Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
Invest Ophthalmol Vis Sci ; 65(8): 51, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-39083309

RESUMEN

Purpose: To investigate the effects of anterior chamber pigment dispersion on ocular immune privilege and the possible mechanisms involved in a DBA/2J mouse model of pigmentary glaucoma. Methods: DBA/2J mice were utilized as a pigment dispersion model, and age-matched C57BL/6J mice were used as the control group in this study. Proteins in the aqueous humor (AH) and serum were quantified using the bicinchoninic acid assay. Immune cells in the AH were detected using hematoxylin and eosin staining and immunocytochemistry. The expression of TGF-ß2 in the AH and cytokine levels (IL-10, IFN-γ) in serum were measured using ELISA. Anterior chamber-associated immune deviation (ACAID) was induced in DBA/2J mice by injecting antigens into the anterior chamber. Delayed-type hypersensitivity (DTH) assays were used to assess the induction of ACAID. In DBA/2J mice, before and after pigment dispersion, following anterior chamber injection of pigment particles, and after ACAID modeling, the expression of regulatory T cells (Tregs) was detected using flow cytometry. Results: Compared to C57BL/6J mice, the protein concentration, immune cell count, and TGF-ß2 levels in the AH were elevated in DBA/2J mice. Protein concentration and IL-10 levels in serum were increased, while IFN-γ levels were decreased in DBA/2J. Additionally, the expression of Treg cells in the spleen of DBA/2J mice was significantly increased after pigment dispersion and anterior chamber injection of pigment particles. At 3 and 6 months, DTH responses in DBA/2J mice were not inhibited, thus preventing ACAID induction. However, the opposite was observed at 9 months in DBA/2J mice. Furthermore, the ACAID group exhibited an augmented expression of Treg cells. Conclusions: Dispersion of pigment particles in the anterior chamber of the eye enhances the state of ocular immune privilege by influencing the immunosuppressive microenvironment and inducing more Treg cells to reestablish ACAID.


Asunto(s)
Humor Acuoso , Modelos Animales de Enfermedad , Glaucoma de Ángulo Abierto , Privilegio Inmunológico , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Linfocitos T Reguladores , Animales , Humor Acuoso/metabolismo , Humor Acuoso/inmunología , Ratones , Linfocitos T Reguladores/inmunología , Glaucoma de Ángulo Abierto/inmunología , Cámara Anterior/inmunología , Factor de Crecimiento Transformador beta2 , Presión Intraocular/fisiología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Interleucina-10 , Hipersensibilidad Tardía/inmunología , Interferón gamma/metabolismo , Inmunohistoquímica , Femenino
2.
Front Immunol ; 15: 1380063, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38863704

RESUMEN

Historically, the central nervous system (CNS) was regarded as 'immune-privileged', possessing its own distinct immune cell population. This immune privilege was thought to be established by a tight blood-brain barrier (BBB) and blood-cerebrospinal-fluid barrier (BCSFB), which prevented the crossing of peripheral immune cells and their secreted factors into the CNS parenchyma. However, recent studies have revealed the presence of peripheral immune cells in proximity to various brain-border niches such as the choroid plexus, cranial bone marrow (CBM), meninges, and perivascular spaces. Furthermore, emerging evidence suggests that peripheral immune cells may be able to infiltrate the brain through these sites and play significant roles in driving neuronal cell death and pathology progression in neurodegenerative disease. Thus, in this review, we explore how the brain-border immune niches may contribute to the pathogenesis of neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). We then discuss several emerging options for harnessing the neuroimmune potential of these niches to improve the prognosis and treatment of these debilitative disorders using novel insights from recent studies.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Enfermedades Neurodegenerativas , Humanos , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/patología , Animales , Barrera Hematoencefálica/inmunología , Encéfalo/inmunología , Encéfalo/patología , Privilegio Inmunológico
3.
J Autoimmun ; 145: 103217, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38581915

