Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Brain Behav Immun ; 120: 208-220, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38823430

ABSTRACT

Chemotherapy is notorious for causing behavioral side effects (e.g., cognitive decline). Notably, the gut microbiome has recently been reported to communicate with the brain to affect behavior, including cognition. Thus, the aim of this clinical longitudinal observational study was to determine whether chemotherapy-induced disruption of the gut microbial community structure relates to cognitive decline and circulating inflammatory signals. Fecal samples, blood, and cognitive measures were collected from 77 patients with breast cancer before, during, and after chemotherapy. Chemotherapy altered the gut microbiome community structure and increased circulating TNF-α. Both the chemotherapy-induced changes in microbial relative abundance and decreased microbial diversity were related to elevated circulating pro-inflammatory cytokines TNF-α and IL-6. Participants reported subjective cognitive decline during chemotherapy, which was not related to changes in the gut microbiome or inflammatory markers. In contrast, a decrease in overall objective cognition was related to a decrease in microbial diversity, independent of circulating cytokines. Stratification of subjects, via a reliable change index based on 4 objective cognitive tests, identified objective cognitive decline in 35% of the subjects. Based on a differential microbial abundance analysis, those characterized by cognitive decline had unique taxonomic shifts (Faecalibacterium, Bacteroides, Fusicatenibacter, Erysipelotrichaceae UCG-003, and Subdoligranulum) over chemotherapy treatment compared to those without cognitive decline. Taken together, gut microbiome change was associated with cognitive decline during chemotherapy, independent of chemotherapy-induced inflammation. These results suggest that microbiome-related strategies may be useful for predicting and preventing behavioral side effects of chemotherapy.

2.
J Neurochem ; 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37084026

ABSTRACT

It is poorly understood how solid peripheral tumors affect brain neuroimmune responses despite the various brain-mediated side effects and higher rates of infection reported in cancer patients. We hypothesized that chronic low-grade peripheral tumor-induced inflammation conditions microglia to drive suppression of neuroinflammatory responses to a subsequent peripheral immune challenge. Here, Balb/c murine mammary tumors attenuated the microglial inflammatory gene expression responses to lipopolysaccharide (LPS) and live Escherichia coli (E. coli) challenges and the fatigue response to an E. coli infection. In contrast, the inflammatory gene expression in response to LPS or a toll-like receptor 2 agonist of Percoll-enriched primary microglia cultures was comparable between tumor-bearing and -free mice, as were the neuroinflammatory and sickness behavioral responses to an intracerebroventricular interleukin (IL)-1ß injection. These data led to the hypothesis that Balb/c mammary tumors blunt the neuroinflammatory responses to an immune challenge via a mechanism involving tumor suppression of the peripheral humoral response. Balb/c mammary tumors modestly attenuated select circulating cytokine responses to LPS and E. coli challenges. Further, a second mammary tumor/mouse strain model (E0771 tumors in C57Bl/6 mice) displayed mildly elevated inflammatory responses to an immune challenge. Taken together, these data indicate that tumor-induced suppression of neuroinflammation and sickness behaviors may be driven by a blunted microglial phenotype, partly because of an attenuated peripheral signal to the brain, which may contribute to infection responses and behavioral side effects reported in cancer patients. Finally, these neuroimmune effects likely vary based on tumor type and/or host immune phenotype.

