Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 789
Filtrar
1.
Herz ; 49(4): 249-253, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38954012

RESUMEN

Cardiovascular diseases are the leading cause of death worldwide. Pathophysiologically, metabolic and inflammatory processes contribute substantially to the development and progression of cardiovascular diseases. Over the past decade, the role of disease-propagating inflammatory processes has been strengthened and reframed, leading to trials testing anti-inflammatory drugs for the treatment of atherosclerosis and its complications. Despite these achievements, further research in both pre-clinical and clinical studies is warranted to explore new targets, to better identify responders, and to refine therapy strategies to combat inflammation in human disease. Environmental disturbances, so-called lifestyle-associated cardiovascular risk factors, greatly alter the immune system in general and leukocytes in particular, thus affecting the progression of atherosclerosis. Epidemiological studies have shown that exposure to mental stress can be closely linked to the occurrence of cardiovascular disease. Here, we describe how acute and chronic mental stress alter the immune system via neuroimmune interactions, thereby modifying vascular inflammation. In addition, we identify gaps that still need to be addressed in the future.


Asunto(s)
Neuroinmunomodulación , Estrés Psicológico , Humanos , Estrés Psicológico/inmunología , Estrés Psicológico/complicaciones , Neuroinmunomodulación/inmunología , Neuroinmunomodulación/fisiología , Inflamación/inmunología , Modelos Inmunológicos , Enfermedades Cardiovasculares/inmunología , Aterosclerosis/inmunología
2.
Neuroimmunomodulation ; 31(1): 143-156, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38934151

RESUMEN

BACKGROUND: Establishing causal relationships is essential in biology and medicine. However, various notions of causality have been operationalized at different times in various fields of the life and health sciences. While this is expected from a history or sociology of science point of view, as different accounts may correspond to what is valued in terms of establishing causal relationships at different times as well as in different fields of biology and medicine, this may come as a surprise for a present-day actor in those fields. If, over time, causal accounts have not been fully dismissed, then they are likely to invite some form of, potentially salutary, explanatory pluralism. SUMMARY: In the decades following WWII, psychosomatic medicine could propose that psychological factors cause somatic diseases. But today, most medicine has to meet the standard of a randomized clinical trial before any causal relationship can be proposed. Instead, in biology, mechanisms seem to be the most-valued causal discourse to explain how phenomena of interest are brought about. Here, the focus will be on how psychoneuroimmunology, an interdisciplinary research field addressing interactions between the nervous system and immune system, and between behavior and health, has considered causal relationships between psychological factors and cancer. KEY MESSAGES: When it comes to causal explanations of links between psychological factors and cancer, psychoneuroimmunology is invited to consider the question of the directionality of these links as well as what and how factors causally contribute to cancer.


Asunto(s)
Neoplasias , Neuroinmunomodulación , Psiconeuroinmunología , Medicina Psicosomática , Humanos , Neuroinmunomodulación/fisiología , Neuroinmunomodulación/inmunología , Neoplasias/inmunología , Neoplasias/psicología , Historia del Siglo XX , Causalidad
3.
Nature ; 630(8017): 695-703, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38692285

RESUMEN

The body-brain axis is emerging as a principal conductor of organismal physiology. It senses and controls organ function1,2, metabolism3 and nutritional state4-6. Here we show that a peripheral immune insult strongly activates the body-brain axis to regulate immune responses. We demonstrate that pro-inflammatory and anti-inflammatory cytokines communicate with distinct populations of vagal neurons to inform the brain of an emerging inflammatory response. In turn, the brain tightly modulates the course of the peripheral immune response. Genetic silencing of this body-brain circuit produced unregulated and out-of-control inflammatory responses. By contrast, activating, rather than silencing, this circuit affords neural control of immune responses. We used single-cell RNA sequencing, combined with functional imaging, to identify the circuit components of this neuroimmune axis, and showed that its selective manipulation can effectively suppress the pro-inflammatory response while enhancing an anti-inflammatory state. The brain-evoked transformation of the course of an immune response offers new possibilities in the modulation of a wide range of immune disorders, from autoimmune diseases to cytokine storm and shock.


