RESUMO
Head trauma often impairs cognitive processes mediated within the prefrontal cortex (PFC), leading to impaired decision making and risk-taking behavior. Mild traumatic brain injury (mTBI) accounts for approximately 80â¯% of reported head injury cases. Most neurological symptoms of a single mTBI are transient; however, growing evidence suggests that repeated mTBI (rmTBI) results in more severe impairments that worsen with each subsequent injury. Although mTBI-induced disruption of risk/reward decision making has been characterized, the potential for rmTBI to exacerbate these effects and the neural mechanisms involved are unknown. Catecholamine neurotransmitters, dopamine (DA) and norepinephrine (NE), modulate PFC-mediated functions. Imbalances in catecholamine function have been associated with TBI and may underlie aberrant decision making. We used a closed head-controlled cortical impact (CH-CCI) model in rats to evaluate the effects of rmTBI on performance of a probabilistic discounting task of risk/reward decision making behavior and expression levels of catecholamine regulatory proteins within the PFC. RmTBI produced transient increases in risky choice preference in both male and female rats, with these effects persisting longer in females. Additionally, rmTBI increased expression of the catecholamine synthetic enzyme, tyrosine hydroxylase (TH), within the orbitofrontal (OFC) region of the PFC in females only. These results suggest females are more susceptible to rmTBI-induced disruption of risk/reward decision making behavior and dysregulation of catecholamine synthesis within the OFC. Together, using the CH-CCI model of rodent rmTBI to evaluate the effects of multiple insults on risk-taking behavior and PFC catecholamine regulation begins to differentiate how mTBI occurrences affect neuropathological outcomes across different sexes.
Assuntos
Concussão Encefálica , Comportamento de Escolha , Córtex Pré-Frontal , Assunção de Riscos , Caracteres Sexuais , Tirosina 3-Mono-Oxigenase , Animais , Córtex Pré-Frontal/metabolismo , Masculino , Tirosina 3-Mono-Oxigenase/metabolismo , Feminino , Concussão Encefálica/metabolismo , Concussão Encefálica/complicações , Concussão Encefálica/fisiopatologia , Comportamento de Escolha/fisiologia , Ratos Sprague-Dawley , Ratos , Recompensa , Modelos Animais de Doenças , Tomada de Decisões/fisiologiaRESUMO
There are no approved therapeutics for psychostimulant use and recurrence of psychostimulant use. However, in preclinical rodent models environmental enrichment can decrease psychostimulant self-administration of low unit doses and cue-induced amphetamine seeking. We have previously demonstrated that glutamate-dependent therapeutics are able to alter amphetamine seeking to amphetamine-associated cues only in enriched rats. In the current experiment, we will determine if enrichment can attenuate responding and cue-induced amphetamine seeking during extended access to a high dose of intravenous amphetamine. We will also determine if N-acetylcysteine (NAC), a glutamate dependent therapeutic, can attenuate amphetamine seeking in differentially reared rats. Female and male Sprague-Dawley rats were reared in enriched, isolated, or standard conditions from postnatal day 21-51. Rats were trained to self-administer intravenous amphetamine (0.1â¯mg/kg/infusion) during twelve 6-hour sessions. During the abstinence period, NAC (100â¯mg/kg) or saline was administered daily. Following a cue-induced amphetamine-seeking test, astrocyte densities within regions of the medial prefrontal cortex (mPFC) and nucleus accumbens (ACb) were quantified using immunohistochemistry. Environmental enrichment decreased responding for amphetamine and during the cue-induced amphetamine-seeking test. NAC did not attenuate cue-induced amphetamine seeking or alter astrocyte density. Across all groups, female rats self-administered less amphetamine but responded more during cue-induced amphetamine seeking than male rats. While amphetamine increased astrocyte densities within the ACb and mPFC, it did not alter mPFC astrocyte densities in female rats. The results suggest that enrichment can attenuate responding during extended access to a high dose of amphetamine and the associated cues. Sex alters amphetamine-induced changes to astrocyte densities in a regionally specific matter.
Assuntos
Acetilcisteína , Anfetamina , Estimulantes do Sistema Nervoso Central , Sinais (Psicologia) , Meio Ambiente , Ratos Sprague-Dawley , Autoadministração , Animais , Masculino , Feminino , Anfetamina/farmacologia , Anfetamina/administração & dosagem , Acetilcisteína/farmacologia , Acetilcisteína/administração & dosagem , Estimulantes do Sistema Nervoso Central/farmacologia , Estimulantes do Sistema Nervoso Central/administração & dosagem , Comportamento de Procura de Droga/efeitos dos fármacos , Comportamento de Procura de Droga/fisiologia , Ratos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Caracteres Sexuais , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismoRESUMO
The Four Core Genotypes (FCG) mouse model has become a valuable model to study the mechanistic basis for biological sex differences. This model allows discrimination between influences of gonadal sex (ovaries or testes) from those associated with genetic sex (presence of XX or XY chromosome complement). FCG mice have illuminated distinct effects of gonadal and chromosomal sex on traits ranging from brain structure and behavior to vulnerability to obesity, atherosclerosis, multiple sclerosis, Alzheimer's and other diseases. A recent study determined that the YSry- chromosome used in a specific line of C57BL/6J FCG mice harbors nine genes that have been duplicated from the X chromosome. This report raised concern that scores of publications that previously used the FCG model may therefore be flawed, but did not provide details regarding how studies can be evaluated for potential impact (or lack of impact) of the translocation. Here we (1) provide a practical description of the genetic translocation for researchers using the FCG model, (2) document that a majority of the studies cited in the recent report are unlikely to be affected by the translocation, (3) provide a scheme for interpreting data from studies with FCG mice harboring the YSry- translocation, and (4) delineate expression levels of the nine translocated genes across tissue/cell types as a filter for evaluating their potential involvement in specific phenotypes.
Assuntos
Genótipo , Translocação Genética , Animais , Masculino , Feminino , Camundongos , Caracteres Sexuais , Camundongos Endogâmicos C57BL , Modelos AnimaisRESUMO
OBJECTIVE: White matter hyperintensities (WMH) on brain MRI images are the most common feature of cerebral small vessel disease (CSVD). Studies have yielded divergent findings on the modifiable risk factors for WMH and WMH's impact on cognitive decline. Mounting evidence suggests sex differences in WMH burden and subsequent effects on cognition. Thus, we aimed to identify sex-specific modifiable risk factors for WMH. We then explored whether there were sex-specific associations of WMH to longitudinal clinical dementia outcomes. METHODS: Participants aged 49-89 years were recruited at memory clinics and underwent a T2-weighted fluid-attenuated inversion recovery (FLAIR) 3T MRI scan to measure WMH volume. Participants were then recruited for two additional follow-up visits, 1-2 years apart, where clinical dementia rating sum of boxes (CDR-SB) scores were measured. We first explored which known modifiable risk factors for WMH were significant when tested for a sex-interaction effect. We additionally tested which risk factors were significant when stratified by sex. We then tested to see whether WMH is longitudinally associated with clinical dementia that is sex-specific. RESULTS: The study utilized data from 713 participants (241 males, 472 females) with a mean age of 72.3 years and 72.8 years for males and females, respectively. 57.3% and 59.5% of participants were diagnosed with mild cognitive impairment (MCI) for males and females, respectively. 40.7% and 39.4% were diagnosed with dementia for males and females, respectively. Of the 713 participants, 181 participants had CDR-SB scores available for three longitudinal time points. Compared to males, females showed stronger association of age to WMH volume. Type 2 Diabetes was associated with greater WMH burden in females but not males. Finally, baseline WMH burden was associated with worse clinical dementia outcomes longitudinally in females but not in males. DISCUSSION: Older females have an accelerated increase in cerebrovascular burden as they age, and subsequently are more vulnerable to clinical dementia decline due to CSVD. Additionally, females are more susceptible to the cerebrovascular consequences of diabetes. These findings emphasize the importance of considering sex when examining the consequences of CSVD. Future research should explore the underlying mechanisms driving these sex differences and personalized prevention and treatment strategies. CLINICAL TRIAL REGISTRATION: The BICWALZS is registered in the Korean National Clinical Trial Registry (Clinical Research Information Service; identifier, KCT0003391). Registration Date 2018/12/14.
Assuntos
Demência , Imageamento por Ressonância Magnética , Substância Branca , Humanos , Masculino , Feminino , Idoso , República da Coreia/epidemiologia , Pessoa de Meia-Idade , Fatores de Risco , Substância Branca/diagnóstico por imagem , Substância Branca/patologia , Idoso de 80 Anos ou mais , Demência/diagnóstico por imagem , Demência/epidemiologia , Estudos de Coortes , Fatores Sexuais , Doenças de Pequenos Vasos Cerebrais/diagnóstico por imagem , Disfunção Cognitiva/diagnóstico por imagem , Caracteres Sexuais , Estudos LongitudinaisRESUMO
BACKGROUND: Aging is a complex process that involves all tissues in an organism and shows sex dimorphism. While transcriptional changes in aging have been well characterized, the majority of studies have focused on a single sex and sex differences in gene expression in aging are poorly understood. In this study, we explore sex dimorphism in gene expression in aging mice across three tissues. METHODS: We collected gastrocnemius muscle, liver and white adipose tissue from young (6 months, n = 14) and old (24 months, n = 14) female and male C57BL/6NIA mice and performed RNA-seq. To investigate sex dimorphism in aging, we considered two levels of comparisons: (a) differentially expressed genes between females and males in the old age group and (b) comparisons between females and males across the aging process. We utilized differential expression analysis and gene feature selection to investigate candidate genes. Gene set enrichment analysis was performed to identify candidate molecular pathways. Furthermore, we performed a co-expression network analysis and chose the gene module(s) associated with aging independent of sex or tissue-type. RESULTS: We identified both tissue-specific and tissue-independent genes associated with sex dimorphism in aged mice. Unique differentially expressed genes between old males and females across tissues were mainly enriched for pathways related to specific tissue function. We found similar results when exploring sex differences in the aging process, with the exception that in the liver genes enriched for lipid metabolism and digestive system were identified in both females and males. Combining enriched pathways across analyses, we identified amino acid metabolism, digestive system, and lipid metabolism as the core mechanisms of sex dimorphism in aging. Although the vast majority of age-related genes were sex and tissue specific, we identified 127 hub genes contributing to aging independent of sex and tissue that were enriched for the immune system and signal transduction. CONCLUSIONS: There are clear sex differences in gene expression in aging across liver, muscle and white adipose. Core pathways, including amino acid metabolism, digestive system and lipid metabolism, contribute to sex differences in aging.
Aging is a complex process that occurs differently across tissues, and in men compared to women. However, the mechanisms that cause sex differences are not well understood. Using naturally aging mouse models we compared how specific genes were differently expressed in muscle, liver and fat of old and young female and male mice. We found that the vast majority of genes that were changed with age were only changed in one sex and specific tissues. Overall, sex differences in aging across tissues were related to genes involved in amino acid metabolism, digestive system and lipid metabolism. Notably, lipid metabolism is important in aging females across all tissues. We also identified a set of genes associated with aging independent of sex and tissue-type involved in immune pathways and signaling. These results enhance our understanding of sex differences in aging.
Assuntos
Envelhecimento , Fígado , Camundongos Endogâmicos C57BL , Músculo Esquelético , Especificidade de Órgãos , Caracteres Sexuais , Animais , Envelhecimento/genética , Feminino , Masculino , Fígado/metabolismo , Músculo Esquelético/metabolismo , Camundongos , Tecido Adiposo Branco/metabolismo , Regulação da Expressão GênicaRESUMO
BACKGROUND: Chronic cigarette smokers report withdrawal symptomology, including affective dysfunction and cognitive deficits. While there are studies demonstrating sex specific withdrawal symptomology in nicotine-dependent individuals, literature examining the underlying biological mediators of this is scant and not in complete agreement. Therefore, in this study, we evaluated the sex specific effects of nicotine and withdrawal on contextual fear memory, a hippocampally dependent aspect of cognition that is disrupted in nicotine withdrawal. METHODS: Male and female B6/129F1 mice (8-13 weeks old) were used in all experiments. For the acute nicotine experiment, mice received intraperitoneal saline or nicotine (0.5 mg/kg) prior to contextual fear conditioning and test. For the chronic nicotine experiment, mice received nicotine (18 mg/kg/day) or saline for 11 days, then underwent contextual fear conditioning and test. Following the test, mice underwent minipump removal to elicit withdrawal or sham surgery, followed by the fear extinction assay. Bulk cortical tissue was used to determine nicotinic acetylcholine receptor levels via single point [3H]Epibatidine binding assay. Gene expression levels in the dorsal and ventral hippocampus were quantified via RT-PCR. RESULTS: We found that female mice had a stronger expression of contextual fear memory than their male counterparts. Further, following acute nicotine treatment, male, but not female, subjects demonstrated augmented contextual fear memory expression. In contrast, no significant effects of chronic nicotine treatment on fear conditioning were observed in either sex. When examining extinction of fear learning, we observed that female mice withdrawn from nicotine displayed impaired extinction learning, but no effect was observed in males. Nicotine withdrawal caused similar suppression of fosb, cfos, and bdnf, our proxy for neuronal activation and plasticity changes, in the dorsal and ventral hippocampus of both sexes. Additionally, we found that ventral hippocampus erbb4 expression, a gene implicated in smoking cessation outcomes, was elevated in both sexes following nicotine withdrawal. CONCLUSIONS: Despite the similar impacts of nicotine withdrawal on gene expression levels, fosb, cfos, bdnf and erbb4 levels in the ventral hippocampus were predictive of delays in female extinction learning alone. This suggests sex specific dysfunction in hippocampal circuitry may contribute to female specific nicotine withdrawal induced deficits in extinction learning.
Smokers undergoing nicotine withdrawal report increased feelings of anxiety, depression, and cognitive deficits. However, there are sex differences in these symptoms, with women reporting higher feelings of anxiety compared to men and men having worse cognitive deficits than women. The mechanisms underlying these sex differences in nicotine withdrawal symptoms are not well understood. The hippocampus is a brain region highly implicated in both the cognitive and anxiety-like symptoms of nicotine withdrawal. Therefore, we evaluated the effects of nicotine and withdrawal on contextual fear memory, a hippocampally dependent learning and memory task, in male and female mice. We found that female mice had a stronger contextual fear memory expression than their male counterparts. However, following acute nicotine treatment male mice had enhanced contextual fear memory compared to non-nicotine treated males, while acute nicotine had no impact on female mice. When examining extinction of contextual fear, we found female mice withdrawn from nicotine displayed impaired extinction learning, but no effect was observed in males. The female specific deficits in extinction learning due to nicotine withdrawal were correlated to hippocampal gene expression related to neuronal activity. This suggests hippocampal dysfunction may be driving the female specific nicotine withdrawal induced deficits in extinction learning.
Assuntos
Extinção Psicológica , Medo , Hipocampo , Nicotina , Caracteres Sexuais , Animais , Medo/efeitos dos fármacos , Nicotina/farmacologia , Nicotina/administração & dosagem , Feminino , Masculino , Extinção Psicológica/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Camundongos , Receptores Nicotínicos/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Agonistas Nicotínicos/farmacologia , Agonistas Nicotínicos/administração & dosagem , Condicionamento Psicológico/efeitos dos fármacosRESUMO
Systemic lupus erythematosus (SLE or lupus) is an immune-mediated disease associated with substantial medical burden. Notably, lupus exhibits a striking female bias, with women having significantly higher susceptibility compared to men, up to 14-fold higher in some ethnicities. Supernumerary X chromosome syndromes, like Klinefelter (XXY) and Triple X syndrome (XXX), also present higher SLE prevalence, whereas Turner syndrome (XO) displays lower prevalence. Taken together, SLE prevalence in different X chromosome dosage sceneries denotes a relationship between the number of X chromosomes and the risk of developing lupus. The dosage of X-linked genes, many of which play roles in the immune system, is compensated between males and females through the inactivation of one of the two X chromosomes in female cells. X-chromosome inactivation (XCI) initiates early in development with a random selection of which X chromosome to inactivate, a choice that is then epigenetically maintained in the daughter cells. This process is regulated by the X-Inactive-Specific Transcript (XIST), encoding for a long non-coding RNA, exclusively expressed from the inactive X chromosome (Xi). XIST interacts with various RNA binding proteins and chromatin modifiers to form a ribonucleoprotein (RNP) complex responsible for the transcriptional silencing and heterochromatinization of the Xi. This ensures stable silencing of most genes on the X chromosome, with only a few genes able to escape this process. Recent findings suggest that the molecular components involved in XCI, or their dysregulation, contribute to the pathogenesis of lupus. Indeed, nonrandom XCI, elevated gene escape from XCI, and the autoimmune potential of the XIST RNP complex have been suggested to contribute to auto-immune diseases, such as lupus. This review examines these current hypotheses concerning how this dosage compensation mechanism might impact the development of lupus, shedding light on potential mechanisms underlying the pathogenesis of the disease.
Lupus is a disease where the immune system mistakenly attacks the body's own tissues, leading to a range of complicated health issues. Interestingly, lupus is much more common in women (XX) than in men (XY), with women being up to 14 times more likely to develop the condition. Additionally, some genetic conditions involving extra or missing X chromosomes can also affect the chances of getting lupus: Klinefelter (XXY) and Triple X (XXX) syndromes show higher rates of lupus, while conditions like Turner syndrome (XO) have a lower risk. This suggests a link between the number of X chromosomes and the likelihood of developing the disease.In female cells, a process called X-chromosome inactivation (XCI) occurs, where one of the two X chromosomes in each cell is switched off to equalize X chromosome dosage with males. This process is regulated by a gene called XIST, which produces a long non-coding RNA. XIST helps to form a complex of RNA and proteins that silence the inactive X chromosome (Xi), ensuring stable gene expression patterns.Recent research suggests that molecular components or problems with this process might be linked to lupus. This review focuses on three hypotheses in which XCI or its dysregulation could impact lupus: nonrandom XCI, incomplete silencing of certain genes on the Xi, and the potential for the XIST ribonucleoprotein complex to activate the immune system. By investigating these mechanisms, researchers aim to better understand how variations in XCI mechanisms contribute to the development of lupus.
Assuntos
Cromossomos Humanos X , Lúpus Eritematoso Sistêmico , Lúpus Eritematoso Sistêmico/genética , Humanos , Feminino , Cromossomos Humanos X/genética , Animais , Inativação do Cromossomo X , Caracteres Sexuais , Masculino , RNA Longo não Codificante/genéticaRESUMO
Introduction: Reducing Optic Atrophy 1 (OPA1) expression in skeletal muscle in male mice induces Activation Transcription Factor 4 (ATF4) and the integrated stress response (ISR). Additionally, skeletal muscle secretion of Fibroblast Growth Factor 21 (FGF21) is increased, which mediates metabolic adaptations including resistance to diet-induced obesity (DIO) and glucose intolerance in these mice. Although FGF21 induction in this model can be reversed with pharmacological attenuation of ER stress, it remains to be determined if ATF4 is responsible for FGF21 induction and its metabolic benefits in this model. Methods: We generated mice with homozygous floxed Opa1 and Atf4 alleles and a tamoxifen-inducible Cre transgene controlled by the human skeletal actin promoter to enable simultaneous depletion of OPA1 and ATF4 in skeletal muscle (mAO DKO). Mice were fed high fat (HFD) or control diet and evaluated for ISR activation, body mass, fat mass, glucose tolerance, insulin tolerance and circulating concentrations of FGF21 and growth differentiation factor 15 (GDF15). Results: In mAO DKO mice, ATF4 induction is absent. Other indices of ISR activation, including XBP1s, ATF6, and CHOP were induced in mAO DKO males, but not in mOPA1 or mAO DKO females. Resistance to diet-induced obesity was not reversed in mAO DKO mice of both sexes. Circulating FGF21 and GDF15 illustrated sexually dimorphic patterns. Loss of OPA1 in skeletal muscle increases circulating FGF21 in mOPA1 males, but not in mOPA1 females. Additional loss of ATF4 decreased circulating FGF21 in mAO DKO male mice, but increased circulating FGF21 in female mAO DKO mice. Conversely, circulating GDF15 was increased in mAO DKO males and mOPA1 females, but not in mAO DKO females. Conclusion: Sex differences exist in the transcriptional outputs of the ISR following OPA deletion in skeletal muscle. Deletion of ATF4 in male and female OPA1 KO mice does not reverse the resistance to DIO. Induction of circulating FGF21 is ATF4 dependent in males, whereas induction of circulating GDF15 is ATF4 dependent in females. Elevated GDF15 in males and FGF21 in females could reflect activation by other transcriptional outputs of the ISR, that maintain mitokine-dependent metabolic protection in an ATF4-independent manner.
Assuntos
Fator 4 Ativador da Transcrição , Fatores de Crescimento de Fibroblastos , GTP Fosfo-Hidrolases , Camundongos Knockout , Músculo Esquelético , Caracteres Sexuais , Animais , Fator 4 Ativador da Transcrição/metabolismo , Fator 4 Ativador da Transcrição/genética , Camundongos , Masculino , Músculo Esquelético/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Feminino , GTP Fosfo-Hidrolases/metabolismo , GTP Fosfo-Hidrolases/genética , Fator 15 de Diferenciação de Crescimento/metabolismo , Fator 15 de Diferenciação de Crescimento/genética , Obesidade/metabolismo , Obesidade/genética , Dieta Hiperlipídica , Camundongos Endogâmicos C57BLRESUMO
The mammalian order Primates is known for widespread sexual dimorphism in size and phenotype. Despite repeated speculation that primate sexual size dimorphism either facilitates or is in part driven by functional differences in how males and females interact with their environments, few studies have directly assessed the influence of sexual dimorphism on performance traits. Here, we use a theoretical morphology framework to show that sexual dimorphism in primate crania is associated with divergent biomechanical performance traits. The degree of dimorphism is a significant covariate in biomechanical trait divergence between sexes. Males exhibit less efficient but stiffer cranial shapes and significant evolutionary allometry in biomechanical performance, whereas females maintain performance stability across their size spectrum. Evolutionary rates are elevated for efficiency in females whereas males emphasize size-dependent cranial stiffness. These findings support a hypothesis of sex-linked bifurcation in masticatory system performance: larger male crania and faster size evolution partially compensate for low efficiency and reflect a de-emphasis of mechanical leverage, whereas female crania maintain higher mechanical efficiency overall and evolve more rapidly in molar-based masticatory performance. The evolutionary checks-and-balances between size dimorphism and cranial mechanical performance may be a more important driver of primate phenotypic evolution than has been hitherto appreciated.
Assuntos
Evolução Biológica , Primatas , Caracteres Sexuais , Crânio , Animais , Feminino , Crânio/anatomia & histologia , Crânio/fisiologia , Masculino , Fenômenos Biomecânicos , Primatas/anatomia & histologia , Primatas/fisiologiaRESUMO
Pork primal weight and primal yield are important indicators for pig breeding, feeding management, commercial distribution systems, and meat processing. Here, we aimed to determine whether primal weight and primal yield could be predicted through non-destructive measurements of pork carcass traits. A total of 4397 carcasses (1958 gilts and 2439 barrows) from eight major meat processing centers were used, and the mean primal weight and primal yield were 56.0 kg and 73.9%, respectively. Significant sex differences were observed for all primal and carcass traits (P < 0.001), except for carcass weight. A maximum of 12 variables were examined, and primal weight was predicted with very high accuracy (R = 0.95, RMSE = 1.7, RPD = 3.0) using four variables. Primal yield was predicted with relatively good accuracy (R = 0.71, RMSE = 2.3, RPD = 1.4) using three variables, and these same variables were also effective for predicting primal weight. These prediction formulas were sufficiently accurate without accounting for the effect of sex. Overall, our results demonstrate that primal weight and primal yield can be accurately predicted using four variables, "carcass weight," "backfat thickness above M. gluteus medius," "spinous process length of 13th thoracic vertebra," and "length from 1st thoracic vertebra to backfat," without accounting for the effect of sex.
Assuntos
Peso Corporal , Animais , Masculino , Feminino , Suínos , Japão , Carne de Porco/análise , Caracteres Sexuais , Carne Vermelha/análise , Manipulação de Alimentos/métodos , Fatores Sexuais , Carne/análiseRESUMO
BACKGROUND: The sex difference in athletic performance has been thoroughly investigated in single sport disciplines such as swimming, cycling, and running. In contrast, only small samples of long-distance triathlons, such as the IRONMAN® triathlon, have been investigated so far. AIM: The aim of the study was to examine potential sex differences in the three split disciplines by age groups in 5-year intervals in a very large data set of IRONMAN® age group triathletes. METHODS: Data from 687,696 (553,608 men and 134,088 women) IRONMAN® age group triathletes (in 5-year intervals from 18-24 to 75+ years) finishing successfully between 2002 and 2022 an official IRONMAN® race worldwide were analyzed. The differences in performance between women and men were determined for each split discipline and for the overall race distance. RESULTS: Most finishers were in the age group 40-44 years. The fastest women were in the age group 25-29 years, and the fastest men were in the age group 30-34 years. For all split disciplines and overall race time, men were always faster than women in all groups. The performance difference between the sexes was more pronounced in cycling compared to swimming and running. From the age group 35-39 years until 60-64 years, the sex differences were nearly identical in swimming and running. For both women and men, the smallest sex difference was least significant in age group 18-24 years for all split disciplines and increased in a U-shaped manner until age group 70-74 years. For age groups 75 years and older, the sex difference decreased in swimming and cycling but increased in running. Considering the different characteristics of the race courses, the smallest performance gaps between men and women were found in river swimming, flat surface cycling and rolling running courses. CONCLUSIONS: The sex difference in the IRONMAN® triathlon was least significant in age group 18-24 years for all split disciplines and increased in a U-shaped manner until age group 70-74 years. For 75 years and older, the sex difference decreased in swimming and cycling but increased in running.
Assuntos
Atletas , Desempenho Atlético , Ciclismo , Corrida , Natação , Humanos , Feminino , Masculino , Pessoa de Meia-Idade , Adulto , Idoso , Natação/fisiologia , Corrida/fisiologia , Desempenho Atlético/fisiologia , Ciclismo/fisiologia , Adolescente , Adulto Jovem , Fatores Etários , Fatores Sexuais , Caracteres SexuaisRESUMO
Background: Hypertension and hypertensive disorders of pregnancy (HDP) cause a significant burden of disease on societies and individuals by increasing cardiovascular disease risk. Environmental risk factors alone do not explain the observed sexual dimorphism in lifetime blood pressure (BP) trajectories nor inter-individual variation in HDP risk.Methods: In this short review, we focus on the genetics of hypertension-related sex differences and HDP and discuss the importance of genetics utilization for sex-specific hypertension risk prediction.Results: Population and twin studies estimate that 28-66% of variation in BP levels and HDP is explained by genetic variation, while genomic wide association studies suggest that BP traits and HDP partly share a common genetic background. Moreover, environmental and epigenetic regulation of these genes differ by sex and oestrogen receptors in particular are shown to convey cardio- and vasculoprotective effects through epigenetic regulation of DNA. The majority of known genetic variation in hypertension and HDP is polygenic. Polygenic risk scores for BP display stronger associations with hypertension risk in women than in men and are associated with sex-specific age of hypertension onset. Monogenic forms of hypertension are rare and mostly present equally in both sexes.Conclusion: Despite recent genetic discoveries providing new insights into HDP and sex differences in BP traits, further research is needed to elucidate the underlying biology. Emphasis should be placed on demonstrating the added clinical value of these genetic discoveries, which may eventually facilitate genomics-based personalized treatments for hypertension and HDP.
Blood pressure trajectories and age-related prevalence of hypertension differ between men and women.Vasculoprotective qualities of oestrogen, mediated by oestrogen receptors, are lost at menopause.Certain genetic variants linked to hypertension are sex-specific, i.e. only observed in one sex.Hypertension and preeclampsia are polygenic diseases, i.e. several different genes influence the susceptibility for these conditions.Hypertension and hypertensive disorders of pregnancy have a partially shared genetic basis.The combined effect of several genetic variants can be evaluated using polygenic risk scores (PRSs). Individuals, and especially women, with a high blood pressure PRS develop hypertension a decade earlier compared to those with a low PRS.Environmental and lifestyle factors can modify genetic risk by altering gene expression. Sex differences in the association between gene expression and hypertension remain largely unknown.
Assuntos
Hipertensão Induzida pela Gravidez , Hipertensão , Caracteres Sexuais , Humanos , Feminino , Gravidez , Hipertensão Induzida pela Gravidez/genética , Masculino , Hipertensão/genética , Hipertensão/fisiopatologia , Epigênese Genética , Estudo de Associação Genômica Ampla , Fatores de Risco , Pressão Sanguínea/genética , Fatores SexuaisRESUMO
The prospect that the ventromedial hypothalamic nucleus (VMN) transcription factor steroidogenic factor-1/NR5A1 (SF-1) may exert sex-dimorphic control of glucose counterregulation is unresolved. Recent studies in male rats show that SF-1 regulates transcription of co-expressed hypoglycemia-sensitive neurochemicals in dorsomedial VMN growth hormone-releasing hormone (Ghrh) neurons. Gene knockdown and laser-catapult-microdissection/single-cell multiplex qPCR techniques were used here in a female rat model to determine if SF-1 control of Ghrh neuron transmitter marker, energy sensor, and estrogen receptor (ER) variant mRNAs varies according to sex. Data show that in females, hypoglycemia elicits a gain of SF-1 inhibitory control of VMNdm Ghrh neuron Ghrh and Ghrh-receptor gene profiles and loss of augmentation of glutaminase transcription; SF-1 gene silencing diminished eu- and hypoglycemic patterns of neuronal nitric oxide gene transcription. SF-1 imposes divergent control of baseline and hypoglycemic glutamate decarboxylase65 (GAD)-1 (stimulatory) versus GAD2 (inhibitory) mRNAs in that sex. SF-1 stimulates baseline VMNdm Ghrh neuron PRKAA1/AMPKα1 and PRKAA2/AMPKα2 gene expression, yet causes opposite changes in these gene profiles during hypoglycemia. SF-1 exerts glucose-dependent control of ER-alpha and G-protein-coupled ER-1 transcription, but blunts ER-beta gene profiles during eu- and hypoglycemia. In females, SF-1 knockdown did not affect hypercorticosteronemia or hyperglucagonemia, but blunted hypoglycemic suppression of growth hormone secretion. Results show that SF-1 expression is critical for female rat VMNdm Ghrh neuron counterregulatory neurochemical, AMPK catalytic subunit, and ER gene transcription responses to hypoglycemia. Sex differences in direction of SF-1 control of distinctive gene profiles may result in observed disparities in SF-1 regulation of counterregulatory hormone secretion between sexes.
Assuntos
Hormônio Liberador de Hormônio do Crescimento , Neurônios , Fator Esteroidogênico 1 , Núcleo Hipotalâmico Ventromedial , Animais , Feminino , Masculino , Ratos , Glutamato Descarboxilase/metabolismo , Glutamato Descarboxilase/genética , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hormônio Liberador de Hormônio do Crescimento/genética , Hormônio Liberador de Hormônio do Crescimento/biossíntese , Hipoglicemia/metabolismo , Neurônios/metabolismo , Ratos Sprague-Dawley , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/biossíntese , Caracteres Sexuais , Fator Esteroidogênico 1/metabolismo , Fator Esteroidogênico 1/genética , Núcleo Hipotalâmico Ventromedial/metabolismoRESUMO
BACKGROUND: Adverse events in early life can have impact lasting into adulthood. We investigated the long-term effects of systemic inflammation during postnatal development on adult microglial responses to lipopolysaccharide (LPS) in two CNS regions (cortex, cervical spinal cord) in male and female rats. METHODS: Inflammation was induced in Sprague-Dawley rats by LPS (1 mg/kg) administered intraperitoneally during postnatal development at P7, P12 or P18. As adults (12 weeks of age), the rats received a second LPS dose (1 mg/kg). Control rats received saline. Microglia were isolated 3 h post-LPS followed by gene expression analysis via qRT-PCR for pro-inflammatory (IL-6, iNOS, Ptgs2, C/EBPb, CD14, CXCL10), anti-inflammatory (CD68, Arg-1), and homeostatic genes (P2Y12, Tmemm119). CSF-1 and CX3CL1 mRNAs were analyzed in microglia-free homogenates. RESULTS: Basal gene expression in adult microglia was largely unaffected by postnatal inflammation. Adult cortical microglial pro-inflammatory gene responses to LPS were either unchanged or attenuated in rats exposed to LPS during postnatal development. Ptgs2, C/EBPb, CXCL10 and Arg-1 were the most affected genes, with expression significantly downregulated vs. rats without postnatal LPS. Spinal microglia were affected most by LPS at P18, with mixed and sometimes opposing effects on proinflammatory genes in males vs. females. Overall, male cortical vs. spinal microglia were more affected by postnatal LPS. Females were affected in both cortex and spinal cord, but the effect was dependent on timing of postnatal LPS. Overall, inflammatory challenge at P18 had greater effect on adult microglia vs. challenge at P12 or P7. CONCLUSIONS: Long-lasting effects of postnatal inflammation on adult microglia depend on postnatal timing, CNS region and sex.
Assuntos
Animais Recém-Nascidos , Inflamação , Lipopolissacarídeos , Microglia , Ratos Sprague-Dawley , Caracteres Sexuais , Animais , Microglia/metabolismo , Microglia/efeitos dos fármacos , Feminino , Ratos , Masculino , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Fatores Etários , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/efeitos dos fármacosRESUMO
G protein-gated inwardly rectifying K+ (GIRK) channels mediate the postsynaptic inhibitory effect of many neurotransmitters in the hippocampus and are implicated in neurological disorders characterized by cognitive deficits. Here, we show that enhancement or suppression of GIRK channel activity in dorsal CA1 pyramidal neurons disrupted novel object recognition in mice, without impacting open field activity or avoidance behavior. Contextual fear learning was also unaffected, but extinction of contextual fear was disrupted by suppression of GIRK channel activity in male mice. Thus, the strength of GIRK channel activity in dorsal CA1 pyramidal neurons regulates select cognitive task performance in mice.
Assuntos
Região CA1 Hipocampal , Medo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Células Piramidais , Animais , Masculino , Células Piramidais/fisiologia , Células Piramidais/metabolismo , Região CA1 Hipocampal/fisiologia , Região CA1 Hipocampal/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Medo/fisiologia , Feminino , Reconhecimento Psicológico/fisiologia , Aprendizagem da Esquiva/fisiologia , Camundongos Endogâmicos C57BL , Camundongos , Caracteres Sexuais , Extinção Psicológica/fisiologia , Memória/fisiologia , Aprendizagem/fisiologia , Comportamento Exploratório/fisiologiaRESUMO
Many complex diseases exhibit pronounced sex differences that can affect both the initial risk of developing the disease, as well as clinical disease symptoms, molecular manifestations, disease progression, and the risk of developing comorbidities. Despite this, computational studies of molecular data for complex diseases often treat sex as a confounding variable, aiming to filter out sex-specific effects rather than attempting to interpret them. A more systematic, in-depth exploration of sex-specific disease mechanisms could significantly improve our understanding of pathological and protective processes with sex-dependent profiles. This survey discusses dedicated bioinformatics approaches for the study of molecular sex differences in complex diseases. It highlights that, beyond classical statistical methods, approaches are needed that integrate prior knowledge of relevant hormone signaling interactions, gene regulatory networks, and sex linkage of genes to provide a mechanistic interpretation of sex-dependent alterations in disease. The review examines and compares the advantages, pitfalls and limitations of various conventional statistical and systems-level mechanistic analyses for this purpose, including tailored pathway and network analysis techniques. Overall, this survey highlights the potential of specialized bioinformatics techniques to systematically investigate molecular sex differences in complex diseases, to inform biomarker signature modeling, and to guide more personalized treatment approaches.
Assuntos
Biologia Computacional , Caracteres Sexuais , Humanos , Biologia Computacional/métodos , Masculino , Feminino , Redes Reguladoras de GenesRESUMO
Ulcerative colitis is a chronic pathology characterized by relapsing-remitting phases of intestinal inflammation. Additionally, some patients develop neuropsychiatric disorders, such as depression and anxiety, or cognitive deficits. We aimed to investigate whether the development of chronic colitis elicits memory, locomotion, and mood impairments. It further examined whether these impairments are influenced by the relapsing-remitting phases of the colitis or by sex. Here, we used a chronic colitis model in male and female rats, induced with sodium dextran sulfate, mirroring the phases of human ulcerative colitis. Our results revealed that the severity of colitis was slightly higher in males than females. Chronic colitis triggered motor and short-term memory deficits and induced anxiety- and depression-like behaviors that remained throughout the development of the disease. There are also sex differences under control or inflammatory conditions. Therefore, in both situations, females compared to males displayed: (i) slightly lower locomotion, (ii) increased anxiety-like behaviors, (iii) similar depression-like behaviors, and (iv) similar short-term memory deficit. Additionally, under control conditions, the mRNA levels of IL-1ß, IL-6, and TNF-α were higher in the female hippocampus. In both sexes, when chronic colitis was established, the neuroinflammation was evidenced by increased mRNA levels of these three cytokines in the hippocampus and in the motor and prefrontal cortices. Interestingly, this neuroinflammation was slightly greater in males. In summary, we show that the development of chronic colitis caused persistent behavioral abnormalities, highlighting sex differences, and that could be a consequence, at least in part, of the increase in IL-1ß, IL-6, and TNF-α in the brain.
Assuntos
Doenças Neuroinflamatórias , Animais , Masculino , Feminino , Ratos , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/etiologia , Transtornos da Memória/etiologia , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Ansiedade , Doença Crônica , Depressão/metabolismo , Depressão/etiologia , Sulfato de Dextrana/toxicidade , Hipocampo/metabolismo , Hipocampo/patologia , Ratos Wistar , Afeto , Inflamação/metabolismo , Colite Ulcerativa/metabolismo , Colite Ulcerativa/psicologia , Fatores Sexuais , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/genética , Caracteres SexuaisRESUMO
In the last few years, immune checkpoint inhibitors (ICIs) improved treatment strategies for advanced non-small cell lung cancer (NSCLC) with no targetable driver mutations. Empirical evidence strongly suggests that males and females differ in outcomes following the use of ICIs for treatments of solid cancers. Women in fact exhibit greater humoral and cell-mediated immune responses and an even more advanced immune editing which plays an important role in controlling cancer rising and evolution. However, at present, no conclusive studies have addressed differences in response to ICIs regarding sex and, to note, reproductive status in women or autoimmune diseases in both sexes are often not recorded in clinical trials. Consequently, it can be argued that to assess cancer responses and study cancer spread, results of published studies in men may not unconditionally be applied on female patients treated with ICIs, and vice versa. In this chapter have been discussed recent data about gender differences in the immune system and in NSCLC patients treated with ICIs, highlighting sex as a key factor in evaluating different responses in the two sexes.
Assuntos
Carcinoma Pulmonar de Células não Pequenas , Imunoterapia , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/terapia , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/genética , Feminino , Masculino , Resultado do Tratamento , Caracteres Sexuais , Inibidores de Checkpoint Imunológico/uso terapêutico , Fatores SexuaisRESUMO
Explaining the evolution of sex differences in cooperation remains a major challenge. Comparative studies highlight that offspring of the more philopatric sex tend to be more cooperative within their family groups than those of the more dispersive sex but we do not understand why. The leading "Philopatry hypothesis" proposes that the more philopatric sex cooperates more because their higher likelihood of natal breeding increases the direct fitness benefits of natal cooperation. However, the "Dispersal trade-off hypothesis" proposes that the more dispersive sex cooperates less because preparations for dispersal, such as extra-territorial prospecting, trade-off against natal cooperation. Here, we test both hypotheses in cooperatively breeding white-browed sparrow weavers (Plocepasser mahali), using a novel high-resolution automated radio-tracking method. First, we show that males are the more dispersive sex (a rare reversal of the typical avian sex difference in dispersal) and that, consistent with the predictions of both hypotheses, females contribute substantially more than males to cooperative care while within the natal group. However, the Philopatry hypothesis cannot readily explain this female-biased cooperation, as females are not more likely than males to breed within their natal group. Instead, our radio-tracking findings support the Dispersal trade-off hypothesis: males conduct pre-dispersal extra-territorial prospecting forays at higher rates than females and prospecting appears to trade-off against natal cooperation. Our findings thus highlight that the evolution of sex differences in cooperation could be widely attributable to trade-offs between cooperation and dispersal; a potentially general explanation that does not demand that cooperation yields direct fitness benefits.
Assuntos
Evolução Biológica , Comportamento Cooperativo , Animais , Feminino , Masculino , Caracteres Sexuais , Pardais/fisiologia , Distribuição Animal , Comportamento Sexual Animal/fisiologiaRESUMO
Severe respiratory syncytial virus (RSV) infection during early life has been linked to gut dysbiosis, which correlates with increased disease severity and a higher risk of developing asthma later in life. However, the impact of such early-life RSV infections on intestinal immunity in adulthood remains unclear. Herein, we show that RSV infection in 3-week-old mice induced persistent differential natural killer (NK) and T cell profiles within the lungs and gastrointestinal (GI) lymphoid tissues (GALT) in adulthood. Notably, male mice exhibited more pronounced RSV-induced changes in immune cell populations in both the lungs and GALT, while female mice displayed greater resilience. Importantly, early-life RSV infection was associated with the chronic downregulation of CD69-expressing T lymphocytes, particularly T regulatory cells in Peyer's patches, which could have a significant impact on T cell functionality and immune tolerance. We propose that RSV infection in early life is a trigger for the breakdown in immune tolerance at mucosal surfaces, with potential implications for airways allergic disease, food allergies, and other GI inflammatory diseases.