Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.297
Filtrar
1.
J Neuroinflammation ; 21(1): 268, 2024 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-39428507

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease affecting the central nervous system (CNS) with the immune system attacking myelin sheaths leading to neuronal death. While several disease-modifying therapies are available to treat MS, these therapies are not universally effective and do not stop disease progression. More personalized long-term treatment options that target specific aspects of the disease, such as reducing relapse frequency, delaying disability accumulation, and addressing symptoms that impact daily functioning, as well as therapies that can promote neuroprotection and repair are needed. Chimeric Antigen Receptor (CAR) Tcell therapies have revolutionized cancer treatment by intravenously (IV) administering a defined dose of T cells with high specificity provided by the CAR. An autologous CAR T cell therapy using suppressive regulatory T cells (Tregs) inducing long-lasting tolerance would be the ideal treatment for patients. Hence, we expanded the application of CAR-T cells by introducing a CAR into Tregs to treat MS patients. We developed a myelin oligodendrocyte glycoprotein (MOG)-specific CAR Treg cell therapy for patients with MS. MOG is expressed on the outer membrane of the myelin sheath, the insulating layer the forms around nerves, making it an ideal target for CAR Treg therapy. Our lead candidate is a 2nd generation CAR, composed of an anti-MOG scFv screened from a large human library. In vitro, we demonstrated CAR-dependent functionality and showed efficacy in vivo using a passive EAE mouse model. Additionally, the MOG-CAR Tregs have very low tonic signaling with a desirable signal-to-noise ratio resulting in a highly potent CAR. In summary our data suggest that MOG-CAR Tregs are a promising MS treatment option with the potential to induce long-lasting tolerance in patients.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Glicoproteína Mielina-Oligodendrócito , Linfocitos T Reguladores , Animales , Linfocitos T Reguladores/inmunología , Humanos , Glicoproteína Mielina-Oligodendrócito/inmunología , Ratones , Esclerosis Múltiple/terapia , Esclerosis Múltiple/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Encefalomielitis Autoinmune Experimental/inmunología , Receptores Quiméricos de Antígenos/inmunología , Inmunoterapia Adoptiva/métodos
2.
Pharmacol Rev ; 76(6): 1133-1158, 2024 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-39406508

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS), with a putative autoimmune origin and complex pathogenesis. Modification of the natural history of MS by reducing relapses and slowing disability accumulation was first attained in the 1990 s with the development of the first-generation disease-modifying therapies. Glatiramer acetate (GA), a copolymer of L-alanine, L-lysine, L-glutamic acid, and L-tyrosine, was discovered due to its ability to suppress the animal model of MS, experimental autoimmune encephalomyelitis. Extensive clinical trials and long-term assessments established the efficacy and the safety of GA. Furthermore, studies of the therapeutic processes induced by GA in animal models and in MS patients indicate that GA affects various levels of the innate and the adaptive immune response, generating deviation from proinflammatory to anti-inflammatory pathways. This includes competition for binding to antigen presenting cells; driving dendritic cells, monocytes, and B-cells toward anti-inflammatory responses; and stimulating T-helper 2 and T-regulatory cells. The immune cells stimulated by GA reach the CNS and secrete in situ anti-inflammatory cytokines alleviating the pathological processes. Furthermore, cumulative findings reveal that in addition to its immunomodulatory effect, GA promotes neuroprotective repair processes such as neurotrophic factors secretion, remyelination, and neurogenesis. This review aims to provide an overview of MS pathology diagnosis and treatment as well as the diverse mechanism of action of GA. SIGNIFICANCE STATEMENT: Understanding the complex MS immune pathogenesis provided multiple targets for therapeutic intervention, resulting in a plethora of agents, with various mechanisms of action, efficacy, and safety profiles. However, promoting repair beyond the body's limited spontaneous extent is still a major challenge. GA, one of the first approved disease-modifying therapies, induces diverse immunomodulatory effects. Furthermore, GA treatment results in elevated neurotrophic factors secretion, remyelination and neurogenesis, supporting the notion that immunomodulatory treatment can support in situ a growth-promoting and repair environment.


Asunto(s)
Acetato de Glatiramer , Esclerosis Múltiple , Acetato de Glatiramer/farmacología , Acetato de Glatiramer/uso terapéutico , Humanos , Animales , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Inmunomodulación/efectos de los fármacos , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Agentes Inmunomoduladores/farmacología , Agentes Inmunomoduladores/uso terapéutico
3.
Int J Mol Sci ; 25(19)2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39408955

RESUMEN

The blood-brain barrier (BBB) acts as a structural and functional barrier for brain homeostasis. This review highlights the pathological contribution of BBB dysfunction to neuroimmunological diseases, including multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD), autoimmune encephalitis (AE), and paraneoplastic neurological syndrome (PNS). The transmigration of massive lymphocytes across the BBB caused by the activation of cell adhesion molecules is involved in the early phase of MS, and dysfunction of the cortical BBB is associated with the atrophy of gray matter in the late phase of MS. At the onset of NMOSD, increased permeability of the BBB causes the entry of circulating AQP4 autoantibodies into the central nervous system (CNS). Recent reports have shown the importance of glucose-regulated protein (GRP) autoantibodies as BBB-reactive autoantibodies in NMOSD, which induce antibody-mediated BBB dysfunction. BBB breakdown has also been observed in MOGAD, NPSLE, and AE with anti-NMDAR antibodies. Our recent report demonstrated the presence of GRP78 autoantibodies in patients with MOGAD and the molecular mechanism responsible for GRP78 autoantibody-mediated BBB impairment. Disruption of the BBB may explain the symptoms in the brain and cerebellum in the development of PNS, as it induces the entry of pathogenic autoantibodies or lymphocytes into the CNS through autoimmunity against tumors in the periphery. GRP78 autoantibodies were detected in paraneoplastic cerebellar degeneration and Lambert-Eaton myasthenic syndrome, and they were associated with cerebellar ataxia with anti-P/Q type voltage-gated calcium channel antibodies. This review reports that therapies affecting the BBB that are currently available for disease-modifying therapies for neuroimmunological diseases have the potential to prevent BBB damage.


Asunto(s)
Autoanticuerpos , Barrera Hematoencefálica , Chaperón BiP del Retículo Endoplásmico , Esclerosis Múltiple , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Humanos , Autoanticuerpos/inmunología , Animales , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Neuromielitis Óptica/inmunología , Neuromielitis Óptica/metabolismo , Neuromielitis Óptica/patología , Encefalitis/inmunología , Encefalitis/metabolismo , Encefalitis/patología
4.
Sci Signal ; 17(856): eadt4125, 2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-39353039

RESUMEN

Obesity exacerbates inflammation to a greater extent in female patients and mice with multiple sclerosis.


Asunto(s)
Autoinmunidad , Esclerosis Múltiple , Obesidad , Animales , Femenino , Humanos , Obesidad/inmunología , Obesidad/metabolismo , Ratones , Esclerosis Múltiple/inmunología , Masculino , Inflamación/inmunología , Inflamación/metabolismo , Factores Sexuales , Caracteres Sexuales
5.
Brain Nerve ; 76(10): 1101-1108, 2024 Oct.
Artículo en Japonés | MEDLINE | ID: mdl-39370834

RESUMEN

B-cell therapy using anti-CD20 antibodies significantly suppresses relapse and is therefore an important treatment option for multiple sclerosis (MS). Based on the production of inflammatory cytokines and enhanced antigen-presenting capacity, B cells trigger MS relapses via activation of pathogenic T cells. Suppression of these abnormal actions of B cells is the primary mechanism underlying relapse prevention using B-cell therapies. Treatments that target B cells are also expected to suppress chronic progression of MS through modulation of B-cell activity within the central nervous system. B-cell therapies based on novel approaches are expected to improve the regulation of acute and chronic MS pathology.


Asunto(s)
Linfocitos B , Esclerosis Múltiple , Humanos , Esclerosis Múltiple/terapia , Esclerosis Múltiple/inmunología , Linfocitos B/inmunología , Animales , Antígenos CD20/inmunología
6.
Sci Rep ; 14(1): 25540, 2024 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-39462090

RESUMEN

Intrathecal synthesis of immunoglobulins (Igs) is a key hallmark of multiple sclerosis (MS). B cells are known to accumulate in the leptomeninges of MS patients and associate with pathology in the underlying cortex and a more severe disease course. However, the role of locally produced antibodies in MS brain pathology is poorly understood. Here, we quantified the protein levels of IgA, IgM, IgG and albumin in serum and cerebrospinal fluid (CSF) samples of 80 MS patients and 28 neurological controls to calculate Ig indices. In addition, we quantified presence of meningeal IgA+, IgM+ and IgG+ B cells in post-mortem brain tissue of 20 MS patients and 6 controls using immunostainings. IgM and IgG, but not IgA, indices were increased in CSF of MS patients compared to controls, with no observed differences between MS disease types. Both IgM and IgG indices correlated significantly with neurofilament light (NfL) levels in CSF, but not with clinical or radiological parameters of disease. Similarly, IgG+ and IgM+ B cells were increased in MS meninges compared to controls, whereas IgA+ B cells were not. Neuronal loss did not differ between sections with low or high IgA+, IgM+ and IgG+ B cells, but was increased in sections with high numbers of all CD19+ meningeal B cells. Similarly, high presence of CD19+ meningeal B cells and IgG+ meningeal B cells associated with increased microglial density in the underlying cortex. Taken together, intrathecal synthesis of IgG and IgM is elevated in MS, which corresponds to an increased number of IgG+ and IgM+ B cells in MS meninges. The significant correlation between intrathecal IgG and IgM production and NfL levels, and increased microglial activation in cortical areas adjacent to meningeal infiltrates with high levels of IgG+ B cells indicate a role for intrathecal IgM- and IgG-producing B cells in neuroinflammatory and degenerative processes in MS.


Asunto(s)
Linfocitos B , Biomarcadores , Inmunoglobulina G , Inmunoglobulina M , Meninges , Esclerosis Múltiple , Humanos , Esclerosis Múltiple/líquido cefalorraquídeo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Inmunoglobulina M/líquido cefalorraquídeo , Inmunoglobulina G/líquido cefalorraquídeo , Inmunoglobulina G/sangre , Masculino , Persona de Mediana Edad , Femenino , Meninges/inmunología , Meninges/patología , Meninges/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Adulto , Biomarcadores/líquido cefalorraquídeo , Anciano , Proteínas de Neurofilamentos/líquido cefalorraquídeo
7.
Nat Rev Neurol ; 20(11): 685-690, 2024 11.
Artículo en Inglés | MEDLINE | ID: mdl-39379493

RESUMEN

The genetic architecture of multiple sclerosis (MS) is similar to that of coeliac disease, with human leukocyte antigen (HLA) being the greatest genetic determinant in both diseases. Furthermore, similar to the involvement of gluten in coeliac disease, Epstein-Barr virus (EBV) infection is now widely considered to be an important environmental factor in MS. The molecular basis for the HLA association in coeliac disease is well defined, and B cells have a clear role in antigen presentation to gluten-specific CD4+ T cells. By contrast, the mechanisms underlying the HLA association of MS are unknown but accumulating evidence indicates a similar role of B cells acting as antigen-presenting cells. The growing parallels suggest that much could be learned about the mechanisms of MS by using coeliac disease as a model. In this Perspective article, we discuss the insights that could be gained from these parallels and consider the possibility of antiviral treatment against EBV as a therapy for MS that is analogous to the gluten-free diet in coeliac disease.


Asunto(s)
Enfermedad Celíaca , Esclerosis Múltiple , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/genética , Humanos , Esclerosis Múltiple/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/complicaciones , Antígenos HLA/genética , Antígenos HLA/inmunología , Herpesvirus Humano 4/inmunología , Linfocitos B/inmunología
8.
Proc Natl Acad Sci U S A ; 121(44): e2416097121, 2024 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-39432795

RESUMEN

Both genetic and environmental factors contribute to multiple sclerosis (MS) risk. Infection with the Epstein-Barr virus (EBV) is the strongest environmental risk factor, and HLA-DR15 is the strongest genetic risk factor for MS. We employed computational methods and in vitro assays for CD4 T cell activation to investigate the DR15-restricted response to EBV. Using a machine learning-based HLA ligand predictor, the EBV glycoprotein B (gB) was predicted to be enriched in epitopes restricted to presentation by DRB1*15:01. In DR15-positive individuals, two epitopes comprised the major CD4 T cell response to gB. Surprisingly, the expression of recombinant gB in a DR15-homozygous B cell line or primary autologous B cells elicited a CD4 T cell response, indicating that intracellular gB was loaded onto HLA class II molecules. By deleting the signal sequence of gB, we determined that this pathway for direct activation of CD4 T cells was dependent on trafficking to the endoplasmic reticulum (ER) within the B cell. We screened seven recombinant EBV antigens from the ER compartment for immune responses in DR15-negative vs. DR15-homozygous individuals. In addition to gB, gH was a key CD4 T cell target in individuals homozygous for DR15. Compared to non-DR15 controls, DR15-homozygotes had significantly higher T cell responses to both gB and gH but not to EBV latent or lytic antigens overall. Responses to gB and gH were slightly elevated in DR15 homozygotes with MS. Our results link MS environmental and genetic risk factors by demonstrating that HLA-DR15 dictates CD4 T cell immunity to EBV antigens.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD4-Positivos , Cadenas HLA-DRB1 , Herpesvirus Humano 4 , Humanos , Linfocitos T CD4-Positivos/inmunología , Herpesvirus Humano 4/inmunología , Cadenas HLA-DRB1/genética , Cadenas HLA-DRB1/inmunología , Presentación de Antígeno/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Linfocitos B/inmunología , Epítopos de Linfocito T/inmunología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/virología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/inmunología , Glicoproteínas/inmunología , Glicoproteínas/genética , Activación de Linfocitos/inmunología
9.
Neurol Neuroimmunol Neuroinflamm ; 11(6): e200312, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39467238

RESUMEN

BACKGROUND AND OBJECTIVES: In multiple sclerosis (MS), immune cells invade the CNS and destroy myelin. Macrophages contribute to demyelination and myelin repair, and their role in each process depends on their ability to acquire specific phenotypes in response to external signals. In this article, we assess whether defects in MS patient macrophage responses may lead to increased inflammation or lack of neuroregenerative effects. METHODS: CD14+CD16- monocytes from patients with MS and healthy controls (HCs) were activated in vitro to obtain homeostatic-like, proinflammatory, and proregenerative macrophages. Macrophage activation profiles were assessed through RNA sequencing and metabolomics. Surface molecule expression of CD14, CD16, and HLA-DR and myelin phagocytic capacity were evaluated with flow cytometry. Macrophage supernatant capacity to influence oligodendrocyte precursor cell differentiation toward an astrocytic or oligodendroglia fate was also tested. RESULTS: We observed that MS patient monocytes ex vivo recapitulate their preferential activation toward the CD16+ phenotype, a subset of proinflammatory cells overrepresented in MS lesions. Functionally, MS patient macrophages display a decreased capacity to phagocytose human myelin and a deficit of processing myelin after ingestion. In addition, MS patient macrophage supernatant favors astrocytes over oligodendrocyte differentiation when compared with HC macrophage supernatant. Furthermore, even when exposed to homeostatic or proregenerative stimuli, MS patient macrophages uphold a proinflammatory transcriptomic profile with higher levels of cytokine/chemokine. Of interest, MS patient macrophages exhibit a distinct metabolic signature with a mitochondrial energy metabolism blockage. Transcriptomic data are further substantiated by metabolomics studies that reveal perturbations in the corresponding metabolic pathways. DISCUSSION: Our results show an intrinsic defect of MS patient macrophages, reminiscent of innate immune cell memory in MS, lifting macrophage importance in the disease and as potential therapeutic targets.


Asunto(s)
Macrófagos , Esclerosis Múltiple , Humanos , Macrófagos/metabolismo , Macrófagos/inmunología , Adulto , Masculino , Femenino , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/inmunología , Persona de Mediana Edad , Activación de Macrófagos/fisiología , Fagocitosis , Oligodendroglía/metabolismo , Vaina de Mielina/metabolismo , Monocitos/metabolismo , Monocitos/inmunología
10.
Nat Commun ; 15(1): 9287, 2024 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-39468055

RESUMEN

Mucosal-associated invariant T (MAIT) cells express semi-invariant T cell receptors (TCR) for recognizing bacterial and yeast antigens derived from riboflavin metabolites presented on the non-polymorphic MHC class I-related protein 1 (MR1). Neuroinflammation in multiple sclerosis (MS) is likely initiated by autoreactive T cells and perpetuated by infiltration of additional immune cells, but the precise role of MAIT cells in MS pathogenesis remains unknown. Here, we use experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, and find an accumulation of MAIT cells in the inflamed central nervous system (CNS) enriched for MAIT17 (RORγt+) and MAIT1/17 (T-bet+RORγt+) subsets with inflammatory and protective features. Results from transcriptome profiling and Nur77GFP reporter mice show that these CNS MAIT cells are activated via cytokines and TCR. Blocking TCR activation with an anti-MR1 antibody exacerbates EAE, whereas enhancing TCR activation with the cognate antigen, 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil, ameliorates EAE severity, potentially via the induction of amphiregulin (AREG). In summary, our findings suggest that TCR-mediated MAIT cell activation is protective in CNS inflammation, likely involving an induction of AREG.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Activación de Linfocitos , Ratones Endogámicos C57BL , Células T Invariantes Asociadas a Mucosa , Receptores de Antígenos de Linfocitos T , Encefalomielitis Autoinmune Experimental/inmunología , Animales , Células T Invariantes Asociadas a Mucosa/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Ratones , Activación de Linfocitos/inmunología , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/inmunología , Esclerosis Múltiple/inmunología , Sistema Nervioso Central/inmunología , Citocinas/metabolismo , Citocinas/inmunología , Ribitol/análogos & derivados , Ribitol/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Uracilo/análogos & derivados , Uracilo/farmacología
11.
Nat Commun ; 15(1): 9403, 2024 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-39477943

RESUMEN

Mimicry of host protein structures, or 'molecular mimicry', is a common mechanism employed by viruses to evade the host's immune system. Short linear amino acid (AA) molecular mimics can elicit cross-reactive antibodies and T cells from the host, but the prevalence of such mimics throughout the human virome has not been fully explored. Here we evaluate 134 human-infecting viruses and find significant usage of linear mimicry across the virome, particularly those in the Herpesviridae and Poxviridae families. Furthermore, host proteins related to cellular replication and inflammation, autosomes, the X chromosome, and thymic cells are enriched as viral mimicry targets. Finally, we find that short linear mimicry from Epstein-Barr virus (EBV) is higher in auto-antibodies found in patients with multiple sclerosis than previously appreciated. Our results thus hint that human-infecting viruses leverage mimicry in the course of their infection, and that such mimicry may contribute to autoimmunity, thereby prompting potential targets for therapies.


Asunto(s)
Autoinmunidad , Evasión Inmune , Imitación Molecular , Imitación Molecular/inmunología , Humanos , Autoinmunidad/inmunología , Evasión Inmune/inmunología , Herpesvirus Humano 4/inmunología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/virología , Virosis/inmunología , Virosis/virología , Autoanticuerpos/inmunología , Herpesviridae/inmunología , Reacciones Cruzadas/inmunología , Poxviridae/inmunología , Poxviridae/genética , Virus/inmunología , Virus/genética
12.
Gut Microbes ; 16(1): 2418415, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39462277

RESUMEN

BACKGROUND: Short-chain fatty acids (SCFAs), produced by the gut microbiota, are thought to exert an anti-inflammatory effect on the host immune system. The levels of SCFAs and abundance of the microbiota that produce them are depleted in multiple sclerosis (MS), an autoimmune disease of the central nervous system (CNS). The mechanisms leading to this depletion are unknown. Using experimental autoimmune encephalomyelitis (EAE) as a model for MS, we have previously shown that gut microbiomes divergent in their abundance of specific commensal Lactobacillaceae, Limosilactobacillus reuteri (L. reuteri) and Ligilactobacillus murinus (L. murinus), differentially impact CNS autoimmunity. To determine the underlying mechanisms, we employed colonization by L. reuteri and L. murinus in disparate gut microbiome configurations in vivo and in vitro, profiling their impact on gut microbiome composition and metabolism, coupled with modulation of dietary fiber in the EAE model. RESULTS: We show that stable colonization by L. reuteri, but not L. murinus, exacerbates EAE, in conjunction with a significant remodeling of gut microbiome composition, depleting SCFA-producing microbiota, including Lachnospiraceae, Prevotellaceae, and Bifidobacterium, with a net decrease in bacterial metabolic pathways involved in butyrate production. In a minimal microbiome culture model in vitro, L. reuteri directly inhibited SCFA-producer growth and depleted butyrate. Genomic analysis of L. reuteri isolates revealed an enrichment in bacteriocins with known antimicrobial activity against SCFA-producing microbiota. Functionally, provision of excess dietary fiber, as the prebiotic substrate for SCFA production, elevated SCFA levels and abrogated the ability of L. reuteri to exacerbate EAE. CONCLUSTIONS: Our data highlight a potential mechanism for reduced SCFAs and their producers in MS through depletion by other members of the gut microbiome, demonstrating that interactions between microbiota can impact CNS autoimmunity in a diet-dependent manner. These data suggest that therapeutic restoration of SCFA levels in MS may require not only dietary intervention, but also modulation of the gut microbiome.


Asunto(s)
Butiratos , Encefalomielitis Autoinmune Experimental , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Lactobacillaceae , Animales , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Butiratos/metabolismo , Ratones , Ácidos Grasos Volátiles/metabolismo , Lactobacillaceae/metabolismo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/microbiología , Ratones Endogámicos C57BL , Autoinmunidad , Femenino , Esclerosis Múltiple/microbiología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/inmunología , Fibras de la Dieta/metabolismo , Limosilactobacillus reuteri/metabolismo , Bacterias/clasificación , Bacterias/metabolismo , Bacterias/aislamiento & purificación , Bacterias/genética
13.
Biomolecules ; 14(10)2024 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-39456143

RESUMEN

Multiple Sclerosis (MS) is an inflammatory, demyelinating, and neurodegenerative disease of the central nervous system (CNS) and is termed as one of the most common causes of neurological disability in young adults. Axonal loss and neuronal cell damage are the primary causes of disease progression and disability. Yet, little is known about the mechanism of neurodegeneration in the disease, a limitation that impairs the development of more effective treatments for progressive MS. MS is characterized by the presence of oligoclonal bands and raised levels of immunoglobulins in the CNS. The role of complement in the demyelinating process has been detected in both experimental animal models of MS and within the CNS of affected MS patients. Furthermore, both IgG antibodies and complement activation can be detected in the demyelinating plaques and cortical gray matter lesions. We propose here that both immunoglobulins and complement play an active role in the neurodegenerative process of MS. We hypothesize that the increased CNS IgG antibodies form IgG aggregates and bind complement C1q with high affinity, activating the classical complement pathway. This results in neuronal cell damage, which leads to neurodegeneration and demyelination in MS.


Asunto(s)
Inmunoglobulina G , Esclerosis Múltiple , Humanos , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Inmunoglobulina G/inmunología , Animales , Proteínas del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/inmunología , Activación de Complemento/inmunología , Complemento C1q/metabolismo , Complemento C1q/inmunología , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología
14.
Int J Mol Sci ; 25(20)2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39456713

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) that progresses with demyelination and neurodegeneration. To date, many studies have revealed the key role of interleukins in the pathogenesis of MS, but their impact has not been fully explained. The aim of the present study was to collect and review the results obtained so far regarding the influence of interleukins on the development and course of MS and to assess the potential for their further use. Through the platform "PubMed", terms related to interleukins and MS were searched. The following interval was set as the time criterion: 2014-2024. A total of 12,731 articles were found, and 100 papers were subsequently used. Cells that produce IL-10 have a neuroprotective effect, whereas those that synthesize IL-6 most likely exacerbate neuroinflammation. IL-12, IL-23 and IL-18 represent pro-inflammatory cytokines. It was found that treatment with an anti-IL-12p40 monoclonal antibody in a study group of MS patients showed a beneficial effect. IL-4 is a pleiotropic cytokine that plays a significant role in type 2 immune responses and inhibits MS progression. IL-13 is an anti-inflammatory cytokine through which the processes of oligodendrogenesis and remyelination occur more efficiently. The group of interleukins discussed in our paper may represent a promising starting point for further research aimed at finding new therapies and prognostic markers for MS.


Asunto(s)
Interleucinas , Esclerosis Múltiple , Humanos , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Interleucinas/metabolismo , Interleucinas/inmunología , Animales
15.
Sci Immunol ; 9(99): eadj8094, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39331727

RESUMEN

Multiple sclerosis (MS) is an inflammatory neurological disease of the central nervous system with a subclinical phase preceding frank neuroinflammation. CD8+ T cells are abundant within MS lesions, but their potential role in disease pathology remains unclear. Using high-throughput single-cell RNA sequencing and single-cell T cell receptor analysis, we compared CD8+ T cell clones from the blood and cerebrospinal fluid (CSF) of monozygotic twin pairs in which the cotwin had either no or subclinical neuroinflammation (SCNI). We identified peripheral MS-associated immunological and metabolic alterations indicative of an enhanced migratory, proinflammatory, and activated CD8+ T cell phenotype, which was also evident in cotwins with SCNI and in an independent validation cohort of people with MS. Together, our in-depth single-cell analysis indicates a disease-driving proinflammatory role of infiltrating CD8+ T cells and identifies potential immunological and metabolic therapeutic targets in both prodromal and definitive stages of the disease.


Asunto(s)
Linfocitos T CD8-positivos , Esclerosis Múltiple , Linfocitos T CD8-positivos/inmunología , Humanos , Esclerosis Múltiple/inmunología , Femenino , Masculino , Adulto , Gemelos Monocigóticos , Persona de Mediana Edad , Análisis de la Célula Individual
16.
Cells ; 13(18)2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39329740

RESUMEN

Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system, leading to significant disability through neurodegeneration. Despite advances in the understanding of MS pathophysiology, effective treatments remain limited. Mesenchymal stem cells (MSCs) have gained attention as a potential therapeutic option due to their immunomodulatory and regenerative properties. This review examines MS pathogenesis, emphasizing the role of immune cells, particularly T cells, in disease progression, and explores MSCs' therapeutic potential. Although preclinical studies in animal models show MSC efficacy, challenges such as donor variability, culture conditions, migratory capacity, and immunological compatibility hinder widespread clinical adoption. Strategies like genetic modification, optimized delivery methods, and advanced manufacturing are critical to overcoming these obstacles. Further research is needed to validate MSCs' clinical application in MS therapy.


Asunto(s)
Inmunidad Adaptativa , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Esclerosis Múltiple , Humanos , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Esclerosis Múltiple/patología , Células Madre Mesenquimatosas/inmunología , Animales
17.
Trends Immunol ; 45(10): 750-759, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39322475

RESUMEN

Neural cells in our central nervous system (CNS) have long been thought to be mere targets of neuroinflammatory events in neurodegenerative diseases such as multiple sclerosis (MS) or Alzheimer's disease. While glial populations such as microglia and astrocytes emerged as active responders and modifiers of pathological environments, oligodendroglia and neurons have been associated with altered homeostasis and eventual cell death. The advent of single-cell and spatial omics technologies has demonstrated transitions of CNS-resident glia, including oligodendroglia, into disease-associated (DA) states. Anchored in recent findings of their roles in MS, we propose that DA glia constitute key nexus of disease progression, with DA oligodendroglia contributing to the modulation of neuroinflammation in certain neurodegenerative diseases, constituting novel putative pharmacological targets for such pathologies.


Asunto(s)
Enfermedades Neurodegenerativas , Oligodendroglía , Humanos , Oligodendroglía/patología , Oligodendroglía/metabolismo , Animales , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/patología , Sistema Nervioso Central/inmunología , Neuronas/metabolismo , Neuronas/patología , Neuronas/inmunología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología
18.
Gut Microbes ; 16(1): 2402547, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39287045

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory demyelination occurring in the central nervous system (CNS). Inulin is a common prebiotic that can improve metabolic disorders by modulating the gut microbiota. However, its capacity to affect CNS autoimmunity is poorly recognized. Experimental autoimmune encephalomyelitis (EAE) is a classical mouse model of MS. Herein, we found that oral administration of inulin ameliorated the severity EAE in mice, accompanied by reductions in inflammatory cell infiltration and demyelination in the CNS. These reductions were associated with decreased proportion and numbers of Th17 cells in brain and spleen. Consistent with the findings, the serum concentrations of IL-17, IL-6, and TNF-α were reduced in inulin treated EAE mice. Moreover, the proliferation of auto-reactive lymphocytes, against MOG35-55 antigen, was attenuated ex vivo. Mechanistically, inulin treatment altered the composition of gut microbiota. It increased Lactobacillus and Dubosiella whereas decreased g_Prevotellaceae_NK3B31_group at the genus level, alongside with elevated concentration of butyric acid in fecal content and serum. In vitro, butyrate, but not inulin, could inhibit the activation of MOG35-55 stimulated lymphocytes. Furthermore, fecal microbiota transplantation assay confirmed that fecal contents of inulin-treated normal mice had an ameliorative effect on EAE mice. In contrast, antibiotic cocktail (ABX) treatment diminished the therapeutic effect of inulin in EAE mice as well as the reduction of Th17 cells, while supplementation with Lactobacillus reuteri restored the amelioration effect. These results confirmed that the attenuation of inulin on Th17 cells and inflammatory demyelination in EAE mice was dependent on its modulation on gut microbiota and metabolites. Our findings provide a potential therapeutic regimen for prebiotic inulin supplementation in patients with multiple sclerosis.


Asunto(s)
Autoinmunidad , Encefalomielitis Autoinmune Experimental , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Inulina , Ratones Endogámicos C57BL , Esclerosis Múltiple , Prebióticos , Células Th17 , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Inulina/administración & dosificación , Inulina/farmacología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/microbiología , Células Th17/inmunología , Ratones , Prebióticos/administración & dosificación , Femenino , Ácidos Grasos Volátiles/metabolismo , Autoinmunidad/efectos de los fármacos , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/microbiología , Sistema Nervioso Central/inmunología , Bacterias/clasificación , Bacterias/aislamiento & purificación
19.
Front Immunol ; 15: 1446748, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39224590

RESUMEN

Multiple sclerosis (MS) is a devastating immune-mediated disorder of the central nervous system resulting in progressive disability accumulation. As there is no cure available yet for MS, the primary therapeutic objective is to reduce relapses and to slow down disability progression as early as possible during the disease to maintain and/or improve health-related quality of life. However, optimizing treatment for people with MS (pwMS) is complex and challenging due to the many factors involved and in particular, the high degree of clinical and sub-clinical heterogeneity in disease progression among pwMS. In this paper, we discuss these many different challenges complicating treatment optimization for pwMS as well as how a shift towards a more pro-active, data-driven and personalized medicine approach could potentially improve patient outcomes for pwMS. We describe how the 'Clinical Impact through AI-assisted MS Care' (CLAIMS) project serves as a recent example of how to realize such a shift towards personalized treatment optimization for pwMS through the development of a platform that offers a holistic view of all relevant patient data and biomarkers, and then using this data to enable AI-supported prognostic modelling.


Asunto(s)
Inteligencia Artificial , Esclerosis Múltiple , Medicina de Precisión , Humanos , Inteligencia Artificial/tendencias , Biomarcadores , Progresión de la Enfermedad , Esclerosis Múltiple/terapia , Esclerosis Múltiple/inmunología , Medicina de Precisión/métodos , Medicina de Precisión/tendencias , Pronóstico , Calidad de Vida
20.
Front Immunol ; 15: 1432348, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39301017

RESUMEN

Background: This study examines the humoral and cellular response in multiple sclerosis (MS) patients on anti-CD20 therapy before and after the 1st to 4th BNT162b2 mRNA SARS-CoV-2 vaccination and the relationship with breakthrough infection. Methods: Participants with McDonald 2017 MS that were treated with ocrelizumab were included. The study duration was throughout the COVID-19 pandemic until four months after fourth mRNA SARS-CoV-2 vaccination (BNT162b2). Longitudinal blood samples were analysed for: IgG antibodies of SARS-CoV-2 spike anti-receptor binding domain (anti-RBD), nucleocapsid IgG antibodies (anti-N) and activation induced marker expressing CD4+, CD8+ T-cells and concentration of ocrelizumab and anti-drug antibodies. Incidences of breakthrough infection were confirmed with SARS-CoV-2 PCR tests. Results: The rate of anti-RBD positive participants increased substantially between the third and fourth vaccination from 22.2% to 55.9% (median 54.7 BAU/mL; IQR: 14.5 - 221.2 BAU/mL and 607.7 BAU/mL; IQR: 29.4 - 784.6 BAU/mL, respectively). Within the same period 75% of participants experienced breakthrough infection. The fourth vaccination resulted in an additional increase in seropositive individuals (64.3%) (median 541.8 BAU/mL (IQR: 19.1-1007 BAU/mL). Breakthrough infection did not influence the cellular response without a significant change after the fourth vaccination. During the study period two participants had detectable anti-N, both after the fourth vaccination. No correlation was found between serum concentration of ocrelizumab and the humoral and cellular response. Discussion: Low levels or absence of specific anti-RBD following vaccination, with a significant increase after breakthrough infections and boosted by the fourth vaccination. T-cell reactivity remained sustained and unaffected by breakthrough infections.


Asunto(s)
Anticuerpos Antivirales , Vacuna BNT162 , COVID-19 , Inmunidad Celular , Inmunidad Humoral , Esclerosis Múltiple , SARS-CoV-2 , Humanos , Masculino , COVID-19/inmunología , COVID-19/prevención & control , Femenino , SARS-CoV-2/inmunología , Vacuna BNT162/inmunología , Adulto , Persona de Mediana Edad , Estudios Longitudinales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/tratamiento farmacológico , Vacunas contra la COVID-19/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Glicoproteína de la Espiga del Coronavirus/inmunología , Antígenos CD20/inmunología , Vacunación , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Infección Irruptiva
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...