Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.763
Filtrar
1.
J Neuroinflammation ; 21(1): 188, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39090741

RESUMEN

The liver, the largest organ in the human body, plays a multifaceted role in digestion, coagulation, synthesis, metabolism, detoxification, and immune defense. Changes in liver function often coincide with disruptions in both the central and peripheral nervous systems. The intricate interplay between the nervous and immune systems is vital for maintaining tissue balance and combating diseases. Signaling molecules and pathways, including cytokines, inflammatory mediators, neuropeptides, neurotransmitters, chemoreceptors, and neural pathways, facilitate this complex communication. They establish feedback loops among diverse immune cell populations and the central, peripheral, sympathetic, parasympathetic, and enteric nervous systems within the liver. In this concise review, we provide an overview of the structural and compositional aspects of the hepatic neural and immune systems. We further explore the molecular mechanisms and pathways that govern neuroimmune communication, highlighting their significance in liver pathology. Finally, we summarize the current clinical implications of therapeutic approaches targeting neuroimmune interactions and present prospects for future research in this area.


Asunto(s)
Hepatopatías , Hígado , Neuroinmunomodulación , Humanos , Animales , Neuroinmunomodulación/fisiología , Hígado/inmunología , Hígado/patología , Hígado/metabolismo , Hepatopatías/inmunología
2.
Artículo en Inglés | MEDLINE | ID: mdl-39111563

RESUMEN

The therapeutic use of many pharmaceuticals, including small molecules and biological therapies, has been associated with the onset of psychiatric and psychological adverse events (PPAEs), posing substantial concerns to patients' health and safety. These events, which encompass mood (e.g., depression, schizophrenia, suicidal ideation) and cognitive changes (e.g., learning and memory impairment, dementia) often remain undetected until advanced stages of clinical trials or pharmacovigilance, mostly because the mechanisms underlying the onset of PPAEs remain poorly understood. In recent years, the role of neuroimmune modulation (comprising an intricate interplay between various cell types and signaling pathways) in PPAEs has garnered substantial interest. Indeed, understanding these complex interactions would substantially contribute to increase the ability to predict the potential onset of PPAEs during preclinical stages of a new drug's R&D. This review provides a comprehensive summary of the most recent advances in neuroimmune modulation-related mechanisms contributing to the onset of PPAEs and their association with specific pharmaceuticals. Reported data strongly support an association between neuroimmune modulation and the onset of PPAEs. Pharmaceuticals may target specific molecular pathways and pathway elements (e.g., cholinergic and serotonergic systems), which in turn may directly or indirectly impact the inflammatory status and the homeostasis of the brain, regulating inflammation and neuronal function. Also, modulation of the peripheral immune system by pharmaceuticals that do not permeate the blood-brain barrier (e.g., monoclonal antibodies) may alter the neuroimmunomodulatory status of the brain, leading to PPAEs. In summary, this review underscores the diverse pathways through which drugs can influence brain inflammation, shedding light on potential targeted interventions.


Asunto(s)
Trastornos Mentales , Neuroinmunomodulación , Humanos , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/fisiología , Trastornos Mentales/inmunología , Trastornos Mentales/tratamiento farmacológico , Animales
3.
J Neuroinflammation ; 21(1): 202, 2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39154174

RESUMEN

Growing evidence has implicated systemic infection as a significant risk factor for the development and advancement of Alzheimer's disease (AD). With the emergence of SARS-CoV-2 (COVID-19) and the resultant pandemic, many individuals from the same aging population vulnerable to AD suffered a severe systemic infection with potentially unidentified long-term consequences for survivors. To study the impact of COVID-19 survival on the brain's intrinsic immune system in a population also suffering from AD, we profiled post-mortem brain tissue from patients in the UF Neuromedicine Human Brain and Tissue Bank with a diagnosis of AD who survived a COVID-19 infection (COVID-AD) and contrasted our findings with AD patients who did not experience a COVID-19 infection, including a group of brain donors who passed away before arrival of SARS-CoV-2 in the United States. We assessed disease-relevant protein pathology and microglial and astrocytic markers by quantitative immunohistochemistry and supplemented these data with whole tissue gene expression analysis performed on the NanoString nCounter® platform. COVID-AD patients showed slightly elevated Aß burden in the entorhinal, fusiform, and inferior temporal cortices compared to non-COVID-AD patients, while tau pathology burden did not differ between groups. Analysis of microglia revealed a significant loss of microglial homeostasis as well as exacerbated microgliosis in COVID-AD patients compared to non-COVID-AD patients in a brain region-dependent manner. Furthermore, COVID-AD patients showed reduced cortical astrocyte numbers, independent of functional subtype. Transcriptomic analysis supported these histological findings and, in addition, identified a dysregulation of oligodendrocyte and myelination pathways in the hippocampus of COVID-AD patients. In summary, our data demonstrate a profound impact of COVID-19 infection on neuroimmune and glial pathways in AD patients persisting for months post-infection, highlighting the importance of peripheral to central neuroimmune crosstalk in neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer , COVID-19 , Homeostasis , Humanos , COVID-19/inmunología , COVID-19/complicaciones , COVID-19/patología , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Masculino , Femenino , Anciano , Anciano de 80 o más Años , Homeostasis/fisiología , Encéfalo/patología , Encéfalo/inmunología , Encéfalo/metabolismo , Neuroinmunomodulación/fisiología , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Persona de Mediana Edad , SARS-CoV-2 , Astrocitos/metabolismo , Astrocitos/inmunología , Astrocitos/patología
4.
J Headache Pain ; 25(1): 129, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39107712

RESUMEN

Migraine, a primary headache disorder whose mechanism remains incompletely understood, appears to involve the activation of the trigeminovascular system (TS) during attacks. Research suggests that inflammatory processes mediated by the immune system may play a role in migraine pathophysiology. Neuroinflammation is often associated with migraine attacks, with cytokines serving as crucial mediators in the process. Elevated levels of pro-inflammatory cytokines, such as interleukin-1 beta (IL-1ß), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α), have been observed in the blood and cerebrospinal fluid of individuals experiencing migraine attacks. These cytokines have the capacity to sensitize pain pathways in the brain, thereby increasing sensitivity to pain stimuli. This phenomenon, known as central sensitization, is believed to contribute to the intensity and persistence of migraine pain. Kynurenines, endogenous mediators of glutamatergic mechanisms, can significantly influence the pathophysiology of primary headache disorders. The kynurenine system is collectively known as the kynurenine pathway (KP), which can act on multiple receptors, such as glutamate receptors, aryl hydrocarbon receptors (AhRs), G protein-coupled receptors 35 (GPR35), and α-7 nicotinic acetylcholine (α7 nACh) receptors. These receptors are also found on various cells of the immune system, so the role of the KP in the pathomechanism of primary headaches may also be mediated through them. In this review, our goal is to show a possible link between the receptors of the KP and immune system in the context of inflammation and migraine. Migraine research in recent years has focused on neuropeptides, such as calcitonin gene-related peptide (CGRP) and pituitary adenylate cyclase-activating polypeptide (PACAP) as potential pathogenic factors and possible therapeutic approaches. These peptides share many similarities in their characteristics and roles. For instance, they exhibit potent vasodilation, occur in both the peripheral and central nervous systems, and play a role in transmitting nociception and neurogenic inflammation. The investigation of potential connections between the aforementioned neuropeptides and the kynurenine pathway could play a significant role in uncovering the pathomechanism of migraine and identifying new drug candidates.


Asunto(s)
Quinurenina , Trastornos Migrañosos , Humanos , Trastornos Migrañosos/inmunología , Trastornos Migrañosos/fisiopatología , Trastornos Migrañosos/metabolismo , Quinurenina/metabolismo , Animales , Neuroinmunomodulación/fisiología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/fisiopatología
5.
Proc Natl Acad Sci U S A ; 121(35): e2406005121, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39172786

RESUMEN

Dynamic brain immune function in individuals with posttraumatic stress disorder is rarely studied, despite evidence of peripheral immune dysfunction. Positron emission tomography brain imaging using the radiotracer [11C]PBR28 was used to measure the 18-kDa translocator protein (TSPO), a microglial marker, at baseline and 3 h after administration of lipopolysaccharide (LPS), a potent immune activator. Data were acquired in 15 individuals with PTSD and 15 age-matched controls. The PTSD group exhibited a significantly lower magnitude LPS-induced increase in TSPO availability in an a priori prefrontal-limbic circuit compared to controls. Greater anhedonic symptoms in the PTSD group were associated with a more suppressed neuroimmune response. In addition, while a reduced granulocyte-macrophage colony-stimulating factor response to LPS was observed in the PTSD group, other measured cytokine responses and self-reported sickness symptoms did not differ between groups; these findings highlight group differences in central-peripheral immune system relationships. The results of this study provide evidence of a suppressed microglia-mediated neuroimmune response to a direct immune system insult in individuals with PTSD that is associated with the severity of symptoms. They also provide further support to an emerging literature challenging traditional concepts of microglial and immune function in psychiatric disease.


Asunto(s)
Anhedonia , Microglía , Tomografía de Emisión de Positrones , Receptores de GABA , Trastornos por Estrés Postraumático , Trastornos por Estrés Postraumático/inmunología , Trastornos por Estrés Postraumático/diagnóstico por imagen , Trastornos por Estrés Postraumático/metabolismo , Humanos , Microglía/inmunología , Microglía/metabolismo , Masculino , Adulto , Tomografía de Emisión de Positrones/métodos , Femenino , Receptores de GABA/metabolismo , Lipopolisacáridos , Persona de Mediana Edad , Neuroinmunomodulación/fisiología , Encéfalo/diagnóstico por imagen , Encéfalo/inmunología , Encéfalo/metabolismo
6.
Neuroimmunomodulation ; 31(1): 157-172, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39008963

RESUMEN

BACKGROUND: That neuroimmune interaction occurs in chronic pain conditions has been established for over a century, since the discovery of neurogenic inflammation in the periphery. However, the central aspects of neuroimmune interactions have not been fully appreciated until the late 1900s, when a growing interest in how cytokines in the cerebrospinal fluid (CSF) might be relevant in chronic pain conditions emerged. Since then, the field has evolved, and nowadays neuroinflammation is considered to be involved in the pathophysiology of chronic pain. Whether or not pain conditions can be called "neuroinflammatory" is a matter of debate. This review summarizes the results from studies investigating cytokines in the CSF in various pain conditions, and critically discusses neuroimmune aspects of pain conditions using previously proposed hallmarks of neuroinflammation as a framework. SUMMARY: Fifty-two papers were summarized and their results evaluated according to (a) the level of the measured cytokines in patients compared to controls, and (b) the correlation between cytokine level and pain intensity. A subdivision based on pain type was also conducted for each of the 52 studies. A total of 49 proteins have been studied in at least 5 studies, 21 of which were upregulated in a majority of studies. IL-8 was specifically upregulated in a majority of studies of nociceptive pain conditions. Regarding correlation to pain intensity, there is a scarcity of data but 31 proteins were upregulated and correlated with pain in at least one study. Of these, 24 proteins were negatively correlated with pain, and 7 were positively correlated. None of the most studied cytokines, such as TNF, IL-1b, IL-6, IL-8, CCL2/MCP1, BDNF, or bNGF, were consistently correlated to pain. KEY MESSAGES: There is sufficient evidence to say that chronic pain conditions come with an upregulation of several cytokines. However, the majority of correlations to symptomatology seem to be negative, indicating that the cytokines might play a protective role that has not been broadly considered. Calling chronic pain conditions neuroinflammatory seems wrong; instead, a more suitable term for depicting the findings would, perhaps, be to talk about neuroimmune activation.


Asunto(s)
Dolor Crónico , Citocinas , Humanos , Citocinas/líquido cefalorraquídeo , Citocinas/inmunología , Dolor Crónico/inmunología , Dolor Crónico/líquido cefalorraquídeo , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/líquido cefalorraquídeo , Neuroinmunomodulación/fisiología , Neuroinmunomodulación/inmunología
7.
Brain Behav Immun ; 121: 56-69, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39043341

RESUMEN

Alzheimer's Disease (AD) is a neurodegenerative disease characterized by profound memory impairments, synaptic loss, neuroinflammation, and hallmark pathological markers. High-fat diet (HFD) consumption increases the risk of developing AD even after controlling for metabolic syndrome, pointing to a role of the diet itself in increasing risk. In AD, the complement system, an arm of the immune system which normally tags redundant or damaged synapses for pruning, becomes pathologically overactivated leading to tagging of healthy synapses. While the unhealthy diet to AD link is strong, the underlying mechanisms are not well understood in part due to confounding variables associated with long-term HFD which can independently influence the brain. Therefore, we experimented with a short-term diet regimen to isolate the diet's impact on brain function without causing obesity. This project investigated the effect of short-term HFD on 1) memory, 2) neuroinflammation including complement, 3) AD pathology markers, 4) synaptic markers, and 5) in vitro microglial synaptic phagocytosis in the 3xTg-AD mouse model. Following the consumption of either standard chow or HFD, 3xTg-AD and non-Tg mice were tested for memory impairments. In a separate cohort of mice, levels of hippocampal inflammatory markers, complement proteins, AD pathology markers, and synaptic markers were measured. For the last set of experiments, BV2 microglial phagocytosis of synapses was evaluated. Synaptoneurosomes isolated from the hippocampus of 3xTg-AD mice fed chow or HFD were incubated with equal numbers of BV2 microglia. The number of BV2 microglia that phagocytosed synaptoneurosomes was tracked over time with a live-cell imaging assay. Finally, we incubated BV2 microglia with a complement receptor inhibitor (NIF) and repeated the assay. Behavioral analysis showed 3xTg-AD mice had significantly impaired long-term contextual and cued fear memory compared to non-Tg mice that was further impaired by HFD. HFD significantly increased inflammatory markers and complement expression while decreasing synaptic marker expression only in 3xTg-AD mice, without altering AD pathology markers. Synaptoneurosomes from HFD-fed 3xTg-AD mice were phagocytosed at a significantly higher rate than those from chow-fed mice, suggesting the synapses were altered by HFD. The complement receptor inhibitor blocked this effect in a dose-dependent manner, demonstrating the HFD-mediated increase in phagocytosis was complement dependent. This study indicates HFD consumption increases neuroinflammation and over-activates the complement cascade in 3xTg-AD mice, resulting in poorer memory. The in vitro data point to complement as a potential mechanistic culprit and therapeutic target underlying HFD's influence in increasing cognitive vulnerability to AD.


Asunto(s)
Enfermedad de Alzheimer , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Ratones Transgénicos , Microglía , Sinapsis , Animales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/inmunología , Dieta Alta en Grasa/efectos adversos , Ratones , Sinapsis/metabolismo , Microglía/metabolismo , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/inmunología , Masculino , Proteínas del Sistema Complemento/metabolismo , Memoria/fisiología , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/inmunología , Ratones Endogámicos C57BL , Encéfalo/metabolismo , Encéfalo/inmunología , Hipocampo/metabolismo , Neuroinmunomodulación/fisiología
8.
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
10.
Pain ; 165(7): e65-e79, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38900144

RESUMEN

ABSTRACT: Recent evidence highlights the importance of the neuroimmune interface, including periphery-to-central nervous system (CNS) neuroimmune crosstalk, in chronic pain. Although neuroinflammatory processes have been implicated in central sensitization for a long time, their potential neuroprotective and analgesic effects remain relatively elusive. We have explored the relationships between cytokine expression and symptom severity, and candidates for periphery-to-CNS crosstalk. Patients with degenerative disk disease (DDD) (nociceptive pain) or patients with lumbar disk herniation (LDH) with radiculopathy (predominantly neuropathic pain) completed questionnaires regarding pain and functional disability, underwent quantitative sensory testing, and provided blood and cerebrospinal fluid (CSF) samples. Proximity extension assay (PEA) was used to measure the levels of 92 inflammatory proteins in the CSF and serum from a total of 160 patients and controls, and CSF/serum albumin quotients was calculated for patients with DDD and patients with LDH. We found signs of neuroimmune activation, in the absence of systemic inflammation. Regarding periphery-to-CNS neuroimmune crosstalk, there were significant associations between several cytokines and albumin quotient, despite the latter being primarily at subclinical levels. The cytokines CCL11, CD5, IL8, and MMP-10 were elevated in the CSF, had positive correlations between CSF and serum levels, and associated in a nonlinear manner with back, but not leg, pain intensity in the LDH, but not the DDD, group. In conclusion, we found evidence for neuroimmune activation in the CNS of both patient groups in the absence of systemic inflammation and signs of a communication between CSF and serum. Complex and disease-specific associations were found between cytokines in CSF and back pain intensity.


Asunto(s)
Dolor Crónico , Citocinas , Degeneración del Disco Intervertebral , Desplazamiento del Disco Intervertebral , Humanos , Masculino , Femenino , Citocinas/líquido cefalorraquídeo , Citocinas/sangre , Persona de Mediana Edad , Desplazamiento del Disco Intervertebral/líquido cefalorraquídeo , Desplazamiento del Disco Intervertebral/complicaciones , Desplazamiento del Disco Intervertebral/inmunología , Degeneración del Disco Intervertebral/líquido cefalorraquídeo , Degeneración del Disco Intervertebral/inmunología , Adulto , Dolor Crónico/líquido cefalorraquídeo , Dolor Crónico/inmunología , Dolor Crónico/sangre , Anciano , Vértebras Lumbares , Dimensión del Dolor/métodos , Neuroinmunomodulación/fisiología
11.
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
12.
J Neuroinflammation ; 21(1): 156, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38872143

RESUMEN

Repetitive mild traumatic brain injuries (rmTBI) sustained within a window of vulnerability can result in long term cognitive deficits, depression, and eventual neurodegeneration associated with tau pathology, amyloid beta (Aß) plaques, gliosis, and neuronal and functional loss. However, a comprehensive study relating acute changes in immune signaling and glial reactivity to neuronal changes and pathological markers after single and repetitive mTBIs is currently lacking. In the current study, we addressed the question of how repeated injuries affect the brain neuroimmune response in the acute phase of injury (< 24 h) by exposing the 3xTg-AD mouse model of tau and Aß pathology to successive (1x-5x) once-daily weight drop closed-head injuries and quantifying immune markers, pathological markers, and transcriptional profiles at 30 min, 4 h, and 24 h after each injury. We used young adult 2-4 month old 3xTg-AD mice to model the effects of rmTBI in the absence of significant tau and Aß pathology. We identified pronounced sexual dimorphism in this model, with females eliciting more diverse changes after injury compared to males. Specifically, females showed: (1) a single injury caused a decrease in neuron-enriched genes inversely correlated with inflammatory protein expression and an increase in AD-related genes within 24 h, (2) each injury significantly increased a group of cortical cytokines (IL-1α, IL-1ß, IL-2, IL-9, IL-13, IL-17, KC) and MAPK phospho-proteins (phospho-Atf2, phospho-Mek1), several of which co-labeled with neurons and correlated with phospho-tau, and (3) repetitive injury caused increased expression of genes associated with astrocyte reactivity and macrophage-associated immune function. Collectively our data suggest that neurons respond to a single injury within 24 h, while other cell types, including astrocytes, transition to inflammatory phenotypes within days of repetitive injury.


Asunto(s)
Conmoción Encefálica , Ratones Transgénicos , Animales , Ratones , Conmoción Encefálica/patología , Conmoción Encefálica/inmunología , Conmoción Encefálica/metabolismo , Conmoción Encefálica/complicaciones , Femenino , Masculino , Modelos Animales de Enfermedad , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Proteínas tau/metabolismo , Proteínas tau/genética , Neuroinmunomodulación/fisiología , Ratones Endogámicos C57BL , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/inmunología , Caracteres Sexuales
13.
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
14.
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
15.
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
16.
Immunity ; 57(4): 815-831, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38599172

RESUMEN

The sensory nervous system possesses the ability to integrate exogenous threats and endogenous signals to mediate downstream effector functions. Sensory neurons have been shown to activate or suppress host defense and immunity against pathogens, depending on the tissue and disease state. Through this lens, pro- and anti-inflammatory neuroimmune effector functions can be interpreted as evolutionary adaptations by host or pathogen. Here, we discuss recent and impactful examples of neuroimmune circuitry that regulate tissue homeostasis, autoinflammation, and host defense. Apparently paradoxical or conflicting reports in the literature also highlight the complexity of neuroimmune interactions that may depend on tissue- and microbe-specific cues. These findings expand our understanding of the nuanced mechanisms and the greater context of sensory neurons in innate immunity.


Asunto(s)
Inmunidad Innata , Células Receptoras Sensoriales , Inmunidad Innata/fisiología , Neuroinmunomodulación/fisiología , Homeostasis
17.
Neuron ; 112(12): 1959-1977.e10, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38614103

RESUMEN

Microglial calcium signaling is rare in a baseline state but strongly engaged during early epilepsy development. The mechanism(s) governing microglial calcium signaling are not known. By developing an in vivo uridine diphosphate (UDP) fluorescent sensor, GRABUDP1.0, we discovered that UDP release is a conserved response to seizures and excitotoxicity across brain regions. UDP can signal through the microglial-enriched P2Y6 receptor to increase calcium activity during epileptogenesis. P2Y6 calcium activity is associated with lysosome biogenesis and enhanced production of NF-κB-related cytokines. In the hippocampus, knockout of the P2Y6 receptor prevents microglia from fully engulfing neurons. Attenuating microglial calcium signaling through calcium extruder ("CalEx") expression recapitulates multiple features of P2Y6 knockout, including reduced lysosome biogenesis and phagocytic interactions. Ultimately, P2Y6 knockout mice retain more CA3 neurons and better cognitive task performance during epileptogenesis. Our results demonstrate that P2Y6 signaling impacts multiple aspects of myeloid cell immune function during epileptogenesis.


Asunto(s)
Señalización del Calcio , Epilepsia , Ratones Noqueados , Microglía , Fagocitosis , Receptores Purinérgicos P2 , Animales , Microglía/metabolismo , Microglía/inmunología , Ratones , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2/genética , Señalización del Calcio/fisiología , Epilepsia/metabolismo , Epilepsia/inmunología , Epilepsia/genética , Uridina Difosfato/metabolismo , Lisosomas/metabolismo , Neuronas/metabolismo , Ratones Endogámicos C57BL , Masculino , Hipocampo/metabolismo , Neuroinmunomodulación/fisiología
18.
Curr Hypertens Rep ; 26(7): 339-347, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38613621

RESUMEN

PURPOSE OF REVIEW: Cardiovascular disease (CVD) is a leading cause of death and chronic disability worldwide. Yet, despite extensive intervention strategies the number of persons affected by CVD continues to rise. Thus, there is great interest in unveiling novel mechanisms that may lead to new treatments. Considering this dilemma, recent focus has turned to the neuroimmune mechanisms involved in CVD pathology leading to a deeper understanding of the brain's involvement in disease pathology. This review provides an overview of new and salient findings regarding the neuroimmune mechanisms that contribute to CVD. RECENT FINDINGS: The brain contains neuroimmune niches comprised of glia in the parenchyma and immune cells at the brain's borders, and there is strong evidence that these neuroimmune niches are important in both health and disease. Mechanistic studies suggest that the activation of glia and immune cells in these niches modulates CVD progression in hypertension and heart failure and contributes to the inevitable end-organ damage to the brain. This review provides evidence supporting the role of neuroimmune niches in CVD progression. However, additional research is needed to understand the effects of prolonged neuroimmune activation on brain function.


Asunto(s)
Encéfalo , Enfermedades Cardiovasculares , Neuroinmunomodulación , Humanos , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/fisiopatología , Neuroinmunomodulación/fisiología , Encéfalo/inmunología , Encéfalo/fisiopatología , Encéfalo/patología , Neuroglía/inmunología , Animales
19.
Am J Physiol Gastrointest Liver Physiol ; 326(6): G712-G725, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38626403

RESUMEN

Gut physiology is the epicenter of a web of internal communication systems (i.e., neural, immune, hormonal) mediated by cell-cell contacts, soluble factors, and external influences, such as the microbiome, diet, and the physical environment. Together these provide the signals that shape enteric homeostasis and, when they go awry, lead to disease. Faced with the seemingly paradoxical tasks of nutrient uptake (digestion) and retarding pathogen invasion (host defense), the gut integrates interactions between a variety of cells and signaling molecules to keep the host nourished and protected from pathogens. When the system fails, the outcome can be acute or chronic disease, often labeled as "idiopathic" in nature (e.g., irritable bowel syndrome, inflammatory bowel disease). Here we underscore the importance of a holistic approach to gut physiology, placing an emphasis on intercellular connectedness, using enteric neuroimmunophysiology as the paradigm. The goal of this opinion piece is to acknowledge the pace of change brought to our field via single-cell and -omic methodologies and other techniques such as cell lineage tracing, transgenic animal models, methods for culturing patient tissue, and advanced imaging. We identify gaps in the field and hope to inspire and challenge colleagues to take up the mantle and advance awareness of the subtleties, intricacies, and nuances of intestinal physiology in health and disease by defining communication pathways between gut resident cells, those recruited from the circulation, and "external" influences such as the central nervous system and the gut microbiota.


Asunto(s)
Microbioma Gastrointestinal , Tracto Gastrointestinal , Humanos , Animales , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/fisiología , Tracto Gastrointestinal/microbiología , Microbioma Gastrointestinal/fisiología , Neuroinmunomodulación/fisiología , Sistema Nervioso Entérico/fisiología , Sistema Nervioso Entérico/inmunología
20.
Neuroimmunomodulation ; 31(1): 66-77, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38471475

RESUMEN

BACKGROUND: Evolutionary medicine builds on evolutionary biology and explains why natural selection has left us vulnerable to disease. Unfortunately, several misunderstandings exist in the medical literature about the levels and mechanisms of evolution. Reasons for these problems start from the lack of teaching evolutionary biology in medical schools. A common mistake is to assume that "traits must benefit the species, as otherwise the species would have gone extinct in the past" confusing evolutionary history (phylogeny) with evolutionary function (fitness). SUMMARY: Here we summarise some basic aspects of evolutionary medicine by pointing out: (1) Evolution has no aim. (2) For adaptive evolution to occur, a trait does not have to be beneficial to its carrier throughout its entire life. (3) Not every single individual carrying an adaptive trait needs to have higher than average fitness. (4) Traits do not evolve for the benefit of the species. Using examples from the field of neuroimmunomodulation like sickness behaviour (nervous system), testosterone (hormones), and cytokines (immunity), we show how misconceptions arise from not differentiating between the explanatory categories of phylogeny (evolutionary history) and evolutionary function (fitness). KEY MESSAGES: Evolution has no aim but is an automatism that does not function for the benefit of the species. In evolution, successful individuals are those that maximise the transmission of their genes, and health and survival are just strategies to have the opportunity to do so. Thus, a trait enabling survival of the individual until reproductive age will spread even if at later age the same trait leads to disease and death. Natural and sexual selection do not select for traits that benefit the health or happiness of the individual, but for traits that increase inclusive fitness even if this increases human suffering. In contrast, our humane aim is to increase individual well-being. Evolutionary medicine can help us achieve this aim against evolutionary constraints.


Asunto(s)
Evolución Biológica , Neuroinmunomodulación , Humanos , Neuroinmunomodulación/fisiología , Animales , Filogenia , Selección Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA