupted by TBI and fructose, adding important information to optimize the action of DHA in therapeutics. The overall evidence seems to indicate that the liver works as a gate for the regulation of the effects of brain injury and foods on brain pathologies.


Subject(s)
Brain Injuries, Traumatic , Liver , Humans , Liver/metabolism , Hepatocytes/metabolism , Fructose/pharmacology , Brain Injuries, Traumatic/metabolism , Thyroid Hormones/metabolism
9.
Sports Med Health Sci ; 4(3): 147-151, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36090919

ABSTRACT

Physical rehabilitation is an effective therapy to normalize weaknesses encountered with neurological disorders such as traumatic brain injury (TBI). However, the efficacy of exercise is limited during the acute period of TBI because of metabolic dysfunction, and this may further compromise neuronal function. Here we discuss the possibility to normalize brain metabolism during the early post-injury convalescence period to support functional plasticity and prevent long-term functional deficits. Although BDNF possesses the unique ability to support molecular events involved with the transmission of information across nerve cells through activation of its TrkB receptor, the poor pharmacokinetic profile of BDNF has limited its therapeutic applicability. The flavonoid derivative, 7,8-dihydroxyflavone (7,8-DHF), signals through the same TrkB receptors and results in the activation of BDNF signaling pathways. We discuss how the pharmacokinetic limitations of BDNF may be avoided by the use of 7,8-DHF, which makes it a promising pharmacological agent for supporting activity-based rehabilitation during the acute post-injury period after TBI. In turn, docosahexaenoic acid (C22:6n-3; DHA) is abundant in the phospholipid composition of plasma membranes in the brain and its action is important for brain development and plasticity. DHA is a major modulator of synaptic membrane fluidity and function, which is fundamental for supporting cell signaling and synaptic plasticity. Exercise influences DHA function by normalizing DHA content in the brain, such that the collaborative action of exercise and DHA can be instrumental to boost BDNF function with strong therapeutic potential for reducing the deleterious effects of TBI on synaptic plasticity and cognition.

10.
Cell Mol Life Sci ; 79(9): 480, 2022 Aug 11.
Article in English | MEDLINE | ID: mdl-35951114

ABSTRACT

BACKGROUND: The etiology of mild traumatic brain injury (mTBI) remains elusive due to the tissue and cellular heterogeneity of the affected brain regions that underlie cognitive impairments and subsequent neurological disorders. This complexity is further exacerbated by disrupted circuits within and between cell populations across brain regions and the periphery, which occur at different timescales and in spatial domains. METHODS: We profiled three tissues (hippocampus, frontal cortex, and blood leukocytes) at the acute (24-h) and subacute (7-day) phases of mTBI at single-cell resolution. RESULTS: We demonstrated that the coordinated gene expression patterns across cell types were disrupted and re-organized by TBI at different timescales with distinct regional and cellular patterns. Gene expression-based network modeling implied astrocytes as a key regulator of the cell-cell coordination following mTBI in both hippocampus and frontal cortex across timepoints, and mt-Rnr2, which encodes the mitochondrial peptide humanin, as a potential target for intervention based on its broad regional and dynamic dysregulation following mTBI. Treatment of a murine mTBI model with humanin reversed cognitive impairment caused by mTBI through the restoration of metabolic pathways within astrocytes. CONCLUSIONS: Our results offer a systems-level understanding of the dynamic and spatial regulation of gene programs by mTBI and pinpoint key target genes, pathways, and cell circuits that are amenable to therapeutics.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Animals , Brain/metabolism , Brain Injuries/metabolism , Brain Injuries, Traumatic/genetics , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins , Mice
11.
Biochim Biophys Acta Mol Basis Dis ; 1868(11): 166491, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35902006

ABSTRACT

Most efforts to understand the pathology of traumatic brain injury (TBI) have been centered on the brain, ignoring the role played by systemic physiology. Gut-derived serotonin is emerging as a major regulator of systemic homeostasis involving various organs and tissues throughout the body. Here, we shed light on the roles occupied by gut-derived serotonin and its downstream metabolic targets in the systemic pathogenesis of TBI. Male C57BL/6J mice were subjected to a fluid percussion injury (FPI) and RT-qPCR was used to examine mRNA levels in intestine, liver, and adipose tissue. In the intestinal tract, TBI transiently downregulated enteric neuronal markers Chat and Nos1 in the duodenum and colon, and altered colonic genes related to synthesis and degradation of serotonin, favoring an overall serotonin downregulation. There also was a decrease in serotonin fluorescence intensity in the colonic mucosa and reduced circulating blood serotonin levels, with concurrent alterations in serotonin-associated gene expression in downstream tissues after TBI (i.e., upregulation of serotonin receptor Htr2a and dysregulation of genes associated with lipid metabolism in liver and adipose). Levels of commensal bacterial species were also altered in the gut and were associated with TBI-mediated changes in the colonic serotonin system. Our findings suggest that TBI alters peripheral serotonin homeostasis, which in turn may impact gastrointestinal function, gut microbiota, and systemic energy balance. These data highlight the importance of building an integrative view of the role of systemic physiology in TBI pathogenesis to assist in the development of effective TBI treatments.


Subject(s)
Brain Injuries, Traumatic , Gastrointestinal Microbiome , Animals , Brain Injuries, Traumatic/pathology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger , Serotonin/metabolism
14.
Mol Syst Biol ; 17(6): e10108, 2021 06.
Article in English | MEDLINE | ID: mdl-34057817

ABSTRACT

RNA hybridization-based spatial transcriptomics provides unparalleled detection sensitivity. However, inaccuracies in segmentation of image volumes into cells cause misassignment of mRNAs which is a major source of errors. Here, we develop JSTA, a computational framework for joint cell segmentation and cell type annotation that utilizes prior knowledge of cell type-specific gene expression. Simulation results show that leveraging existing cell type taxonomy increases RNA assignment accuracy by more than 45%. Using JSTA, we were able to classify cells in the mouse hippocampus into 133 (sub)types revealing the spatial organization of CA1, CA3, and Sst neuron subtypes. Analysis of within cell subtype spatial differential gene expression of 80 candidate genes identified 63 with statistically significant spatial differential gene expression across 61 (sub)types. Overall, our work demonstrates that known cell type expression patterns can be leveraged to improve the accuracy of RNA hybridization-based spatial transcriptomics while providing highly granular cell (sub)type information. The large number of newly discovered spatial gene expression patterns substantiates the need for accurate spatial transcriptomic measurements that can provide information beyond cell (sub)type labels.


Subject(s)
Gene Expression Profiling , Transcriptome , Animals , Computer Simulation , Mice , Neurons , RNA, Messenger , Transcriptome/genetics
15.
Acta Neuropathol Commun ; 9(1): 74, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33892818

ABSTRACT

INTRODUCTION: Traumatic brain injury (TBI) is considered as the most robust environmental risk factor for Alzheimer's disease (AD). Besides direct neuronal injury and neuroinflammation, vascular impairment is also a hallmark event of the pathological cascade after TBI. However, the vascular connection between TBI and subsequent AD pathogenesis remains underexplored. METHODS: In a closed-head mild TBI (mTBI) model in mice with controlled cortical impact, we examined the time courses of microvascular injury, blood-brain barrier (BBB) dysfunction, gliosis and motor function impairment in wild type C57BL/6 mice. We also evaluated the BBB integrity, amyloid pathology as well as cognitive functions after mTBI in the 5xFAD mouse model of AD. RESULTS: mTBI induced microvascular injury with BBB breakdown, pericyte loss, basement membrane alteration and cerebral blood flow reduction in mice, in which BBB breakdown preceded gliosis. More importantly, mTBI accelerated BBB leakage, amyloid pathology and cognitive impairment in the 5xFAD mice. DISCUSSION: Our data demonstrated that microvascular injury plays a key role in the pathogenesis of AD after mTBI. Therefore, restoring vascular functions might be beneficial for patients with mTBI, and potentially reduce the risk of developing AD.


Subject(s)
Alzheimer Disease/pathology , Brain Concussion/pathology , Cognitive Dysfunction/pathology , Disease Progression , Microvessels/pathology , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Animals , Brain Concussion/complications , Brain Concussion/genetics , Cognitive Dysfunction/etiology , Cognitive Dysfunction/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
16.
Biochim Biophys Acta Mol Basis Dis ; 1867(4): 166078, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33444711

ABSTRACT

To shed light on the impact of systemic physiology on the pathology of traumatic brain injury (TBI), we examine the effects of TBI (concussive injury) and dietary fructose on critical aspects of lipid homeostasis in the brain and liver of young-adult rats. Lipids are integral components of brain structure and function, and the liver has a role on the synthesis and metabolism of lipids. Fructose is mainly metabolized in the liver with potential implications for brain function. Lipidomic analysis accompanied by unbiased sparse partial least squares discriminant analysis (sPLS-DA) identified lysophosphatidylcholine (LPC) and cholesterol ester (CE) as the top lipid families impacted by TBI and fructose in the hippocampus, and only LPC (16:0) was associated with hippocampal-dependent memory performance. Fructose and TBI elevated liver pro-inflammatory markers, interleukin-1α (IL-1α), Interferon-γ (IFN-γ) that correlated with hippocampal-dependent memory dysfunction, and monocyte chemoattractant protein-1 (MCP-1) positively correlated with LPC levels in the hippocampus. The effects of fructose were more pronounced in the liver, in agreement with the role of liver on fructose metabolism and suggest that fructose could exacerbate liver inflammation caused by TBI. The overall results indicate that TBI and fructose interact to influence systemic and central inflammation by engaging liver lipids. The impact of TBI and fructose diet on the periphery provides a therapeutic target to counteract the TBI pathogenesis.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Brain/physiopathology , Lipid Metabolism , Liver/physiopathology , Animals , Brain/metabolism , Brain Injuries, Traumatic/metabolism , Inflammation/metabolism , Inflammation/physiopathology , Liver/metabolism , Male , Rats , Rats, Sprague-Dawley
17.
Biochim Biophys Acta Mol Basis Dis ; 1867(5): 166036, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33508421

ABSTRACT

High consumption of fructose has paralleled an explosion in metabolic disorders including obesity and type 2 diabetes. Even more problematic, sustained consumption of fructose is perceived as a threat for brain function and development of neurological disorders. The action of fructose on peripheral organs is an excellent model to understand how systemic physiology impacts the brain. Given the recognized action of fructose on liver metabolism, here we discuss mechanisms by which fructose can impact the brain by interacting with liver and other organs. The interaction between peripheral and central mechanisms is a suitable target to reduce the pathophysiological consequences of neurological disorders.


Subject(s)
Brain Diseases/etiology , Dietary Sucrose/adverse effects , Fructose/adverse effects , Metabolic Diseases/pathology , Neuronal Plasticity , Brain Diseases/pathology , Humans , Metabolic Diseases/chemically induced , Nervous System Physiological Phenomena , Sweetening Agents/pharmacology
18.
Mol Psychiatry ; 26(1): 134-150, 2021 01.
Article in English | MEDLINE | ID: mdl-33144709

ABSTRACT

The field of nutritional psychiatry has generated observational and efficacy data supporting a role for healthy dietary patterns in depression onset and symptom management. To guide future clinical trials and targeted dietary therapies, this review provides an overview of what is currently known regarding underlying mechanisms of action by which diet may influence mental and brain health. The mechanisms of action associating diet with health outcomes are complex, multifaceted, interacting, and not restricted to any one biological pathway. Numerous pathways were identified through which diet could plausibly affect mental health. These include modulation of pathways involved in inflammation, oxidative stress, epigenetics, mitochondrial dysfunction, the gut microbiota, tryptophan-kynurenine metabolism, the HPA axis, neurogenesis and BDNF, epigenetics, and obesity. However, the nascent nature of the nutritional psychiatry field to date means that the existing literature identified in this review is largely comprised of preclinical animal studies. To fully identify and elucidate complex mechanisms of action, intervention studies that assess markers related to these pathways within clinically diagnosed human populations are needed.


Subject(s)
Depression/metabolism , Depression/physiopathology , Diet/psychology , Animals , Depression/genetics , Epigenesis, Genetic , Gastrointestinal Microbiome , Humans , Inflammation , Oxidative Stress
19.
Front Aging Neurosci ; 12: 560865, 2020.
Article in English | MEDLINE | ID: mdl-33024433

ABSTRACT

The loss of cognitive function in Alzheimer's disease is pathologically linked with neurofibrillary tangles, amyloid deposition, and loss of neuronal communication. Cerebral insulin resistance and mitochondrial dysfunction have emerged as important contributors to pathogenesis supporting our hypothesis that cerebral fructose metabolism is a key initiating pathway for Alzheimer's disease. Fructose is unique among nutrients because it activates a survival pathway to protect animals from starvation by lowering energy in cells in association with adenosine monophosphate degradation to uric acid. The fall in energy from fructose metabolism stimulates foraging and food intake while reducing energy and oxygen needs by decreasing mitochondrial function, stimulating glycolysis, and inducing insulin resistance. When fructose metabolism is overactivated systemically, such as from excessive fructose intake, this can lead to obesity and diabetes. Herein, we present evidence that Alzheimer's disease may be driven by overactivation of cerebral fructose metabolism, in which the source of fructose is largely from endogenous production in the brain. Thus, the reduction in mitochondrial energy production is hampered by neuronal glycolysis that is inadequate, resulting in progressive loss of cerebral energy levels required for neurons to remain functional and viable. In essence, we propose that Alzheimer's disease is a modern disease driven by changes in dietary lifestyle in which fructose can disrupt cerebral metabolism and neuronal function. Inhibition of intracerebral fructose metabolism could provide a novel way to prevent and treat this disease.

20.
Mol Nutr Food Res ; 64(23): e2000788, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33063454

ABSTRACT

SCOPE: The influence of docosahexaenoic acid (DHA) on cardiometabolic and cognitive phenotypes, and multi-omic alterations in the brain under two metabolic conditions is explored to understand context-specific nutritional effects. METHODS AND RESULTS: Rats are randomly assigned to a DHA-rich or a control chow diet while drinking water or high fructose solution, followed by profiling of metabolic and cognitive phenotypes and the transcriptome and DNA methylome of the hypothalamus and hippocampus. DHA reduces serum triglyceride and improves insulin resistance and memory exclusively in the fructose-consuming rats. In hippocampus, DHA affects genes related to synapse functions in the chow group but immune functions in the fructose group; in hypothalamus, DHA alters immune pathways in the chow group but metabolic pathways in the fructose group. Network modeling reveals context-specific regulators of DHA effects, including Klf4 and Dusp1 for chow condition and Lum, Fn1, and Col1a1 for fructose condition in hippocampus, as well as Cyr61, JunB, Ier2, and Pitx2 under chow condition and Hcar1, Cdh1, and Osr1 under fructose condition in hypothalamus. CONCLUSION: DHA exhibits differential influence on epigenetic loci, genes, pathways, and metabolic and cognitive phenotypes under different dietary contexts, supporting population stratification in DHA studies to achieve precision nutrition.

SELECTION OF CITATIONS
SEARCH DETAIL
...