Asunto(s)
Encéfalo , Citocinas , Inflamación , Neuroinmunomodulación , Animales , Femenino , Masculino , Ratones , Encéfalo/citología , Encéfalo/inmunología , Encéfalo/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Ratones Endogámicos C57BL , Neuroinmunomodulación/inmunología , Neuroinmunomodulación/fisiología , Neuronas/fisiología , Nervio Vago/citología , Nervio Vago/fisiología , Análisis de Expresión Génica de una Sola Célula
4.
Neuroimmunomodulation ; 31(1): 102-113, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38697052

RESUMEN

BACKGROUND: More than a century ago, experimental work and clinical observations revealed the functional communication between the brain and the peripheral immune system. This is documented on the one hand by studies first demonstrating the effects of catecholamines on the circulation of leukocytes in experimental animals and humans, and on the other hand via the work of Russian physiologist Ivan Petrovic Pavlov and his coworkers, reporting observations that associative learning can modify peripheral immune functions. This work later fell into oblivion since little was known about the endocrine and immune system's function and even less about the underlying mechanisms of how learning, a central nervous system activity, could affect peripheral immune responses. SUMMARY: In this article, we embark on a fascinating exploration of the historical trajectory of behaviorally conditioned immune responses. KEY MESSAGE: We will pay homage to the visionary scientists who laid the groundwork for this field of research, tracing its evolution from early theories of how associative learning can affect immunity to the modern-day insights that behavioral conditioning of pharmacological responses can be exploited to improve the efficacy of medical interventions for patients.


Asunto(s)
Aprendizaje por Asociación , Humanos , Animales , Historia del Siglo XX , Historia del Siglo XXI , Aprendizaje por Asociación/fisiología , Sistema Inmunológico/fisiología , Sistema Inmunológico/inmunología , Neuroinmunomodulación/fisiología , Neuroinmunomodulación/inmunología
5.
J Neuroinflammation ; 21(1): 135, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802931

RESUMEN

Traumatic brain injury (TBI) is a major cause of disability and mortality worldwide, particularly among the elderly, yet our mechanistic understanding of what renders the post-traumatic brain vulnerable to poor outcomes, and susceptible to neurological disease, is incomplete. It is well established that dysregulated and sustained immune responses elicit negative consequences after TBI; however, our understanding of the neuroimmune interface that facilitates crosstalk between central and peripheral immune reservoirs is in its infancy. The meninges serve as the interface between the brain and the immune system, facilitating important bi-directional roles in both healthy and disease settings. It has been previously shown that disruption of this system exacerbates neuroinflammation in age-related neurodegenerative disorders such as Alzheimer's disease; however, we have an incomplete understanding of how the meningeal compartment influences immune responses after TBI. In this manuscript, we will offer a detailed overview of the holistic nature of neuroinflammatory responses in TBI, including hallmark features observed across clinical and animal models. We will highlight the structure and function of the meningeal lymphatic system, including its role in immuno-surveillance and immune responses within the meninges and the brain. We will provide a comprehensive update on our current knowledge of meningeal-derived responses across the spectrum of TBI, and identify new avenues for neuroimmune modulation within the neurotrauma field.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Meninges , Enfermedades Neuroinflamatorias , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Humanos , Animales , Meninges/inmunología , Meninges/patología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/patología , Neuroinmunomodulación/fisiología , Neuroinmunomodulación/inmunología
6.
Neuroimmunomodulation ; 31(1): 78-88, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38527434

RESUMEN

BACKGROUND: The brain and the immune systems represent the two primary adaptive systems within the body. Both are involved in a dynamic process of communication, vital for the preservation of mammalian homeostasis. This interplay involves two major pathways: the hypothalamic-pituitary-adrenal axis and the sympathetic nervous system. SUMMARY: The establishment of infection can affect immunoneuroendocrine interactions, with functional consequences for immune organs, particularly the thymus. Interestingly, the physiology of this primary organ is not only under the control of the central nervous system (CNS) but also exhibits autocrine/paracrine regulatory circuitries mediated by hormones and neuropeptides that can be altered in situations of infectious stress or chronic inflammation. In particular, Chagas disease, caused by the protozoan parasite Trypanosoma cruzi (T. cruzi), impacts upon immunoneuroendocrine circuits disrupting thymus physiology. Here, we discuss the most relevant findings reported in relation to brain-thymic connections during T. cruzi infection, as well as their possible implications for the immunopathology of human Chagas disease. KEY MESSAGES: During T. cruzi infection, the CNS influences thymus physiology through an intricate network involving hormones, neuropeptides, and pro-inflammatory cytokines. Despite some uncertainties in the mechanisms and the fact that the link between these abnormalities and chronic Chagasic cardiomyopathy is still unknown, it is evident that the precise control exerted by the brain over the thymus is markedly disrupted throughout the course of T. cruzi infection.


Asunto(s)
Encéfalo , Enfermedad de Chagas , Timo , Humanos , Enfermedad de Chagas/inmunología , Enfermedad de Chagas/fisiopatología , Animales , Encéfalo/inmunología , Timo/inmunología , Timo/fisiología , Trypanosoma cruzi/fisiología , Trypanosoma cruzi/inmunología , Sistema Hipotálamo-Hipofisario/inmunología , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Neuroinmunomodulación/fisiología , Neuroinmunomodulación/inmunología , Sistema Hipófiso-Suprarrenal/inmunología , Sistema Hipófiso-Suprarrenal/fisiopatología , Sistema Hipófiso-Suprarrenal/metabolismo
8.
Nature ; 612(7940): 417-429, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36517712

RESUMEN

The concept of immune privilege suggests that the central nervous system is isolated from the immune system. However, recent studies have highlighted the borders of the central nervous system as central sites of neuro-immune interactions. Although the nervous and immune systems both function to maintain homeostasis, under rare circumstances, they can develop pathological interactions that lead to neurological or psychiatric diseases. Here we discuss recent findings that dissect the key anatomical, cellular and molecular mechanisms that enable neuro-immune responses at the borders of the brain and spinal cord and the implications of these interactions for diseases of the central nervous system.


Asunto(s)
Encéfalo , Sistema Inmunológico , Neuroinmunomodulación , Encéfalo/inmunología , Encéfalo/fisiología , Encéfalo/fisiopatología , Sistema Inmunológico/inmunología , Sistema Inmunológico/fisiología , Sistema Inmunológico/fisiopatología , Neuroinmunomodulación/inmunología , Neuroinmunomodulación/fisiología , Médula Espinal/inmunología , Médula Espinal/fisiología , Médula Espinal/fisiopatología , Humanos , Enfermedades del Sistema Nervioso/inmunología , Enfermedades del Sistema Nervioso/fisiopatología , Enfermedades del Sistema Nervioso/psicología
9.
Signal Transduct Target Ther ; 7(1): 307, 2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-36064538

RESUMEN

Mesenchymal stromal cells (MSCs) have been considered a promising alternative for treatment of acute respiratory distress syndrome (ARDS). However, there is significant heterogeneity in their therapeutic efficacy, largely owing to the incomplete understanding of the mechanisms underlying the therapeutic activities of MSCs. Here, we hypothesize that the cholinergic anti-inflammatory pathway (CAP), which is recognized as a neuroimmunological pathway, may be involved in the therapeutic mechanisms by which MSCs mitigate ARDS. Using lipopolysaccharide (LPS) and bacterial lung inflammation models, we found that inflammatory cell infiltration and Evans blue leakage were reduced and that the expression levels of choline acetyltransferase (ChAT) and vesicular acetylcholine transporter (VAChT) in lung tissue were significantly increased 6 hours after MSC infusion. When the vagus nerve was blocked or α7 nicotinic acetylcholine (ACh) receptor (α7nAChR)-knockout mice were used, the therapeutic effects of MSCs were significantly reduced, suggesting that the CAP may play an important role in the effects of MSCs in ARDS treatment. Our results further showed that MSC-derived prostaglandin E2 (PGE2) likely promoted ACh synthesis and release. Additionally, based on the efficacy of nAChR and α7nAChR agonists, we found that lobeline, the nicotinic cholinergic receptor excitation stimulant, may attenuate pulmonary inflammation and alleviate respiratory symptoms of ARDS patients in a clinical study (ChiCTR2100047403). In summary, we reveal a previously unrecognized MSC-mediated mechanism of CAP activation as the means by which MSCs alleviate ARDS-like syndrome, providing insight into the clinical translation of MSCs or CAP-related strategies for the treatment of patients with ARDS.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Neuroinmunomodulación , Síndrome de Dificultad Respiratoria , Receptor Nicotínico de Acetilcolina alfa 7 , Animales , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Noqueados , Neuroinmunomodulación/genética , Neuroinmunomodulación/inmunología , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/terapia , Receptor Nicotínico de Acetilcolina alfa 7/genética , Receptor Nicotínico de Acetilcolina alfa 7/inmunología
11.
J Exp Med ; 219(3)2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-35195682

RESUMEN

Leukocyte trafficking between blood and tissues is an essential function of the immune system that facilitates humoral and cellular immune responses. Within tissues, leukocytes perform surveillance and effector functions via cell motility and migration toward sites of tissue damage, infection, or inflammation. Neurotransmitters that are produced by the nervous system influence leukocyte trafficking around the body and the interstitial migration of immune cells in tissues. Neural regulation of leukocyte dynamics is influenced by circadian rhythms and altered by stress and disease. This review examines current knowledge of neuro-immune interactions that regulate leukocyte migration and consequences for protective immunity against infections and cancer.


Asunto(s)
Leucocitos/inmunología , Neuroinmunomodulación/inmunología , Movimiento Celular/inmunología , Quimiotaxis de Leucocito/inmunología , Ritmo Circadiano/inmunología , Humanos , Modelos Inmunológicos , Modelos Neurológicos , Vías Nerviosas/inmunología , Sistema Nervioso Simpático/inmunología , Microambiente Tumoral/inmunología
12.
Ann Neurol ; 91(3): 342-352, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35067959

RESUMEN

OBJECTIVE: The study was undertaken to assess the impact of B cell depletion on humoral and cellular immune responses to severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) vaccination in patients with various neuroimmunologic disorders on anti-CD20 therapy. This included an analysis of the T cell vaccine response to the SARS-CoV-2 Delta variant. METHODS: We investigated prospectively humoral and cellular responses to SARS-CoV-2 mRNA vaccination in 82 patients with neuroimmunologic disorders on anti-CD20 therapy and 82 age- and sex-matched healthy controls. For quantification of antibodies, the Elecsys anti-SARS-CoV-2 viral spike (S) immunoassay against the receptor-binding domain (RBD) was used. IFN-gamma enzyme-linked immunosorbent spot assays were performed to assess T cell responses against the SARS-CoV-2 Wuhan strain and the Delta variant. RESULTS: SARS-CoV-2-specific antibodies were found less frequently in patients (70% [57/82]) compared with controls (82/82 [100%], p < 0.001). In patients without detectable B cells (<1 B cell/mcl), seroconversion rates and antibody levels were lower compared to nondepleted (≥1 B cell/mcl) patients (p < 0.001). B cell levels ≥1 cell/mcl were sufficient to induce seroconversion in our cohort of anti-CD20 treated patients. In contrast to the antibody response, the T-cell response against the Wuhan strain and the Delta variant was more pronounced in frequency (p < 0.05) and magnitude (p < 0.01) in B-cell depleted compared to nondepleted patients. INTERPRETATION: Antibody responses to SARS-CoV-2 mRNA vaccinnation can be attained in patients on anti-CD20 therapy by the onset of B cell repopulation. In the absence of B cells, a strong T cell response is generated which may help to protect against severe coronavirus disease 2019 (COVID-19) in this high-risk population. ANN NEUROL 2022;91:342-352.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Linfocitos B/inmunología , Vacunas contra la COVID-19/administración & dosificación , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , SARS-CoV-2/inmunología , Adulto , Enfermedades Autoinmunes del Sistema Nervioso/sangre , Enfermedades Autoinmunes del Sistema Nervioso/epidemiología , Linfocitos B/metabolismo , COVID-19/epidemiología , COVID-19/prevención & control , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuroinmunomodulación/inmunología , Estudios Prospectivos , SARS-CoV-2/metabolismo
14.
Front Immunol ; 12: 742173, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34603329

RESUMEN

Although anatomically distant from the central nervous system (CNS), gut-derived signals can dynamically regulate both peripheral immune cells and CNS-resident glial cells to modulate disease. Recent discoveries of specific microbial taxa and microbial derived metabolites that modulate neuroinflammation and neurodegeneration have provided mechanistic insight into how the gut may modulate the CNS. Furthermore, the participation of the gut in regulation of peripheral and CNS immune activity introduces a potential therapeutic target. This review addresses emerging literature on how the microbiome can affect glia and circulating lymphocytes in preclinical models of human CNS disease. Critically, this review also discusses how the host may in turn influence the microbiome, and how this may impact CNS homeostasis and disease, potentially through the production of IgA.


Asunto(s)
Enfermedades del Sistema Nervioso Central/inmunología , Microbioma Gastrointestinal/inmunología , Inmunoglobulina A/inmunología , Neuroinmunomodulación/inmunología , Animales , Humanos , Enfermedades Neuroinflamatorias/inmunología
15.
Front Immunol ; 12: 742449, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34707612

RESUMEN

The gut-brain axis refers to the bidirectional communication between the gut and brain, and regulates intestinal homeostasis and the central nervous system via neural networks and neuroendocrine, immune, and inflammatory pathways. The development of sequencing technology has evidenced the key regulatory role of the gut microbiota in several neurological disorders, including Parkinson's disease, Alzheimer's disease, and multiple sclerosis. Epilepsy is a complex disease with multiple risk factors that affect more than 50 million people worldwide; nearly 30% of patients with epilepsy cannot be controlled with drugs. Interestingly, patients with inflammatory bowel disease are more susceptible to epilepsy, and a ketogenic diet is an effective treatment for patients with intractable epilepsy. Based on these clinical facts, the role of the microbiome and the gut-brain axis in epilepsy cannot be ignored. In this review, we discuss the relationship between the gut microbiota and epilepsy, summarize the possible pathogenic mechanisms of epilepsy from the perspective of the microbiota gut-brain axis, and discuss novel therapies targeting the gut microbiota. A better understanding of the role of the microbiota in the gut-brain axis, especially the intestinal one, would help investigate the mechanism, diagnosis, prognosis evaluation, and treatment of intractable epilepsy.


Asunto(s)
Eje Cerebro-Intestino/inmunología , Encéfalo/inmunología , Epilepsia/inmunología , Microbioma Gastrointestinal/inmunología , Neuroinmunomodulación/inmunología , Animales , Humanos
16.
Molecules ; 26(20)2021 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-34684720

RESUMEN

Nicotinic acetylcholine receptors (nAChRs) are widely expressed in or on various cell types and have diverse functions. In immune cells nAChRs regulate proliferation, differentiation and cytokine release. Specifically, activation of the α7 nAChR reduces inflammation as part of the cholinergic anti-inflammatory pathway. Here we review numerous effects of α7 nAChR activation on immune cell function and differentiation. Further, we also describe evidence implicating this receptor and its chaperone RIC-3 in diseases of the central nervous system and in neuroinflammation, focusing on multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Deregulated neuroinflammation due to dysfunction of α7 nAChR provides one explanation for involvement of this receptor and of RIC-3 in neurodegenerative diseases. In this review, we also provide evidence implicating α7 nAChRs and RIC-3 in neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) involving neuroinflammation. Besides, we will describe the therapeutic implications of activating the cholinergic anti-inflammatory pathway for diseases involving neuroinflammation.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Neuroinmunomodulación/fisiología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Humanos , Inflamación/metabolismo , Chaperonas Moleculares/metabolismo , Neuroinmunomodulación/inmunología , Transducción de Señal
17.
J Neuroinflammation ; 18(1): 231, 2021 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-34645457

RESUMEN

It is well accepted that environmental stressors experienced over a one's life, from microbial infections to chemical toxicants to even psychological stressors, ultimately shape central nervous system (CNS) functioning but can also contribute to its eventual breakdown. The severity, timing and type of such environmental "hits", woven together with genetic factors, likely determine what CNS outcomes become apparent. This focused review assesses the current COVID-19 pandemic through the lens of a multi-hit framework and disuses how the SARS-COV-2 virus (causative agent) might impact the brain and potentially interact with other environmental insults. What the long-term consequences of SAR2 COV-2 upon neuronal processes is yet unclear, but emerging evidence is suggesting the possibility of microglial or other inflammatory factors as potentially contributing to neurodegenerative illnesses. Finally, it is critical to consider the impact of the virus in the context of the substantial psychosocial stress that has been associated with the global pandemic. Indeed, the loneliness, fear to the future and loss of social support alone has exerted a massive impact upon individuals, especially the vulnerable very young and the elderly. The substantial upswing in depression, anxiety and eating disorders is evidence of this and in the years to come, this might be matched by a similar spike in dementia, as well as motor and cognitive neurodegenerative diseases.


Asunto(s)
COVID-19/inmunología , Mediadores de Inflamación/inmunología , Trastornos Mentales/inmunología , Enfermedades Neurodegenerativas/inmunología , Neuroinmunomodulación/inmunología , Animales , Encéfalo/inmunología , COVID-19/epidemiología , Humanos , Inmunoterapia/tendencias , Trastornos Mentales/epidemiología , Trastornos Mentales/terapia , Enfermedades Neurodegenerativas/epidemiología , Enfermedades Neurodegenerativas/terapia , Estrés Psicológico/epidemiología , Estrés Psicológico/inmunología , Estrés Psicológico/terapia
18.
J Neuroinflammation ; 18(1): 212, 2021 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-34530858

RESUMEN

BACKGROUND: Binge ethanol exposure during adolescence reduces hippocampal neurogenesis, a reduction which persists throughout adulthood despite abstinence. This loss of neurogenesis, indicated by reduced doublecortin+ immunoreactivity (DCX+IR), is paralleled by an increase in hippocampal proinflammatory signaling cascades. As galantamine, a cholinesterase inhibitor, has anti-inflammatory actions, we tested the hypothesis that galantamine would prevent (study 1) or restore (study 2) AIE induction of proinflammatory signals within the hippocampus as well as AIE-induced loss of hippocampal neurogenesis. METHODS: Galantamine (4 mg/kg) or vehicle (saline) was administered to Wistar rats during adolescent intermittent ethanol (AIE; 5.0 g/kg ethanol, 2 days on/2 days off, postnatal day [P] 25-54) (study 1, prevention) or after AIE during abstinent maturation to adulthood (study 2, restoration). RESULTS: Results indicate AIE reduced DCX+IR and induced cleaved caspase3 (Casp3) in DCX-expressing immature neurons. Excitingly, AIE induction of activated Casp3 in DCX-expressing neurons is both prevented and reversed by galantamine treatment, which also resulted in prevention and restoration of neurogenesis (DCX+IR). Similarly, galantamine prevented and/or reversed AIE induction of proinflammatory markers, including the chemokine (C-C motif) ligand 2 (CCL2), cyclooxygenase-2 (COX-2), and high mobility group box 1 (HMGB1) protein, suggesting that AIE induction of proinflammatory signaling mediates both cell death cascades and hippocampal neurogenesis. Interestingly, galantamine treatment increased Ki67+IR generally as well as increased pan-Trk expression specifically in AIE-treated rats but failed to reverse AIE induction of NADPH-oxidase (gp91phox). CONCLUSIONS: Collectively, our studies suggest that (1) loss of neurogenesis after AIE is mediated by persistent induction of proinflammatory cascades which drive activation of cell death machinery in immature neurons, and (2) galantamine can prevent and restore AIE disruptions in the hippocampal environmental milieu to then prevent and restore AIE-mediated loss of neurogenesis.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/tratamiento farmacológico , Etanol/toxicidad , Galantamina/uso terapéutico , Hipocampo/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuroinmunomodulación/efectos de los fármacos , Factores de Edad , Animales , Consumo Excesivo de Bebidas Alcohólicas/inmunología , Consumo Excesivo de Bebidas Alcohólicas/patología , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Femenino , Galantamina/farmacología , Hipocampo/inmunología , Hipocampo/patología , Masculino , Neurogénesis/inmunología , Neuroinmunomodulación/inmunología , Ratas , Ratas Wistar
19.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34502395

RESUMEN

Stroke disrupts the homeostatic balance within the brain and is associated with a significant accumulation of necrotic cellular debris, fluid, and peripheral immune cells in the central nervous system (CNS). Additionally, cells, antigens, and other factors exit the brain into the periphery via damaged blood-brain barrier cells, glymphatic transport mechanisms, and lymphatic vessels, which dramatically influence the systemic immune response and lead to complex neuroimmune communication. As a result, the immunological response after stroke is a highly dynamic event that involves communication between multiple organ systems and cell types, with significant consequences on not only the initial stroke tissue injury but long-term recovery in the CNS. In this review, we discuss the complex immunological and physiological interactions that occur after stroke with a focus on how the peripheral immune system and CNS communicate to regulate post-stroke brain homeostasis. First, we discuss the post-stroke immune cascade across different contexts as well as homeostatic regulation within the brain. Then, we focus on the lymphatic vessels surrounding the brain and their ability to coordinate both immune response and fluid homeostasis within the brain after stroke. Finally, we discuss how therapeutic manipulation of peripheral systems may provide new mechanisms to treat stroke injury.


Asunto(s)
Neuroinmunomodulación/inmunología , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/patología , Animales , Transporte Biológico , Barrera Hematoencefálica/patología , Encéfalo/patología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/fisiología , Homeostasis , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/patología , Inmunidad , Leucocitos , Linfangiogénesis , Vasos Linfáticos , Neuroinmunomodulación/fisiología
20.
Nat Rev Neurol ; 17(9): 564-579, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34341569

RESUMEN

Maternal health during pregnancy plays a major role in shaping health and disease risks in the offspring. The maternal immune activation hypothesis proposes that inflammatory perturbations in utero can affect fetal neurodevelopment, and evidence from human epidemiological studies supports an association between maternal inflammation during pregnancy and offspring neurodevelopmental disorders (NDDs). Diverse maternal inflammatory factors, including obesity, asthma, autoimmune disease, infection and psychosocial stress, are associated with an increased risk of NDDs in the offspring. In addition to inflammation, epigenetic factors are increasingly recognized to operate at the gene-environment interface during NDD pathogenesis. For example, integrated brain transcriptome and epigenetic analyses of individuals with NDDs demonstrate convergent dysregulated immune pathways. In this Review, we focus on the emerging human evidence for an association between maternal immune activation and childhood NDDs, including autism spectrum disorder, attention-deficit/hyperactivity disorder and Tourette syndrome. We refer to established pathophysiological concepts in animal models, including immune signalling across the placenta, epigenetic 'priming' of offspring microglia and postnatal immune-brain crosstalk. The increasing incidence of NDDs has created an urgent need to mitigate the risk and severity of these conditions through both preventive strategies in pregnancy and novel postnatal therapies targeting disease mechanisms.


Asunto(s)
Interacción Gen-Ambiente , Trastornos del Neurodesarrollo/inmunología , Neuroinmunomodulación/inmunología , Enfermedades Neuroinflamatorias/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Animales , Femenino , Humanos , Trastornos del Neurodesarrollo/epidemiología , Trastornos del Neurodesarrollo/genética , Enfermedades Neuroinflamatorias/epidemiología , Enfermedades Neuroinflamatorias/genética , Embarazo , Efectos Tardíos de la Exposición Prenatal/epidemiología , Efectos Tardíos de la Exposición Prenatal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA