Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 627(8002): 165-173, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38326613

RESUMO

The arachnoid barrier delineates the border between the central nervous system and dura mater. Although the arachnoid barrier creates a partition, communication between the central nervous system and the dura mater is crucial for waste clearance and immune surveillance1,2. How the arachnoid barrier balances separation and communication is poorly understood. Here, using transcriptomic data, we developed transgenic mice to examine specific anatomical structures that function as routes across the arachnoid barrier. Bridging veins create discontinuities where they cross the arachnoid barrier, forming structures that we termed arachnoid cuff exit (ACE) points. The openings that ACE points create allow the exchange of fluids and molecules between the subarachnoid space and the dura, enabling the drainage of cerebrospinal fluid and limited entry of molecules from the dura to the subarachnoid space. In healthy human volunteers, magnetic resonance imaging tracers transit along bridging veins in a similar manner to access the subarachnoid space. Notably, in neuroinflammatory conditions such as experimental autoimmune encephalomyelitis, ACE points also enable cellular trafficking, representing a route for immune cells to directly enter the subarachnoid space from the dura mater. Collectively, our results indicate that ACE points are a critical part of the anatomy of neuroimmune communication in both mice and humans that link the central nervous system with the dura and its immunological diversity and waste clearance systems.


Assuntos
Aracnoide-Máter , Encéfalo , Dura-Máter , Animais , Humanos , Camundongos , Aracnoide-Máter/anatomia & histologia , Aracnoide-Máter/irrigação sanguínea , Aracnoide-Máter/imunologia , Aracnoide-Máter/metabolismo , Transporte Biológico , Encéfalo/anatomia & histologia , Encéfalo/irrigação sanguínea , Encéfalo/imunologia , Encéfalo/metabolismo , Dura-Máter/anatomia & histologia , Dura-Máter/irrigação sanguínea , Dura-Máter/imunologia , Dura-Máter/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Perfilação da Expressão Gênica , Imageamento por Ressonância Magnética , Camundongos Transgênicos , Espaço Subaracnóideo/anatomia & histologia , Espaço Subaracnóideo/irrigação sanguínea , Espaço Subaracnóideo/imunologia , Espaço Subaracnóideo/metabolismo , Líquido Cefalorraquidiano/metabolismo , Veias/metabolismo
2.
Neuron ; 111(20): 3244-3254.e8, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37582366

RESUMO

Aging is a complex process involving various systems and behavioral changes. Altered immune regulation, dysbiosis, oxidative stress, and sleep decline are common features of aging, but their interconnection is poorly understood. Using Drosophila, we discover that IM33, a novel immune modulator, and its mammalian homolog, secretory leukocyte protease inhibitor (SLPI), are upregulated in old flies and old mice, respectively. Knockdown of IM33 in glia elevates the gut reactive oxygen species (ROS) level and alters gut microbiota composition, including increased Lactiplantibacillus plantarum abundance, leading to a shortened lifespan. Additionally, dysbiosis induces sleep fragmentation through the activation of insulin-producing cells in the brain, which is mediated by the binding of Lactiplantibacillus plantarum-produced DAP-type peptidoglycan to the peptidoglycan recognition protein LE (PGRP-LE) receptor. Therefore, IM33 plays a role in the glia-microbiota-neuronal axis, connecting neuroinflammation, dysbiosis, and sleep decline during aging. Identifying molecular mediators of these processes could lead to the development of innovative strategies for extending lifespan.


Assuntos
Proteínas de Drosophila , Longevidade , Inibidor Secretado de Peptidases Leucocitárias , Animais , Camundongos , Drosophila/fisiologia , Proteínas de Drosophila/metabolismo , Disbiose , Neuroglia/metabolismo , Inibidor Secretado de Peptidases Leucocitárias/metabolismo
3.
Neuron ; 111(14): 2155-2169.e9, 2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37148871

RESUMO

Spinal cord injury (SCI) causes lifelong debilitating conditions. Previous works demonstrated the essential role of the immune system in recovery after SCI. Here, we explored the temporal changes of the response after SCI in young and aged mice in order to characterize multiple immune populations within the mammalian spinal cord. We revealed substantial infiltration of myeloid cells to the spinal cord in young animals, accompanied by changes in the activation state of microglia. In contrast, both processes were blunted in aged mice. Interestingly, we discovered the formation of meningeal lymphatic structures above the lesion site, and their role has not been examined after contusive injury. Our transcriptomic data predicted lymphangiogenic signaling between myeloid cells in the spinal cord and lymphatic endothelial cells (LECs) in the meninges after SCI. Together, our findings delineate how aging affects the immune response following SCI and highlight the participation of the spinal cord meninges in supporting vascular repair.


Assuntos
Células Endoteliais , Traumatismos da Medula Espinal , Camundongos , Animais , Células Endoteliais/patologia , Traumatismos da Medula Espinal/patologia , Medula Espinal/patologia , Microglia/patologia , Células Mieloides , Mamíferos
4.
J Exp Med ; 220(7)2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-37027179

RESUMO

The meningeal lymphatic network enables the drainage of cerebrospinal fluid (CSF) and facilitates the removal of central nervous system (CNS) waste. During aging and in Alzheimer's disease, impaired meningeal lymphatic drainage promotes the buildup of toxic misfolded proteins in the CNS. Reversing this age-related dysfunction represents a promising strategy to augment CNS waste clearance; however, the mechanisms underlying this decline remain elusive. Here, we demonstrate that age-related alterations in meningeal immunity underlie this lymphatic impairment. Single-cell RNA sequencing of meningeal lymphatic endothelial cells from aged mice revealed their response to IFNγ, which was increased in the aged meninges due to T cell accumulation. Chronic elevation of meningeal IFNγ in young mice via AAV-mediated overexpression attenuated CSF drainage-comparable to the deficits observed in aged mice. Therapeutically, IFNγ neutralization alleviated age-related impairments in meningeal lymphatic function. These data suggest manipulation of meningeal immunity as a viable approach to normalize CSF drainage and alleviate the neurological deficits associated with impaired waste removal.


Assuntos
Doença de Alzheimer , Vasos Linfáticos , Camundongos , Animais , Células Endoteliais , Sistema Nervoso Central , Meninges , Sistema Linfático , Encéfalo/fisiologia
5.
Nat Commun ; 14(1): 2314, 2023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-37085492

RESUMO

Genetic studies of Alzheimer disease (AD) have prioritized variants in genes related to the amyloid cascade, lipid metabolism, and neuroimmune modulation. However, the cell-specific effect of variants in these genes is not fully understood. Here, we perform single-nucleus RNA-sequencing (snRNA-seq) on nearly 300,000 nuclei from the parietal cortex of AD autosomal dominant (APP and PSEN1) and risk-modifying variant (APOE, TREM2 and MS4A) carriers. Within individual cell types, we capture genes commonly dysregulated across variant groups. However, specific transcriptional states are more prevalent within variant carriers. TREM2 oligodendrocytes show a dysregulated autophagy-lysosomal pathway, MS4A microglia have dysregulated complement cascade genes, and APOEε4 inhibitory neurons display signs of ferroptosis. All cell types have enriched states in autosomal dominant carriers. We leverage differential expression and single-nucleus ATAC-seq to map GWAS signals to effector cell types including the NCK2 signal to neurons in addition to the initially proposed microglia. Overall, our results provide insights into the transcriptional diversity resulting from AD genetic architecture and cellular heterogeneity. The data can be explored on the online browser ( http://web.hararilab.org/SNARE/ ).


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Heterozigoto , Microglia/metabolismo , Lobo Parietal/metabolismo , RNA/metabolismo
6.
Nature ; 611(7936): 585-593, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36352225

RESUMO

Macrophages are important players in the maintenance of tissue homeostasis1. Perivascular and leptomeningeal macrophages reside near the central nervous system (CNS) parenchyma2, and their role in CNS physiology has not been sufficiently well studied. Given their continuous interaction with the cerebrospinal fluid (CSF) and strategic positioning, we refer to these cells collectively as parenchymal border macrophages (PBMs). Here we demonstrate that PBMs regulate CSF flow dynamics. We identify a subpopulation of PBMs that express high levels of CD163 and LYVE1 (scavenger receptor proteins), closely associated with the brain arterial tree, and show that LYVE1+ PBMs regulate arterial motion that drives CSF flow. Pharmacological or genetic depletion of PBMs led to accumulation of extracellular matrix proteins, obstructing CSF access to perivascular spaces and impairing CNS perfusion and clearance. Ageing-associated alterations in PBMs and impairment of CSF dynamics were restored after intracisternal injection of macrophage colony-stimulating factor. Single-nucleus RNA sequencing data obtained from patients with Alzheimer's disease (AD) and from non-AD individuals point to changes in phagocytosis, endocytosis and interferon-γ signalling on PBMs, pathways that are corroborated in a mouse model of AD. Collectively, our results identify PBMs as new cellular regulators of CSF flow dynamics, which could be targeted pharmacologically to alleviate brain clearance deficits associated with ageing and AD.


Assuntos
Sistema Nervoso Central , Líquido Cefalorraquidiano , Macrófagos , Tecido Parenquimatoso , Animais , Camundongos , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Líquido Cefalorraquidiano/metabolismo , Macrófagos/fisiologia , Meninges/citologia , Reologia , Proteínas da Matriz Extracelular/metabolismo , Envelhecimento/metabolismo , Fagocitose , Endocitose , Interferon gama/metabolismo , Tecido Parenquimatoso/citologia , Humanos
7.
Neuron ; 110(11): 1788-1805.e10, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35381189

RESUMO

Resolving glial contributions to Alzheimer's disease (AD) is necessary because changes in neuronal function, such as reduced synaptic density, altered electrophysiological properties, and degeneration, are not entirely cell autonomous. To improve understanding of transcriptomic heterogeneity in glia during AD, we used single-nuclei RNA sequencing (snRNA-seq) to characterize astrocytes and oligodendrocytes from apolipoprotein (APOE) Ɛ2/3 human AD and age- and genotype-matched non-symptomatic (NS) brains. We enriched astrocytes before sequencing and characterized pathology from the same location as the sequenced material. We characterized baseline heterogeneity in both astrocytes and oligodendrocytes and identified global and subtype-specific transcriptomic changes between AD and NS astrocytes and oligodendrocytes. We also took advantage of recent human and mouse spatial transcriptomics resources to localize heterogeneous astrocyte subtypes to specific regions in the healthy and inflamed brain. Finally, we integrated our data with published AD snRNA-seq datasets, highlighting the power of combining datasets to resolve previously unidentifiable astrocyte subpopulations.


Assuntos
Doença de Alzheimer , Astrócitos , Doença de Alzheimer/patologia , Animais , Astrócitos/patologia , Humanos , Camundongos , Neuroglia/patologia , Oligodendroglia/patologia , RNA Nuclear Pequeno , Transcriptoma
8.
Nat Neurosci ; 25(5): 555-560, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35301477

RESUMO

It remains unclear how immune cells from skull bone marrow niches are recruited to the meninges. Here we report that cerebrospinal fluid (CSF) accesses skull bone marrow via dura-skull channels, and CSF proteins signal onto diverse cell types within the niches. After spinal cord injury, CSF-borne cues promote myelopoiesis and egress of myeloid cells into meninges. This reveals a mechanism of CNS-to-bone-marrow communication via CSF that regulates CNS immune responses.


Assuntos
Medula Óssea , Crânio , Medula Óssea/fisiologia , Líquido Cefalorraquidiano , Cabeça , Meninges , Células Mieloides/metabolismo
9.
Nat Aging ; 2(8): 704-713, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-37065770

RESUMO

Peripheral inflammation triggers a transient, well-defined set of behavioral changes known as sickness behavior1-3, but the mechanisms by which inflammatory signals originating in the periphery alter activity in the brain remain obscure. Emerging evidence has established meningeal lymphatic vasculature as an important interface between the central nervous system (CNS) and the immune system, responsible for facilitating brain solute clearance and perfusion by cerebrospinal fluid (CSF)4,5. Here, we demonstrate that meningeal lymphatics both assist microglial activation and support the behavioral response to peripheral inflammation. Ablation of meningeal lymphatics results in a heightened behavioral response to IL-1ß-induced inflammation and a dampened transcriptional and morphological microglial phenotype. Moreover, our findings support a role for microglia in tempering the severity of sickness behavior with specific relevance to aging-related meningeal lymphatic dysfunction. Transcriptional profiling of brain myeloid cells shed light on the impact of meningeal lymphatic dysfunction on microglial activation. Furthermore, we demonstrate that experimental enhancement of meningeal lymphatic function in aged mice is sufficient to reduce the severity of exploratory abnormalities but not pleasurable consummatory behavior. Finally, we identify dysregulated genes and biological pathways, common to both experimental meningeal lymphatic ablation and aging, in microglia responding to peripheral inflammation that may result from age-related meningeal lymphatic dysfunction.


Assuntos
Vasos Linfáticos , Microglia , Camundongos , Animais , Microglia/metabolismo , Meninges , Sistema Nervoso Central/anatomia & histologia , Vasos Linfáticos/anatomia & histologia , Inflamação/genética
10.
Neuron ; 109(22): 3609-3618.e9, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34793707

RESUMO

Mechanisms governing how immune cells and their derived molecules impact homeostatic brain function are still poorly understood. Here, we elucidate neuronal mechanisms underlying T cell effects on synaptic function and episodic memory. Depletion of CD4 T cells led to memory deficits and impaired long-term potentiation. Severe combined immune-deficient mice exhibited amnesia, which was reversible by repopulation with T cells from wild-type but not from IL-4-knockout mice. Behaviors impacted by T cells were mediated via IL-4 receptors expressed on neurons. Exploration of snRNA-seq of neurons participating in memory processing provided insights into synaptic organization and plasticity-associated pathways regulated by immune cells. IL-4Rα knockout in inhibitory (but not in excitatory) neurons was sufficient to impair contextual fear memory, and snRNA-seq from these mice pointed to IL-4-driven regulation of synaptic function in promoting memory. These findings provide new insights into complex neuroimmune interactions at the transcriptional and functional levels in neurons under physiological conditions.


Assuntos
Plasticidade Neuronal , Linfócitos T , Animais , Neurônios GABAérgicos , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Memória/fisiologia , Camundongos , Camundongos Knockout , Plasticidade Neuronal/fisiologia
11.
Science ; 373(6553)2021 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-34083447

RESUMO

The meninges are a membranous structure enveloping the central nervous system (CNS) that host a rich repertoire of immune cells mediating CNS immune surveillance. Here, we report that the mouse meninges contain a pool of monocytes and neutrophils supplied not from the blood but by adjacent skull and vertebral bone marrow. Under pathological conditions, including spinal cord injury and neuroinflammation, CNS-infiltrating myeloid cells can originate from brain borders and display transcriptional signatures distinct from their blood-derived counterparts. Thus, CNS borders are populated by myeloid cells from adjacent bone marrow niches, strategically placed to supply innate immune cells under homeostatic and pathological conditions. These findings call for a reinterpretation of immune-cell infiltration into the CNS during injury and autoimmunity and may inform future therapeutic approaches that harness meningeal immune cells.


Assuntos
Células da Medula Óssea/fisiologia , Doenças do Sistema Nervoso Central/imunologia , Sistema Nervoso Central/imunologia , Meninges/imunologia , Células Mieloides/fisiologia , Crânio/anatomia & histologia , Coluna Vertebral/anatomia & histologia , Animais , Medula Óssea/fisiologia , Encéfalo/citologia , Encéfalo/imunologia , Encéfalo/fisiologia , Movimento Celular , Sistema Nervoso Central/citologia , Doenças do Sistema Nervoso Central/patologia , Dura-Máter/citologia , Dura-Máter/imunologia , Dura-Máter/fisiologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Homeostase , Meninges/citologia , Meninges/fisiologia , Camundongos , Monócitos/fisiologia , Neutrófilos/fisiologia , Medula Espinal/citologia , Medula Espinal/imunologia , Medula Espinal/fisiologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/patologia
12.
Sci Adv ; 7(21)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34020948

RESUMO

Aging leads to a progressive deterioration of meningeal lymphatics and peripheral immunity, which may accelerate cognitive decline. We hypothesized that an age-related reduction in C-C chemokine receptor type 7 (CCR7)-dependent egress of immune cells through the lymphatic vasculature mediates some aspects of brain aging and potentially exacerbates cognitive decline and Alzheimer's disease-like brain ß-amyloid (Aß) pathology. We report a reduction in CCR7 expression by meningeal T cells in old mice that is linked to increased effector and regulatory T cells. Hematopoietic CCR7 deficiency mimicked the aging-associated changes in meningeal T cells and led to reduced glymphatic influx and cognitive impairment. Deletion of CCR7 in 5xFAD transgenic mice resulted in deleterious neurovascular and microglial activation, along with increased Aß deposition in the brain. Treating old mice with anti-CD25 antibodies alleviated the exacerbated meningeal regulatory T cell response and improved cognitive function, highlighting the therapeutic potential of modulating meningeal immunity to fine-tune brain function in aging and in neurodegenerative diseases.

13.
Nature ; 593(7858): 255-260, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33911285

RESUMO

Alzheimer's disease (AD) is the most prevalent cause of dementia1. Although there is no effective treatment for AD, passive immunotherapy with monoclonal antibodies against amyloid beta (Aß) is a promising therapeutic strategy2,3. Meningeal lymphatic drainage has an important role in the accumulation of Aß in the brain4, but it is not known whether modulation of meningeal lymphatic function can influence the outcome of immunotherapy in AD. Here we show that ablation of meningeal lymphatic vessels in 5xFAD mice (a mouse model of amyloid deposition that expresses five mutations found in familial AD) worsened the outcome of mice treated with anti-Aß passive immunotherapy by exacerbating the deposition of Aß, microgliosis, neurovascular dysfunction, and behavioural deficits. By contrast, therapeutic delivery of vascular endothelial growth factor C improved clearance of Aß by monoclonal antibodies. Notably, there was a substantial overlap between the gene signature of microglia from 5xFAD mice with impaired meningeal lymphatic function and the transcriptional profile of activated microglia from the brains of individuals with AD. Overall, our data demonstrate that impaired meningeal lymphatic drainage exacerbates the microglial inflammatory response in AD and that enhancement of meningeal lymphatic function combined with immunotherapies could lead to better clinical outcomes.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Imunoterapia , Vasos Linfáticos/imunologia , Meninges/imunologia , Microglia/imunologia , Envelhecimento/efeitos dos fármacos , Envelhecimento/imunologia , Doença de Alzheimer/genética , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/efeitos dos fármacos , Animais , Anticorpos Monoclonais Humanizados/imunologia , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Modelos Animais de Doenças , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/imunologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Masculino , Meninges/irrigação sanguínea , Meninges/citologia , Camundongos , Microglia/citologia , Microglia/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/farmacologia
14.
Cell ; 184(4): 1000-1016.e27, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33508229

RESUMO

Despite the established dogma of central nervous system (CNS) immune privilege, neuroimmune interactions play an active role in diverse neurological disorders. However, the precise mechanisms underlying CNS immune surveillance remain elusive; particularly, the anatomical sites where peripheral adaptive immunity can sample CNS-derived antigens and the cellular and molecular mediators orchestrating this surveillance. Here, we demonstrate that CNS-derived antigens in the cerebrospinal fluid (CSF) accumulate around the dural sinuses, are captured by local antigen-presenting cells, and are presented to patrolling T cells. This surveillance is enabled by endothelial and mural cells forming the sinus stromal niche. T cell recognition of CSF-derived antigens at this site promoted tissue resident phenotypes and effector functions within the dural meninges. These findings highlight the critical role of dural sinuses as a neuroimmune interface, where brain antigens are surveyed under steady-state conditions, and shed light on age-related dysfunction and neuroinflammatory attack in animal models of multiple sclerosis.


Assuntos
Cavidades Cranianas/imunologia , Cavidades Cranianas/fisiologia , Dura-Máter/imunologia , Dura-Máter/fisiologia , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos/líquido cefalorraquidiano , Senescência Celular , Quimiocina CXCL12/farmacologia , Dura-Máter/irrigação sanguínea , Feminino , Homeostase , Humanos , Imunidade , Masculino , Camundongos Endogâmicos C57BL , Fenótipo , Células Estromais/citologia , Linfócitos T/citologia
15.
PM R ; 10(7): 724-729, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29288142

RESUMO

BACKGROUND: For the majority of patients, spinal surgery is an elective treatment. The decision as to whether and when to pursue surgery is complicated and influenced by myriad factors, including pain intensity and duration, impact on functional activities, referring physician recommendation, and surgeon preference. By understanding the factors that lead a patient to choose surgery, we may better understand the decision-making process, improve outcomes, and provide more effective care. OBJECTIVE: To investigate the relationship between patient-reported outcome measures (PROMs) at initial physiatry clinic consultation and subsequent decision to pursue surgical treatment. We hypothesized that measures of function, pain, and mental health might identify which patients eventually elect to pursue surgical management. DESIGN: Retrospective chart review study. SETTING: Physiatry spine clinic in a tertiary hospital. PATIENTS: A total of 395 consecutive patients meeting our inclusion criteria were assessed for the presence of chronic pain, self-perceived disability, history of prior spinal surgery, and provision of chronic opioid therapy at the time of their initial visit to the integrated spine clinic. METHODS: Retrospective chart review of all patients presenting to our spine clinic between August 1, 2014, and July 31, 2015, was performed. At the initial spine clinic consultation, patients were asked to complete the General Anxiety Disorder-7 (GAD-7), Patient Health Questionnaire-8 (PHQ-8), Oswestry Disability Index (ODI), and Patient-Reported Outcomes Measurement Information System (PROMIS) 10-item short-form questionnaire. The primary outcome was surgical intervention within 18 months from their first visit to the integrated spine clinic. We surveyed all patient records until February 2017 for CPT codes associated with spinal surgery, excluding from analysis those patients who were lost to follow-up within 1 year of the index visit. Analysis focused on the risk of spinal surgery, with data points treated as both continuous and categorical variables. We used logistic regression models to determine whether PROMs, either alone or in combination, predicted later decision to pursue surgical intervention. MAIN OUTCOME MEASUREMENTS: Decision to pursue spinal surgery. RESULTS: The baseline PROM scores spanning functional, mental health, and pain domains were collected for 94% of the patients presenting to our spine program during the interval of this study. In total, 146 patients were excluded because of missing patient-reported outcome data or less than 1 year of follow-up, leaving 395 patients for analysis. Of these, 40.3% were male with a median age of 58 years, 4.6% presented with a history of prior spinal surgery, and 3.8% were being treated with chronic opioids at their initial consultation. Male gender (P = .01) and older age (P = .05) were associated with subsequent surgery, but no relationship was observed between measured patient-reported outcomes and decision to undergo spinal surgery within 18 months of the index visit. CONCLUSIONS: Contrary to our hypothesis, this analysis demonstrates that the PROMs evaluated in this study, alone are insufficient to identify patients who may elect to pursue spinal surgery. Male gender and increasing age correlate with decision for later spinal surgery. LEVEL OF EVIDENCE: II.


Assuntos
Dor Crônica/reabilitação , Procedimentos Ortopédicos , Medidas de Resultados Relatados pelo Paciente , Qualidade de Vida , Sistema de Registros , Doenças da Coluna Vertebral/cirurgia , Adulto , Idoso , Dor Crônica/diagnóstico , Dor Crônica/etiologia , Avaliação da Deficiência , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Medição da Dor , Período Pré-Operatório , Estudos Retrospectivos , Doenças da Coluna Vertebral/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...