Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
1.
J Clin Invest ; 134(18)2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39286970

RESUMEN

Autoimmune diseases are a leading cause of disability worldwide. Most autoimmune diseases occur more often in women than men, with rheumatic autoimmune diseases being among those most highly expressed in women. Several key factors, identified mainly in animal models and cell culture experiments, are important in increasing autoimmune disease in females. These include sex hormones, immune genes including those found on the X chromosome, sex-specific epigenetic effects on genes by estrogen and the environment, and regulation of genes and messenger RNA by microRNAs found in extracellular vesicles. Evidence is also emerging that viruses as well as drugs or toxins that damage mitochondria may contribute to increased levels of autoantibodies against nuclear and mitochondrial antigens, which are common in many autoimmune diseases. The purpose of this Review is to summarize our current understanding of mechanisms that may determine sex differences in autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes , Autoinmunidad , Caracteres Sexuales , Humanos , Femenino , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/genética , Animales , Masculino , Epigénesis Genética , Hormonas Esteroides Gonadales/inmunología , Hormonas Esteroides Gonadales/metabolismo , Autoanticuerpos/inmunología , MicroARNs/genética , MicroARNs/inmunología , MicroARNs/metabolismo , Cromosomas Humanos X/genética , Cromosomas Humanos X/inmunología , Mitocondrias/inmunología , Mitocondrias/metabolismo , Mitocondrias/genética
2.
J Clin Invest ; 134(18)2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39286972

RESUMEN

Globally, the majority of people living with HIV are women or girls, but they have been a minority of participants in clinical trials and observational studies of HIV. Despite this underrepresentation, differences in the pathogenesis of HIV have been observed between men and women, with contributions from both gender- and sex-based factors. These include differences in the risk of HIV acquisition, in viral load set point and immune activation in responses to viremia, and differences in HIV reservoir maintenance. These differences obligate adequate study in both males and females in order to optimize treatments, but also provide a powerful leverage point for delineating the mechanisms of HIV pathogenesis. The shifts in exposure to sex steroid hormones across a lifespan introduce additional complexity, which again can be used to focus on either genetic or hormonal influences as the driver of an outcome. In this Review, we discuss consistent and reproducible differences by sex across the spectrum of HIV, from acquisition through pathogenesis, treatment, and cure, and explore potential mechanisms and gaps in knowledge.


Asunto(s)
Infecciones por VIH , Humanos , Infecciones por VIH/inmunología , Femenino , Masculino , Caracteres Sexuales , VIH-1/inmunología , Carga Viral , Hormonas Esteroides Gonadales/inmunología , Factores Sexuales
3.
J Reprod Immunol ; 165: 104295, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39053203

RESUMEN

During pregnancy, a unique immune milieu is established systemically and locally at the maternal-fetal interface. While preparing for embryonic implantation, endometrial effectors significantly change their proportions and function, which are synchronized with hormonal changes. During assisted reproductive technology cycles, various cytokines, chemokines, and immune factors dynamically change with the altered receptor expressions on the immune effectors. Thus, the hormonal regulation of immune effectors is critical to maintaining the immune milieu. In this review, hormonal effects on T cell subsets are reviewed. Sex hormones affect T cell ontogeny and development, consequently affecting their functions. Like other T cell subsets, CD4+ T helper (Th) cells are modulated by estrogen, where low estrogen concentration promotes Th1-driven cell-mediated immunity in the uterus and in vitro by enhancing IFN-γ production, while a high estrogen level decreases it. The abundance and differentiation of T regulatory (Treg) cells are controlled by estrogen, inducing Treg expansion. Conversely, progesterone maintains immune homeostasis by balancing Th1/Th2 and Th17/Treg immunity, leading to maternal-fetal tolerance. Therefore, the understanding of the hormonal impact on various T cell subsets during the reproductive cycles is critical to improving reproductive outcomes in women with recurrent pregnancy losses, repeated implantation failures, and undergoing assisted reproductive cycles.


Asunto(s)
Técnicas Reproductivas Asistidas , Humanos , Femenino , Embarazo , Animales , Linfocitos T Reguladores/inmunología , Estrógenos/metabolismo , Estrógenos/inmunología , Implantación del Embrión/inmunología , Tolerancia Inmunológica , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Hormonas Esteroides Gonadales/inmunología , Progesterona/metabolismo
4.
Front Immunol ; 15: 1379754, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38835761

RESUMEN

An increasing number of studies have highlighted the existence of a sex-specific immune response, wherein men experience a worse prognosis in cases of acute inflammatory diseases. Initially, this sex-dependent inflammatory response was attributed to the influence of sex hormones. However, a growing body of evidence has shifted the focus toward the influence of chromosomes rather than sex hormones in shaping these inflammatory sex disparities. Notably, certain pattern recognition receptors, such as Toll-like receptors (TLRs), and their associated immune pathways have been implicated in driving the sex-specific immune response. These receptors are encoded by genes located on the X chromosome. TLRs are pivotal components of the innate immune system, playing crucial roles in responding to infectious diseases, including bacterial and viral pathogens, as well as trauma-related conditions. Importantly, the TLR-mediated inflammatory responses, as indicated by the production of specific proteins and cytokines, exhibit discernible sex-dependent patterns. In this review, we delve into the subject of sex bias in TLR activation and explore its clinical implications relatively to both the X chromosome and the hormonal environment. The overarching objective is to enhance our understanding of the fundamental mechanisms underlying these sex differences.


Asunto(s)
Inflamación , Receptores Toll-Like , Animales , Femenino , Humanos , Masculino , Hormonas Esteroides Gonadales/metabolismo , Hormonas Esteroides Gonadales/inmunología , Inmunidad Innata , Inflamación/inmunología , Factores Sexuales , Transducción de Señal , Receptores Toll-Like/metabolismo , Receptores Toll-Like/inmunología
5.
Nat Rev Cancer ; 24(5): 338-355, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38589557

RESUMEN

Sex differences are present across multiple non-reproductive organ cancers, with male individuals generally experiencing higher incidence of cancer with poorer outcomes. Although some mechanisms underlying these differences are emerging, the immunological basis is not well understood. Observations from clinical trials also suggest a sex bias in conventional immunotherapies with male individuals experiencing a more favourable response and female individuals experiencing more severe adverse events to immune checkpoint blockade. In this Perspective article, we summarize the major biological hallmarks underlying sex bias in immuno-oncology. We focus on signalling from sex hormones and chromosome-encoded gene products, along with sex hormone-independent and chromosome-independent epigenetic mechanisms in tumour and immune cells such as myeloid cells and T cells. Finally, we highlight opportunities for future studies on sex differences that integrate sex hormones and chromosomes and other emerging cancer hallmarks such as ageing and the microbiome to provide a more comprehensive view of how sex differences underlie the response in cancer that can be leveraged for more effective immuno-oncology approaches.


Asunto(s)
Inmunoterapia , Neoplasias , Femenino , Humanos , Masculino , Epigénesis Genética , Hormonas Esteroides Gonadales/metabolismo , Hormonas Esteroides Gonadales/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Caracteres Sexuales , Factores Sexuales , Sexismo
6.
Nat Rev Immunol ; 24(7): 487-502, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38383754

RESUMEN

There are notable sex-based differences in immune responses to pathogens and self-antigens, with female individuals exhibiting increased susceptibility to various autoimmune diseases, and male individuals displaying preferential susceptibility to some viral, bacterial, parasitic and fungal infections. Although sex hormones clearly contribute to sex differences in immune cell composition and function, the presence of two X chromosomes in female individuals suggests that differential gene expression of numerous X chromosome-linked immune-related genes may also influence sex-biased innate and adaptive immune cell function in health and disease. Here, we review the sex differences in immune system composition and function, examining how hormones and genetics influence the immune system. We focus on the genetic and epigenetic contributions responsible for altered X chromosome-linked gene expression, and how this impacts sex-biased immune responses in the context of pathogen infection and systemic autoimmunity.


Asunto(s)
Caracteres Sexuales , Humanos , Femenino , Masculino , Animales , Hormonas Esteroides Gonadales/inmunología , Hormonas Esteroides Gonadales/metabolismo , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/genética , Epigénesis Genética/inmunología , Cromosomas Humanos X/genética , Genes Ligados a X , Autoinmunidad/inmunología , Autoinmunidad/genética , Inmunidad Innata/inmunología , Factores Sexuales , Inmunidad Adaptativa/genética , Inmunidad Adaptativa/inmunología
7.
Circ Res ; 130(4): 593-610, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35175848

RESUMEN

Immune responses differ between men and women, with women at higher risk of developing chronic autoimmune diseases and having more robust immune responses to many viruses, including HIV and hepatitis C virus. Although immune dysregulation plays a prominent role in chronic systemic inflammation, a key driver in the development of atherosclerotic cardiovascular disease (ASCVD), standard ASCVD risk prediction scores underestimate risk in populations with immune disorders, particularly women. This review focuses on the ASCVD implications of immune dysregulation due to disorders with varying global prevalence by sex: autoimmune disorders (female predominant), HIV (male-female equivalent), and hepatitis C virus (male predominant). Factors contributing to ASCVD in women with immune disorders, including traditional risk factors, dysregulated innate and adaptive immunity, sex hormones, and treatment modalities, are discussed. Finally, the need to develop new ASCVD risk stratification tools that incorporate variables specific to populations with chronic immune disorders, particularly in women, is emphasized.


Asunto(s)
Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/inmunología , Hormonas Esteroides Gonadales/inmunología , Enfermedades del Sistema Inmune/epidemiología , Enfermedades del Sistema Inmune/inmunología , Inmunidad Adaptativa/inmunología , Enfermedades Cardiovasculares/diagnóstico , Femenino , Infecciones por VIH/diagnóstico , Infecciones por VIH/epidemiología , Infecciones por VIH/inmunología , Hepatitis C/diagnóstico , Hepatitis C/epidemiología , Hepatitis C/inmunología , Humanos , Enfermedades del Sistema Inmune/diagnóstico
8.
Int Immunopharmacol ; 101(Pt B): 108255, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34688149

RESUMEN

The coronavirus disease (COVID-19) has once again reminded us of the significance of host immune response and consequential havocs of the immune dysregulation. The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) inflicts severe complications to the infected host, including cough, dyspnoea, fever, septic shock, acute respiratory distress syndrome (ARDs), and multiple organ failure. These manifestations are the consequence of the dysregulated immune system, which gives rise to excessive and unattended production of pro-inflammatory mediators. Elevated circulatory cytokine and chemokine levels are accompanied by spontaneous haemorrhage, thrombocytopenia and systemic inflammation, which are the cardinal features of life-threatening cytokine storm syndrome in advanced COVID-19 diseases. Coronavirus hijacked NF-kappa B (NF-κB) is responsible for upregulating the expressions of inflammatory cytokine, chemokine, alarmins and inducible enzymes, which paves the pathway for cytokine storm. Given the scenario, the systemic approach of simultaneous inhibition of NF-κB offers an attractive therapeutic intervention. Targeted therapies with proteasome inhibitor (VL-01, bortezomib, carfilzomib and ixazomib), bruton tyrosine kinase inhibitor (acalabrutinib), nucleotide analogue (remdesivir), TNF-α monoclonal antibodies (infliximab and adalimumab), N-acetylcysteine and corticosteroids (dexamethasone), focusing the NF-κB inhibition have demonstrated effectiveness in terms of the significant decrease in morbidity and mortality in severe COVID-19 patients. Hence, this review highlights the activation, signal transduction and cross-talk of NF-κB with regard to cytokine storm in COVID-19. Moreover, the development of therapeutic strategies based on NF-κB inhibition are also discussed herein.


Asunto(s)
COVID-19/inmunología , Síndrome de Liberación de Citoquinas/inmunología , FN-kappa B/inmunología , SARS-CoV-2 , Animales , Cromosomas Humanos X/inmunología , Exoftalmia , Hormonas Esteroides Gonadales/inmunología , Humanos , Transducción de Señal
9.
J Ovarian Res ; 14(1): 100, 2021 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-34330312

RESUMEN

OBJECTIVE: The design of this study was due to the report of the antioxidant properties of Ellagic acid (EA) for its evaluation on the Insulin resistance (IR), oxidative stress and sex hormones levels in women with polycystic ovarian syndrome (PCOS). METHODS: In this randomized, double-blind, placebo-controlled clinical trial, 60 patients were recruited. Patients were randomly allocated consumed a capsule containing 200 mg of EA per day (n = 30) or placebo (n = 30) for 8 weeks. The fasting blood sugar (FBS), insulin, IR, total cholesterol (TC), triglycerides (TG), low density lipoprotein (LDL), high density lipoprotein (HDL), total antioxidant capacity (TAC), Malondialdehyde (MDA), C-reactive protein (CRP), Tumor necrosis factor-alpha (TNF-α), sex hormones and anti-mullerian hormone (AMH) were measured at the beginning and end of the study. RESULT: At the end of the study, the mean of FBS, insulin, IR, TC, TG, LDL, MDA, CRP, TNF-α, total testosterone, prolactin and AMH were significantly decreased in the intervention group compared to the placebo group (P < 0.05). Also, there was a significant increase in the mean of TAC after supplementation with EA (P < 0.05). At the end of the study, no significant changes were observed in the mean of anthropometric factors, physical activity and food intake (P > 0.05). CONCLUSION: EA supplementation can be helpful as a diet supplement in women with PCOS through improvement in insulin resistance. This supplement may be used to reduce metabolic disorders in women. TRIAL REGISTRATION: This study was retrospectively (07-07-2019) registered in the Iranian website ( www.irct.ir ) for registration of clinical trials ( IRCT20141025019669N12 ).


Asunto(s)
Ácido Elágico/uso terapéutico , Hormonas Esteroides Gonadales/inmunología , Resistencia a la Insulina/inmunología , Estrés Oxidativo/efectos de los fármacos , Adolescente , Adulto , Método Doble Ciego , Ácido Elágico/farmacología , Femenino , Humanos , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
10.
Front Immunol ; 12: 672853, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34248954

RESUMEN

Through the release of hormones, the neuro-endocrine system regulates the immune system function promoting adaptation of the organism to the external environment and to intrinsic physiological changes. Glucocorticoids (GCs) and sex hormones not only regulate immune responses, but also control the hematopoietic stem cell (HSC) differentiation and subsequent maturation of immune cell subsets. During the development of an organism, this regulation has long-term consequences. Indeed, the effects of GC exposure during the perinatal period become evident in the adulthood. Analogously, in the context of HSC transplantation (HSCT), the immune system development starts de novo from the donor HSCs. In this review, we summarize the effects of GCs and sex hormones on the regulation of HSC, as well as of adaptive and innate immune cells. Moreover, we discuss the short and long-term implications on hematopoiesis of sex steroid ablation and synthetic GC administration upon HSCT.


Asunto(s)
Glucocorticoides/inmunología , Hormonas Esteroides Gonadales/inmunología , Sistema Inmunológico/crecimiento & desarrollo , Sistema Inmunológico/inmunología , Animales , Humanos
11.
Eur J Clin Invest ; 51(5): e13485, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33393082

RESUMEN

The new coronavirus (SARS-CoV-2) appearance in Wuhan, China, did rise the new virus disease (COVID-19), which spread globally in a short time, leading the World Health Organization to declare a new global pandemic. To contain and mitigate the spread of SARS-CoV-2, specific public health procedures were implemented in virtually all countries, with a significant impact on society, making it difficult to keep the regular practice of physical activity. It is widely accepted that an active lifestyle contributes to the improvement of general health and preservation of cardiovascular, respiratory, osteo-muscular and immune system capacities. The positive effects of regular physical activity on the immune system have emerged as a pivotal trigger of general health, underlying the beneficial effects of physical activity on multiple physiological systems. Accordingly, recent studies have already pointed out the negative impact of physical inactivity caused by the social isolation imposed by the public sanitary authorities due to COVID-19. Nevertheless, there are still no current narrative reviews evaluating the real impact of COVID-19 on active lifestyle or even discussing the possible beneficial effects of exercise-promoted immune upgrade against the severity or progression of COVID-19. Based on the consensus in the scientific literature, in this review, we discuss how an exercise adherence could adequately improve immune responses in times of the 'COVID-19 Era and beyond'.


Asunto(s)
COVID-19 , Ejercicio Físico/fisiología , Inmunidad/inmunología , Inflamación/inmunología , Leucocitos/inmunología , Control de Enfermedades Transmisibles , Citocinas/inmunología , Hormonas Esteroides Gonadales/inmunología , Humanos , Hidrocortisona/inmunología , Células Asesinas Naturales/inmunología , Neutrófilos/inmunología , Cooperación del Paciente , Fagocitosis/inmunología , Política Pública , SARS-CoV-2 , Conducta Sedentaria , Linfocitos T/inmunología
12.
Trends Microbiol ; 29(9): 849-859, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33257138

RESUMEN

The role of sex steroids in mammalian maturation is well established. Recently, it has been increasingly appreciated that sex steroids also play an important role in the propensity of adults to develop a myriad of diseases. The exposure and responsiveness of tissues to sex steroids varies among individuals and between the sexes, and this has been correlated with gender-specific differences in the composition of the intestinal microbiota and in susceptibility to metabolic, autoimmune, and neoplastic diseases. Here we focus on recent studies that demonstrate an interplay between sex steroids, the intestinal immune response, and the intestinal microbiota. While correlations between biological sex, the intestinal innate immune response, intestinal inflammation, and intestinal microbiota have been established, many gaps in our knowledge prevent the emergence of an overarching model for this complex interaction. Such a model could aid in the development of prebiotic, probiotic, or synthetic therapeutics that decrease the risk of autoimmune, metabolic, neoplastic, and infectious diseases of the intestine and mitigate the particular health risks faced by individuals receiving sex steroid treatment.


Asunto(s)
Microbioma Gastrointestinal , Hormonas Esteroides Gonadales/inmunología , Intestinos/inmunología , Intestinos/microbiología , Animales , Humanos , Inmunidad Innata
13.
Neuroendocrinology ; 111(11): 1066-1085, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33242856

RESUMEN

The coronavirus disease 2019 (COVID-19) outbreak, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a global public health issue which has profound effects on most aspects of societal well-being, including physical and mental health. A plethora of studies globally have suggested the existence of a sex disparity in the severity and outcome of COVID-19 patients, mainly due to mechanisms of virus infection, immune response to the virus, development of systemic inflammation, and consequent systemic complications, particularly thromboembolism. Epidemiological data report a sex difference in the severity of COVID-19, with a more favorable course of the disease in women compared to men regardless of age, although the rate of SARS-CoV-2 infection seems to be similar in both sexes. Sex hormones, including androgens and estrogens, may not only impact virus entry and load, but also shape the clinical manifestations, complications, and ultimately the outcome of the disease. The current review comprehensively summarizes the current literature on sex disparities in susceptibility and outcome of COVID-19 as well as the literature underpinning the pathophysiological and molecular mechanisms, which may provide a rationale to a sex disparity. These mechanisms include sex hormone influence on factors that facilitate virus entry and priming, immune and inflammatory response, as well as coagulation and thrombosis diathesis. Based on present evidence, women appear to be relatively protected from COVID-19 because of a more effective immune response and a less pronounced systemic inflammation, with consequent moderate clinical manifestations of the disease, together with a lesser predisposition to thromboembolism. Conversely, men appear to be particularly susceptible to COVID-19 because of a less effective immune response with consequent severe clinical manifestations of the disease, together with a greater predisposition to thromboembolism. In the elderly, generally characterized by the phenomenon of inflammaging, sex disparities in overall mortality following SARS-CoV-2 infection are even more palpable as elderly men appear to be more prone to severe COVID-19 because of a greater predisposition to infections, a weaker immune defense, and an enhanced thrombotic state compared to women. The information revealed from the review highlights potential novel therapeutic approaches employing the administration of hormonal or antihormonal therapy in combination with antiviral drugs in COVID-19 patients.


Asunto(s)
Enzima Convertidora de Angiotensina 2/inmunología , COVID-19/inmunología , COVID-19/mortalidad , Hormonas Esteroides Gonadales/inmunología , Índice de Severidad de la Enfermedad , Caracteres Sexuales , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Antivirales/farmacología , Antivirales/uso terapéutico , Femenino , Hormonas Esteroides Gonadales/antagonistas & inhibidores , Antagonistas de Hormonas/farmacología , Antagonistas de Hormonas/uso terapéutico , Humanos , Masculino , Factores de Riesgo , Resultado del Tratamiento , Tratamiento Farmacológico de COVID-19
15.
Biol Sex Differ ; 11(1): 53, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32948238

RESUMEN

Coronavirus disease 2019 (COVID-19) has shown high infection and mortality rates all over the world, and despite the global efforts, there is so far no specific therapy available for COVID-19. Interestingly, while the severity and mortality of COVID-19 are higher in males than in females, the underlying molecular mechanisms are unclear. In this review, we explore sex-related differences that may be contributing factors to the observed male-biased mortality from COVID-19. Males are considered the weaker sex in aspects related to endurance and infection control. Studies show that viral RNA clearance is delayed in males with COVID-19. A recent study has indicated that the testis can harbor coronavirus, and consequently, males show delayed viral clearance. However, the role of testis involvement in COVID-19 severity and mortality needs further research. Males and females show a distinct difference in immune system responses with females eliciting stronger immune responses to pathogens. This difference in immune system responses may be a major contributing factor to viral load, disease severity, and mortality. In addition, differences in sex hormone milieus could also be a determinant of viral infections as estrogen has immunoenhancing effects while testosterone has immunosuppressive effects. The sex-specific severity of COVID-19 infections indicates that further research on understanding the sex differences is needed. Inclusion of both males and females in basic research and clinical trials is required to provide critical information on sex-related differences that may help to better understand disease outcome and therapy.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/mortalidad , Neumonía Viral/mortalidad , Secuencia de Aminoácidos , Betacoronavirus/aislamiento & purificación , COVID-19 , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Femenino , Hormonas Esteroides Gonadales/inmunología , Humanos , Inmunidad , Masculino , Pandemias , Neumonía Viral/epidemiología , Neumonía Viral/inmunología , Neumonía Viral/virología , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Factores Sexuales , Testículo/inmunología
16.
Front Immunol ; 11: 2147, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32983176

RESUMEN

The novel severe acute respiratory syndrome coronavirus 2, the cause of the coronavirus disease 2019 (COVID-19) pandemic, has ravaged the world, with over 22 million total cases and over 770,000 deaths worldwide as of August 18, 2020. While the elderly are most severely affected, implicating an age bias, a striking factor in the demographics of this deadly disease is the gender bias, with higher numbers of cases, greater disease severity, and higher death rates among men than women across the lifespan. While pre-existing comorbidities and social, behavioral, and lifestyle factors contribute to this bias, biological factors underlying the host immune response may be crucial contributors. Women mount stronger immune responses to infections and vaccinations and outlive men. Sex-based biological factors underlying the immune response are therefore important determinants of susceptibility to infections, disease outcomes, and mortality. Despite this, gender is a profoundly understudied and often overlooked variable in research related to the immune response and infectious diseases, and it is largely ignored in drug and vaccine clinical trials. Understanding these factors will not only help better understand the pathogenesis of COVID-19, but it will also guide the design of effective therapies and vaccine strategies for gender-based personalized medicine. This review focuses on sex-based differences in genes, sex hormones, and the microbiome underlying the host immune response and their relevance to infections with a focus on coronaviruses.


Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/mortalidad , Interacciones Huésped-Patógeno/inmunología , Neumonía Viral/inmunología , Neumonía Viral/mortalidad , Inmunidad Adaptativa/genética , COVID-19 , Infecciones por Coronavirus/virología , Femenino , Predisposición Genética a la Enfermedad , Hormonas Esteroides Gonadales/inmunología , Interacciones Huésped-Patógeno/genética , Humanos , Inmunidad Innata/genética , Masculino , Microbiota/inmunología , Pandemias , Neumonía Viral/virología , SARS-CoV-2 , Factores Sexuales
17.
J Pharmacol Exp Ther ; 375(1): 161-174, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32759370

RESUMEN

Women are at significantly greater risk of developing stress-related disorders such as depression. The increased risk begins during puberty and continues throughout life until menopause, suggesting a role for ovarian hormones in this increased susceptibility. Importantly, inflammation has been gaining momentum in its role in the pathogenesis of depression. Herein, clinical and preclinical studies have been reviewed to better understand how sex differences within the immune system may contribute to exaggerated risk of depression in females. First, studies that investigate the ability of psychologic stress episodes to engage the inflammatory systems both in the brain and periphery are reviewed with a special focus on sex-specific effects. Moreover, studies are discussed that identify whether imbalanced inflammatory milieu contributes to the development of depression in males versus females and whether these effects are regulated by estradiol. Importantly, we propose a locus coeruleus-norepinephrine-cytokine circuit as a conduit through which stress could increase stress susceptibly in females. Finally, the anti-inflammatory capacity of traditional and nontraditional antidepressants is investigated, with the goal of providing a better understanding of pharmacotherapeutics to enhance strategies to personalize antidepressant treatments between the sexes. The studies reviewed herein strongly support the need for further studies to elucidate whether females are especially sensitive to anti-inflammatory compounds as adjuvants to traditional therapies. SIGNIFICANCE STATEMENT: Women have hve an increased risk of developing stress-related disorders such as depression. In this review, literature from clinical and preclinical studies are integrated to define sex differences in stress-induced inflammatory responses as a potential source for the etiology of sex differences in depressive disorders. Moreover, the anti-inflammatory capacity of traditional and nontraditional antidepressants is reviewed to inform on potential pharmacotherapeutic strategies to personalize antidepressant therapy in a sex-dependent manner.


Asunto(s)
Encéfalo/efectos de los fármacos , Depresión/tratamiento farmacológico , Neuroinmunomodulación/efectos de los fármacos , Caracteres Sexuales , Estrés Psicológico/tratamiento farmacológico , Animales , Antidepresivos/uso terapéutico , Encéfalo/inmunología , Depresión/etiología , Depresión/inmunología , Femenino , Hormonas Esteroides Gonadales/inmunología , Humanos , Inflamación , Masculino , Microglía/efectos de los fármacos , Microglía/inmunología , Ovario/inmunología , Medicina de Precisión , Estrés Psicológico/complicaciones , Estrés Psicológico/inmunología
18.
Prehosp Disaster Med ; 35(4): 438-441, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32600476

RESUMEN

Studies have reported a sex bias in case fatalities of COVID-19 patients. Moreover, it is observed that men have a higher risk of developing a severe form of the disease compared to women, highlighting the importance of disaggregated data of male and female COVID-19 patients. On the other hand, other factors (eg, hormonal levels and immune functions) also need to be addressed due to the effects of sex differences on the outcomes of COVID-19 patients. An insight into the underlying causes of sex differences in COVID-19 patients may provide an opportunity for better care of the patients or prevention of the disease. The current study reviews the reports concerning with the sex differences in COVID-19 patients. It is explained how sex can affect angiotensin converting enzyme-2 (ACE2), that is a key component for the pathogenesis of COVID-19, and summarized the gender differences in immune responses and how sex hormones are involved in immune processes. Furthermore, the available data about the impact of sex hormones on the immune functions of COVID-19 cases are looked into.


Asunto(s)
Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/inmunología , Neumonía Viral/epidemiología , Neumonía Viral/inmunología , Enzima Convertidora de Angiotensina 2 , Betacoronavirus , COVID-19 , Infecciones por Coronavirus/mortalidad , Femenino , Hormonas Esteroides Gonadales/inmunología , Humanos , Masculino , Pandemias , Peptidil-Dipeptidasa A/fisiología , Neumonía Viral/mortalidad , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Factores Sexuales
19.
Int Immunopharmacol ; 87: 106776, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32682255

RESUMEN

During pregnancy, the fetal-maternal interface underlies several dynamic alterations to permit the fetus to be cultivated and developed in the uterus, in spite of being identifies by the maternal immune system. A large variety of decidual leukocyte populations, including natural killer cells, NKT cells, innate lymphoid cells, dendritic cells, B cells, T cells, subpopulations of helper T cells play a vital role in controlling the trophoblast invasion, angiogenesis as well as vascular remodeling. In contrast, several regulatory immunosuppressive mechanisms, including regulatory T cells, regulatory B cells, several cytokines and mediators are involved in maintain the homeostasis of immune system in the fetal-maternal interface. Nonetheless, aberrant alterations in the balance of immune inflammatory or immunosuppressive arms have been associated with various pregnancy losses and infertilities. As a result, numerous strategies have been developed to revers dysregulated balance of immune players to increase the chance of successful pregnancy. Lymphocyte immunotherapy has been developed through utilization of peripheral white blood cells of the husband or others and administered into the mother to confer an immune tolerance for embryo's antigens. However, the results have not always been promising, implying to further investigations to improve the approach. This review attempts to clarify the involvement of lymphocytes in contributing to the pregnancy outcome and the potential of lymphocyte immunotherapy in treatment of infertilities with dysregulated immune system basis.


Asunto(s)
Inmunoterapia , Infertilidad/terapia , Linfocitos/inmunología , Embarazo/inmunología , Animales , Femenino , Hormonas Esteroides Gonadales/inmunología , Humanos , Infertilidad/inmunología
20.
PLoS One ; 15(6): e0235174, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32574226

RESUMEN

AIM: To investigate markers of systemic inflammation in pre- and postmenopausal women and identify possible predictors of systemic inflammation with menopause. METHODS: Cross-sectional study of 69 healthy women between 45- and 60 years. Blood samples were collected to assess leukocyte subsets and plasma cytokines. MRI and DXA scans were performed to assess body composition. Through uni- and multivariate analyses, follicle-stimulating hormone (FSH), visceral fat mass and age were evaluated as predictors of systemic inflammation in relation to menopause. RESULTS: Postmenopausal women tended to have higher leukocyte counts (5.4 x109 vs. 4.9 x109 cells/l, p = 0.05) reflected in increased total lymphocytes (1.8 x109 vs. 1.6 x109 cells/l, p = 0.01) and monocytes (0.5 x109 vs. 0.4 x109 cells/l, p = 0.02), compared to premenopausal women. Increased visceral fat mass was a strong predictor of high leukocyte subsets. Postmenopausal women had higher plasma TNF-α (2.24 vs. 1.91 pg/ml, p = 0.01) and IL-6 (0.45 vs. 0.33 pg/ml, p = 0.004) compared to premenopausal women and high FSH was a significant predictor of increased plasma TNF-α, IL-1ß and IL-6. Menopause was further associated with increased T-cells (1,336 vs. 1,128 cells/µl, p = 0.04) reflected in significantly higher counts of exhausted-, senescent-, and memory CD4+ T-cell subsets. CONCLUSIONS: Menopause is associated with increased systemic inflammation as well as exhausted- and senescent T-cells. We suggest, that both increased visceral fat mass and declining sex hormone levels might contribute to postmenopausal systemic inflammation and calls for further large-scale studies to confirm these findings.


Asunto(s)
Citocinas/inmunología , Inflamación/inmunología , Posmenopausia/inmunología , Subgrupos de Linfocitos T/inmunología , Absorciometría de Fotón/métodos , Composición Corporal , Citocinas/sangre , Citocinas/metabolismo , Femenino , Hormona Folículo Estimulante/sangre , Hormona Folículo Estimulante/inmunología , Hormonas Esteroides Gonadales/sangre , Hormonas Esteroides Gonadales/inmunología , Humanos , Inflamación/sangre , Inflamación/metabolismo , Grasa Intraabdominal/diagnóstico por imagen , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/metabolismo , Linfocitos/citología , Linfocitos/inmunología , Linfocitos/metabolismo , Imagen por Resonancia Magnética/métodos , Persona de Mediana Edad , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Análisis Multivariante , Posmenopausia/sangre , Posmenopausia/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...