3.
Brain Behav Immun ; 95: 401-412, 2021 07.
Article in English | MEDLINE | ID: mdl-33895287

ABSTRACT

Chemotherapy treatment is associated with acute behavioral side effects (fatigue, anorexia) that significantly reduce patient quality of life and are dose-limiting, thereby increasing mortality (Kidwell et al., 2014). Disruptions to gut homeostasis (diarrhea, constipation, microbial dysbiosis) are also observed in patients receiving chemotherapy. In non-oncological patients, facets of mental health (fatigue, anxiety, depression) correlate with alterations in the gut microbiome, suggestive of a contribution of the gut in CNS disease etiology. The potential gut-to-brain pathway is poorly understood in patients receiving chemotherapy. Our prior studies have demonstrated a correlation between chemotherapy treatment, gut changes, peripheral and central inflammation, and behavioral symptoms in mice. Here we aimed to determine the extent to which chemotherapy-associated gut manipulations modulate the behavioral and biological consequences of chemotherapy. We measured sickness behaviors, peripheral and central inflammatory mediators, and anxiety in conventional or germ-free female mice: 1) cohabitating with mice of the opposite treatment group, 2) pre-treated with broad-spectrum antibiotics, or 3) given an intra-gastric gavage of gut content from chemotherapy-treated mice. In cohabitation studies, presumed coprophagia promoted body mass recovery, however strong associations with inflammation and behavior were not observed. Reduction of gut microbial alpha diversity via antibiotics did not prevent chemotherapy-associated side effects, however the relative abundances of the genera Tyzzerella, Romboutsia, and Turicibacter correlated with circulating inflammatory (IL-1ß) and behavioral outcomes (lethargy, anxiety-like behavior). A gut microbiota transplant from chemotherapy-treated mice decreased central locomotion in open field testing, increased circulating CXCL1, and increased hippocampal Il6 and Tnfa in germ-free mice compared to germ-free mice that received a transplant from vehicle-treated mice. Taken together, these data provide further evidence that the gut microbiota likely contributes to the development of chemotherapy-associated side effects. This work has significant implications in the future treatment of anxiety in patients, and warrants future studies using microbe-based treatment options.


Subject(s)
Antineoplastic Agents , Gastrointestinal Microbiome , Animals , Dysbiosis/chemically induced , Female , Humans , Inflammation , Mice , Quality of Life
4.
Sci Rep ; 9(1): 16490, 2019 11 11.
Article in English | MEDLINE | ID: mdl-31712703

ABSTRACT

Chemotherapy treatment negatively affects the nervous and immune systems and alters gastrointestinal function and microbial composition. Outside of the cancer field, alterations in commensal bacteria and immune function have been implicated in behavioral deficits; however, the extent to which intestinal changes are related to chemotherapy-associated behavioral comorbidities is not yet known. Thus, this study identified concurrent changes in behavior, central and peripheral immune activation, colon histology, and bacterial community structure in mice treated with paclitaxel chemotherapy. In paclitaxel-treated mice, increased fatigue and decreased cognitive performance occurred in parallel with reduced microglia immunoreactivity, increased circulating chemokine expression (CXCL1), as well as transient increases in pro-inflammatory cytokine/chemokine (Il-1ß, Tnfα, Il-6, and Cxcl1) gene expression in the brain. Furthermore, mice treated with paclitaxel had altered colonic bacterial community composition and increased crypt depth. Relative abundances of multiple bacterial taxa were associated with paclitaxel-induced increases in colon mass, spleen mass, and microglia activation. Although microbial community composition was not directly related to available brain or behavioral measures, structural differences in colonic tissue were strongly related to microglia activation in the dentate gyrus and the prefrontal cortex. These data indicate that the chemotherapeutic paclitaxel concurrently affects the gut microbiome, colonic tissue integrity, microglia activation, and fatigue in female mice, thus identifying a novel relationship between colonic tissue integrity and behavioral responses that is not often assessed in studies of the brain-gut-microbiota axis.


Subject(s)
Antineoplastic Agents/adverse effects , Bacteria/drug effects , Colon/drug effects , Dysbiosis/etiology , Gastrointestinal Microbiome/drug effects , Immune System/drug effects , Inflammation/etiology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers , Brain/drug effects , Brain/metabolism , Brain/pathology , Cognitive Dysfunction/etiology , Colon/metabolism , Colon/pathology , Cytokines/blood , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Fatigue/etiology , Female , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Illness Behavior , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Mice
5.
Neuroscience ; 175: 127-32, 2011 Feb 23.
Article in English | MEDLINE | ID: mdl-21145376

ABSTRACT

Adult mammalian brains are capable of some structural plasticity. Although the basic cellular mechanisms underlying learning and memory are being revealed, extrinsic factors contributing to this plasticity remain unspecified. White-footed mice (Peromyscus leucopus) are particularly well suited to investigate brain plasticity because they show marked seasonal changes in structure and function of the hippocampus induced by a distinct environmental signal, viz., photoperiod (i.e. the number of hours of light/day). Compared to animals maintained in 16 h of light/day, exposure to 8 h of light/day for 10 weeks induces several phenotypic changes in P. leucopus, including reduction in brain mass and hippocampal volume. To investigate the functional consequences of reduced hippocampal size, we examined the effects of photoperiod on spatial learning and memory in the Barnes maze, and on long-term potentiation (LTP) in the hippocampus, a leading candidate for a synaptic mechanism underlying spatial learning and memory in rodents. Exposure to short days for 10 weeks decreased LTP in the Schaffer collateral-CA1 pathway of the hippocampus and impaired spatial learning and memory ability in the Barnes maze. Taken together, these results demonstrate a functional change in the hippocampus in male white-footed mice induced by day length.


Subject(s)
Circadian Rhythm/physiology , Hippocampus/physiopathology , Learning/physiology , Long-Term Potentiation/physiology , Memory Disorders/physiopathology , Photoperiod , Animals , Atrophy , Hippocampus/pathology , Male , Maze Learning/physiology , Memory Disorders/etiology , Memory Disorders/pathology , Neural Pathways/pathology , Neural Pathways/physiopathology , Peromyscus , Photic Stimulation/adverse effects
6.
J Neuroendocrinol ; 21(12): 1007-14, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19840238

ABSTRACT

Seasonally breeding animals use a combination of photic (i.e. day length) and nonphotic (e.g. food availability, temperature) cues to regulate their reproduction. How these environmental cues are integrated is not understood. To assess the potential role of two candidate neuropeptides, kisspeptin and RFamide-related peptide-3 (RFRP), we monitored regional changes in their gene expression in a seasonally breeding mammal exposed to moderate changes in photoperiod and food availability. Adult male Siberian hamsters (Phodopus sungorus) were housed under a long (16 h light/day; 16 L) or intermediate (13.5 L) photoperiod and fed ad lib. or a progressive food restriction schedule (FR; reduced to 80% of ad lib.) for 11 weeks. Gonadal regression occurred only in FR hamsters housed under 13.5 L. Immunohistochemistry was used to identify diencephalic populations of kisspeptin- and RFRP-immunoreactive cells, and quantitative PCR was used to measure gene expression in adjacent coronal brain sections. Photoperiod, but not food availability, altered RFRP mRNA expression in the dorsomedial sections, whereas food availability but not photoperiod altered Kiss1 expression in the arcuate sections; intermediate photoperiods elevated RFRP expression, and food restriction suppressed Kiss1 expression. Regional- and neuropeptide-specific activity of RFamides may provide a mechanism for integration of multi-modal environmental information in the seasonal control of reproduction.


Subject(s)
Hypothalamus/metabolism , Light , Neuropeptides/metabolism , Seasons , Tumor Suppressor Proteins/metabolism , Animals , Cerebrum/physiology , Cricetinae , Cues , Diet , Food Deprivation/physiology , Gene Expression Regulation , Male , Neuropeptides/genetics , Organ Size , Phodopus , Photoperiod , RNA, Messenger/metabolism , Testis/anatomy & histology , Time Factors , Tumor Suppressor Proteins/genetics
7.
Neuroscience ; 131(4): 779-84, 2005.
Article in English | MEDLINE | ID: mdl-15749332

ABSTRACT

Rodents that live in changing environments display different immune responses mediated in part by photoperiod (day length) cues. Siberian hamsters maintained in winter-like (short) photoperiods display smaller physiological and behavioral responses to immune challenges as compared with hamsters housed in summer-like (long) photoperiods. We hypothesized that these different response patterns are attributable to altered cytokine production in the hypothalamus in response to photoperiod changes. Female hamsters were housed in long or short days for 10 weeks to induce photoperiodic alterations, then injected with either LPS (a bacterial endotoxin) or saline. Fever and food intake were assessed 3 h post-injection; hypothalami and blood were collected 3, 6, and 12 h post-injection. LPS induced lower fever and reduction in food intake responses in short-day hamsters as compared with long-day hamsters. Additionally, short-day hamsters reduced IL-1beta and Tnfalpha expression in the hypothalamus 6 h after LPS injection, as measured by quantitative RT-PCR. Plasma estradiol concentrations did not differ between long- and short-day hamsters. These data suggest that differences in cytokine production in the hypothalamus may underlie the photoperiod-induced differences in sickness responses, and that these changes are not mediated by estradiol.


Subject(s)
Cytokines/biosynthesis , Cytokines/genetics , Eating/drug effects , Fever/physiopathology , Hypothalamus/metabolism , Lipopolysaccharides/toxicity , Photoperiod , Animals , Body Weight/drug effects , Body Weight/physiology , Cricetinae , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Eating/physiology , Estradiol/metabolism , Female , Fever/chemically induced , Interleukin-1/biosynthesis , Organ Size/drug effects , Organ Size/physiology , Phodopus , RNA/biosynthesis , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...