RESUMEN

The autoimmunity-promoting cytokine, Interleukin-15 (IL-15), is often claimed to be a key pathogenic cytokine in alopecia areata (AA). Yet, rhIL-15 promotes human hair follicle (HF) growth ex vivo. We have asked whether the expression of IL-15 and its receptor (IL-15R) isoforms is altered in human AA and how IL-15 impacts on human HF immune privilege (HF-IP) in the presence/absence of interferon-γ (IFNγ), the well-documented key AA-pathogenic cytokine, as well as on hair regrowth after experimental AA induction in vivo. Quantitative immunohistomorphometry showed the number of perifollicular IL-15+ T cells in AA skin biopsies to be significantly increased compared to healthy control skin, while IL-15, IL-15Rα, and IL-15Rγ protein expression within the hair bulb were significantly down-regulated in AA HFs. In organ-cultured human scalp HFs, rhIL-15 significantly reduced hair bulb expression of MICA, the key "danger" signal in AA pathogenesis, and increased production of the HF-IP guardian, α-MSH. Crucially, ex vivo, rhIL-15 prevented IFNγ-induced HF-IP collapse, restored a collapsed HF-IP by IL-15Rα-dependent signaling (as documented by IL-15Rα-silencing), and protected AA-preventive immunoinhibitory iNKT10 cells from IFNγ-induced apoptosis. rhIL-15 even promoted hair regrowth after experimental AA induction in human scalp skin xenotransplants on SCID/beige mice in vivo. Our data introduce IL-15 as a novel, functionally important HF-IP guardian whose signaling is constitutively defective in scalp HFs of AA patients. Our data suggest that selective stimulation of intrafollicular IL-15Rα signaling could become a novel therapeutic approach in AA management, while blocking it pharmacologically may hinder both HF-IP restoration and hair re-growth and may thus make HFs more vulnerable to AA relapse.


Asunto(s)
Alopecia Areata , Folículo Piloso , Privilegio Inmunológico , Interferón gamma , Interleucina-15 , Interleucina-15/metabolismo , Interleucina-15/inmunología , Folículo Piloso/inmunología , Folículo Piloso/metabolismo , Humanos , Animales , Alopecia Areata/inmunología , Alopecia Areata/metabolismo , Ratones , Interferón gamma/metabolismo , Femenino , Receptores de Interleucina-15/metabolismo , Receptores de Interleucina-15/inmunología , Masculino , Adulto , Persona de Mediana Edad , Subunidad alfa del Receptor de Interleucina-15/metabolismo , Subunidad alfa del Receptor de Interleucina-15/inmunología , Piel/inmunología , Piel/metabolismo , Piel/patología , Modelos Animales de Enfermedad
4.
Adv Mater ; 36(31): e2312088, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38638030

RESUMEN

Disorders in the regulatory arm of the adaptive immune system result in autoimmune-mediated diseases. While systemic immunosuppression is the prevailing approach to manage them, it fails to achieve long-lasting remission due to concomitant suppression of the regulatory arm and carries the risk of heightened susceptibility to infections and malignancies. Alopecia areata is a condition characterized by localized hair loss due to autoimmunity. The accessibility of the skin allows local rather than systemic intervention to avoid broad immunosuppression. It is hypothesized that the expansion of endogenous regulatory T cells (Tregs) at the site of antigen encounter can restore the immune balance and generate a long-lasting tolerogenic response. A hydrogel microneedle (MN) patch is therefore utilized for delivery of CCL22, a Treg-chemoattractant, and IL-2, a Treg survival factor to amplify them. In an immune-mediated murine model of alopecia, local bolstering of Treg numbers is shown, leading to sustained hair regrowth and attenuation of inflammatory pathways. In a humanized skin transplant mouse model, expansion of Tregs within human skin is confirmed without engendering peripheral immunosuppression. The patch offers high-loading capacity and shelf-life stability for prospective clinical translation. By harmonizing immune responses locally, the aim is to reshape the landscape of autoimmune skin disease management.


Asunto(s)
Alopecia , Folículo Piloso , Linfocitos T Reguladores , Animales , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Ratones , Humanos , Agujas , Privilegio Inmunológico , Hidrogeles/química , Factores Inmunológicos/química , Factores Inmunológicos/farmacología , Interleucina-2/metabolismo , Agentes Inmunomoduladores/química , Agentes Inmunomoduladores/farmacología
5.
Int J Mol Sci ; 25(8)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38673994

RESUMEN

Both alopecia areata (AA) and vitiligo are distinct, heterogenous, and complex disease entities, characterized by nonscarring scalp terminal hair loss and skin pigment loss, respectively. In AA, inflammatory cell infiltrates are in the deep reticular dermis close to the hair bulb (swarm of bees), whereas in vitiligo the inflammatory infiltrates are in the epidermis and papillary dermis. Immune privilege collapse has been extensively investigated in AA pathogenesis, including the suppression of immunomodulatory factors (e.g., transforming growth factor-ß (TGF-ß), programmed death-ligand 1 (PDL1), interleukin-10 (IL-10), α-melanocyte-stimulating hormone (α-MSH), and macrophage migration inhibitory factor (MIF)) and enhanced expression of the major histocompatibility complex (MHC) throughout hair follicles. However, immune privilege collapse in vitiligo remains less explored. Both AA and vitiligo are autoimmune diseases that share commonalities in pathogenesis, including the involvement of plasmacytoid dendritic cells (and interferon-α (IFN- α) signaling pathways) and cytotoxic CD8+ T lymphocytes (and activated IFN-γ signaling pathways). Blood chemokine C-X-C motif ligand 9 (CXCL9) and CXCL10 are elevated in both diseases. Common factors that contribute to AA and vitiligo include oxidative stress, autophagy, type 2 cytokines, and the Wnt/ß-catenin pathway (e.g., dickkopf 1 (DKK1)). Here, we summarize the commonalities and differences between AA and vitiligo, focusing on their pathogenesis.


Asunto(s)
Alopecia Areata , Vitíligo , Alopecia Areata/inmunología , Alopecia Areata/patología , Alopecia Areata/etiología , Alopecia Areata/metabolismo , Humanos , Vitíligo/inmunología , Vitíligo/patología , Vitíligo/metabolismo , Vitíligo/etiología , Animales , Privilegio Inmunológico , Citocinas/metabolismo
6.
Int Immunopharmacol ; 133: 112070, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38640716

RESUMEN

Skin, the largest organ of body, is a highly immunogenic tissue with a diverse collection of immune cells. Highly polymorphic human leukocyte antigen (HLA) molecules have a central role in coordinating immune responses as recognition molecules. Nevertheless, HLA gene expression patterns among diverse cell types within a specific organ, like the skin, have yet to be thoroughly investigated, with stromal cells attracting much less attention than immune cells. To illustrate HLA expression profiles across different cell types in the skin, we performed single-cell RNA sequencing (scRNA-seq) analyses on skin datasets, covering adult and fetal skin, and hair follicles as the skin appendages. We revealed the variation in HLA expression between different skin populations by examining normal adult skin datasets. Moreover, we evaluated the potential immunogenicity of multiple skin populations based on the expression of classical HLA class I genes, which were well represented in all cell types. Furthermore, we generated scRNA-seq data of developing skin from fetuses of 15 post conception weeks (PCW), 17 PCW, and 22 PCW, delineating the dynamic expression of HLA genes with cell type-dependent variation among various cell types during development. Notably, the pseudotime trajectory analysis unraveled the significant variance in HLA genes during the evolution of vascular endothelial cells. Moreover, we uncovered the immune-privileged properties of hair follicles at single-cell resolution. Our study presents a comprehensive single-cell transcriptomic landscape of HLA genes in the skin, which provides new insights into variation in HLA molecules and offers a clue for allogeneic skin transplantation.


Asunto(s)
Perfilación de la Expresión Génica , Antígenos HLA , Análisis de la Célula Individual , Piel , Transcriptoma , Humanos , Piel/inmunología , Piel/metabolismo , Antígenos HLA/genética , Antígenos HLA/inmunología , Folículo Piloso/inmunología , Folículo Piloso/metabolismo , Feto/inmunología , Adulto , Privilegio Inmunológico
7.
Int J Cosmet Sci ; 46(5): 717-733, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38488328

RESUMEN

OBJECTIVE: Dandruff is characterised by the presence of perivascular leukocytes and mild inflammation; however, the immune microenvironment of dandruff-affected scalp skin and the potential changes to the hair follicle's (HF) physiological immune privilege (HF IP) remain unknown. Here, we characterised the HF immune microenvironment and immune privilege status in dandruff-affected scalp skin. METHODS: We assessed relevant key parameters in healthy versus dandruff-affected human scalp biopsies using quantitative immunohistomorphometry, laser capture microdissection, and RNA sequencing. RESULTS: The number of epidermal CD4+ and CD8+ T cells was increased in lesional dandruff scalp skin, while the number of MHC class II+/CD1a+ Langerhans cells was decreased in the infundibulum. The number of intrafollicular and perifollicular CD4+ T cells and CD8+ T cells, perifollicular CD68+ macrophages, and tryptase+ mast cells remained unchanged. Interestingly, MHC class Ia and ß2-microglobulin protein expression were significantly increased specifically in the suprabulbar outer root sheath (ORS) compartment of dandruff-associated HFs. RNAseq analysis of laser capture micro-dissected suprabulbar ORS compartment revealed antigen presentation pathway as the top regulated canonical pathway, along with the upregulation of HF-IP genes such as HLA-C, HLA-DP, and TAP1, which are normally down-regulated in healthy HFs. Intrafollicular protein expression of known HF IP guardians (CD200 and α-MSH) and 'danger signals' (MICA and CXCL10) remained unaltered at the IP sites of dandruff lesional HFs compared to non-lesional and healthy HFs. Instead, the expression of macrophage migration inhibiting factor (MIF), another HF IP guardian, was reduced. CONCLUSION: Together, this work shows that dandruff is associated with epidermal T-cell infiltration and a weakened HF IP in the suprabulbar ORS of HFs in dandruff lesional scalp.


OBJECTIF: Les pellicules sont caractérisées par la présence de leucocytes périvasculaires et une légère inflammation. Cependant, le microenvironnement immunitaire de la peau du cuir chevelu affectée par les pellicules et les modifications potentielles du privilège immunitaire physiologique du follicule pileux (PI FP) restent inconnus. Ici, nous avons caractérisé le microenvironnement immunitaire du follicule pileux (FP) et l'état du privilège immunitaire de la peau du cuir chevelu affectée par les pellicules. MÉTHODES: Nous avons évalué les principaux paramètres pertinents dans des biopsies de cuir chevelu humain sain par rapport à ceux touchés par les pellicules, à l'aide d'une immuno­histomorphométrie quantitative, d'une microdissection au laser et d'un séquençage de l'ARN. RÉSULTATS: Le nombre de lymphocytes T CD4+ et CD8+ épidermiques a augmenté dans la peau du cuir chevelu atteinte de pellicules lésionnelles, tandis que le nombre de cellules de Langerhans du CMH de classe II+/CD1a+ a diminué dans l'infundibulum. Le nombre de lymphocytes T CD4+ et de lymphocytes T CD8+ intrafolliculaires et périfolliculaires, de macrophages CD68+ périfolliculaires et de mastocytes tryptase+ est resté inchangé. Il est intéressant de noter que l'expression des protéines du CMH de classe Ia et de la ß2­microglobuline a augmenté de manière significative dans le compartiment suprabulbaire de la gaine radiculaire externe (GRE) en particulier des FP associés aux pellicules. L'analyse par séquençage ARN du compartiment suprabulbaire de la GRE micro­disséquée au laser a révélé que la voie de présentation de l'antigène était la voie canonique la plus régulée, ainsi que la régulation à la hausse des gènes PI­FP tels que HLA­C, HLA­DP et TAP1, qui sont normalement régulés à la baisse dans les FP sains. L'expression protéique intrafolliculaire des gardiens connus du PI FP (CD200 et α­MSH) et des « signaux de danger ¼ (MICA et CXCL10) est restée inchangée au niveau des sites du PI des FP à pellicules lésionnelles par rapport aux FP sans pellicules lésionnelles et sains. En revanche, l'expression du facteur d'inhibition de la migration des macrophages (MIF), un autre gardien du PI FP, a été réduite. CONCLUSION: L'ensemble de ces travaux montrent que les pellicules sont associées à une infiltration épidermique des lymphocytes T et à un affaiblissement du PI FP dans la GRE suprabulbaire des FP du cuir chevelu atteint de pellicules lésionnelles.


Asunto(s)
Caspa , Folículo Piloso , Privilegio Inmunológico , Cuero Cabelludo , Humanos , Folículo Piloso/inmunología , Caspa/inmunología , Femenino , Persona de Mediana Edad , Linfocitos T/inmunología , Masculino , Adulto , Epidermis/inmunología , Epidermis/metabolismo
8.
Sci Immunol ; 9(93): eadi7038, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38517952

RESUMEN

The persistent murine norovirus strain MNVCR6 is a model for human norovirus and enteric viral persistence. MNVCR6 causes chronic infection by directly infecting intestinal tuft cells, rare chemosensory epithelial cells. Although MNVCR6 induces functional MNV-specific CD8+ T cells, these lymphocytes fail to clear infection. To examine how tuft cells promote immune escape, we interrogated tuft cell interactions with CD8+ T cells by adoptively transferring JEDI (just EGFP death inducing) CD8+ T cells into Gfi1b-GFP tuft cell reporter mice. Unexpectedly, some intestinal tuft cells partially resisted JEDI CD8+ T cell-mediated killing-unlike Lgr5+ intestinal stem cells and extraintestinal tuft cells-despite seemingly normal antigen presentation. When targeting intestinal tuft cells, JEDI CD8+ T cells predominantly adopted a T resident memory phenotype with decreased effector and cytotoxic capacity, enabling tuft cell survival. JEDI CD8+ T cells neither cleared nor prevented MNVCR6 infection in the colon, the site of viral persistence, despite targeting a virus-independent antigen. Ultimately, we show that intestinal tuft cells are relatively resistant to CD8+ T cells independent of norovirus infection, representing an immune-privileged niche that can be leveraged by enteric microbes.


Asunto(s)
Linfocitos T CD8-positivos , Norovirus , Ratones , Humanos , Animales , Células en Penacho , Norovirus/fisiología , Privilegio Inmunológico , Intestinos
9.
J Virol ; 98(1): e0078923, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38168677

RESUMEN

Zika virus (ZIKV) infection caused neurological complications and male infertility, leading to the accumulation of antigen-specific immune cells in immune-privileged organs (IPOs). Thus, it is important to understand the immunological responses to ZIKV in IPOs. We extensively investigated the ZIKV-specific T cell immunity in IPOs in Ifnar1-/- mice, based on an immunodominant epitope E294-302 tetramer. The distinct kinetics and functions of virus-specific CD8+ T cells infiltrated into different IPOs were characterized, with late elevation in the brain and spinal cord. Single epitope E294-302-specific T cells can account for 20-60% of the total CD8+ T cells in the brain, spinal cord, and testicle and persist for at least 90 days in the brain and spinal cord. The E294-302-specific TCRαßs within the IPOs are featured with the majority of clonotypes utilizing TRAV9N-3 paired with diverse TRBV chains, but with distinct αß paired clonotypes in 7 and 30 days post-infection. Specific chemokine receptors, Ccr2 and Ccr5, were selectively expressed in the E294-302-specific CD8+ T cells within the brain and testicle, indicating an IPO-oriented migration of virus-specific CD8+ T cells after infection. Overall, this study adds to the understanding of virus-specific CD8+ T cell responses for controlling and clearing ZIKV infection in IPOs.IMPORTANCEThe immune-privileged organs (IPOs), such as the central nervous system and testicles, presented pathogenicity and inflammation after Zika virus (ZIKV) infection with infiltrated CD8+ T cells. Our data show that CD8+ T cells keep up with virus increases and decreases in immune-privileged organs. Furthermore, our study provides the first ex vivo comparative analyses of the composition and diversity related to TCRα/ß clonotypes across anatomical sites and ZIKV infection phases. We show that the vast majority of TCRα/ß clonotypes in tissues utilize TRAV9N-3 with conservation. Specific chemokine expression, including Ccr2 and Ccr5, was found to be selectively expressed in the E294-302-specific CD8+ T cells within the brain and testicle, indicating an IPO-oriented migration of the virus-specific CD8+ T cells after the infection. Our study adds insights into the anti-viral immunological characterization and chemotaxis mechanism of virus-specific CD8+ T cells after ZIKV infection in different IPOs.


Asunto(s)
Linfocitos T CD8-positivos , Privilegio Inmunológico , Infección por el Virus Zika , Animales , Masculino , Ratones , Encéfalo/inmunología , Encéfalo/virología , Linfocitos T CD8-positivos/inmunología , Receptor de Interferón alfa y beta/genética , Virus Zika , Infección por el Virus Zika/inmunología , Ratones Noqueados , Testículo/inmunología , Testículo/virología
10.
Sci Immunol ; 9(91): eadh0152, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38181095

RESUMEN

Immune tolerance is maintained in lymphoid organs (LOs). Despite the presence of complex immune cell networks in non-LOs, it is unknown whether self-tolerance is maintained in these tissues. We developed a technique to restrict genetic recombination to regulatory T cells (Tregs) only in skin. Selective depletion of skin Tregs resulted in T cell-mediated inflammation of hair follicles (HFs). Suppression did not rely on CTLA-4, but instead on high-affinity interleukin-2 (IL-2) receptor expression by skin Tregs, functioning exclusively in a cell-extrinsic manner. In a novel model of HF stem cell (HFSC)-driven autoimmunity, we reveal that skin Tregs immunologically protect the HFSC niche. Finally, we used spatial transcriptomics to identify aberrant IL-2 signaling at stromal-HF interfaces in a rare form of human alopecia characterized by HFSC destruction and alopecia areata. Collectively, these results reveal the fundamental biology of Tregs in skin uncoupled from the systemic pool and elucidate a mechanism of self-tolerance.


Asunto(s)
Privilegio Inmunológico , Linfocitos T Reguladores , Humanos , Folículo Piloso , Interleucina-2 , Nicho de Células Madre
11.
Viral Immunol ; 36(10): 627-641, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38064537

RESUMEN

Hepatitis E virus (HEV) exhibits tropism toward hepatocytes and thus affects the liver; however, HEV may also affect other tissues, including the heart, kidneys, intestines, testicles, and central nervous system. To date, the pathophysiological links between HEV infection and extrahepatic manifestations have not yet been established. Considering that HEV infects multiple types of cells, the direct effects of virus replication in peripheral tissues represent a plausible explanation for extrahepatic manifestations. In addition, since the immune response is crucial in the development of the disease, the immune characteristics of affected tissues should be revisited to identify commonalities explaining the effects of the virus. This review summarizes the most recent advances in understanding the virus biology and immune-privileged status of specific tissues as major elements for HEV replication in diverse organs. These discoveries may open avenues to explain the multiple extrahepatic manifestations associated with HEV infection and ultimately to design effective strategies for infection control.


Asunto(s)
Virus de la Hepatitis E , Hepatitis E , Humanos , Privilegio Inmunológico , Biología
12.
Biochemistry (Mosc) ; 88(11): 1818-1831, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38105201

RESUMEN

Immune privileges of cancer stem cells is a well-known and widely studied problem, as presence of such cells in tumors is associated with refractoriness, recurrence, and metastasis. Accumulating evidence also suggests presence of immune privileges in non-pathological stem cells in addition to their other defense mechanisms against damaging factors. This similarity between pathological and normal stem cells raises the question of why stem cells have such a potentially dangerous property. Regulation of vital processes of autoimmunity control and regeneration realized through interactions between immune cells, stem cells, and their microenvironment are reviewed in this work as causes of formation of the stem cell immune privilege. Deep mutual integration between regulations of stem and immune cells is noted. Considering diversity and complexity of mutual regulation of stem cells, their microenvironment, and immune system, I suggest the term "stem system".


Asunto(s)
Privilegio Inmunológico , Neoplasias , Humanos , Neoplasias/patología , Células Madre Neoplásicas/patología , Microambiente Tumoral
13.
Int J Mol Sci ; 24(15)2023 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-37569464

RESUMEN

The eye is an immune privileged tissue that insulates the visual system from local and systemic immune provocation to preserve homeostatic functions of highly specialized retinal neural cells. If immune privilege is breached, immune stimuli will invade the eye and subsequently trigger acute inflammatory responses. Local resident microglia become active and release numerous immunological factors to protect the integrity of retinal neural cells. Although acute inflammatory responses are necessary to control and eradicate insults to the eye, chronic inflammation can cause retinal tissue damage and cell dysfunction, leading to ocular disease and vision loss. In this review, we summarized features of immune privilege in the retina and the key inflammatory responses, factors, and intracellular pathways activated when retinal immune privilege fails, as well as a highlight of the recent clinical and research advances in ocular immunity and ocular vascular diseases including retinopathy of prematurity, age-related macular degeneration, and diabetic retinopathy.


Asunto(s)
Retinopatía Diabética , Oftalmopatías , Enfermedades de la Retina , Recién Nacido , Humanos , Privilegio Inmunológico , Retina/fisiología , Inflamación
14.
Int J Mol Sci ; 24(4)2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36834607

RESUMEN

Based on the postulate that glioblastoma (GBM) tumors generate anti-inflammatory prostaglandins and bile salts to gain immune privilege, we analyzed 712 tumors in-silico from three GBM transcriptome databases for prostaglandin and bile synthesis/signaling enzyme-transcript markers. A pan-database correlation analysis was performed to identify cell-specific signal generation and downstream effects. The tumors were stratified by their ability to generate prostaglandins, their competency in bile salt synthesis, and the presence of bile acid receptors nuclear receptor subfamily 1, group H, member 4 (NR1H4) and G protein-coupled bile acid receptor 1 (GPBAR1). The survival analysis indicates that tumors capable of prostaglandin and/or bile salt synthesis are linked to poor outcomes. Tumor prostaglandin D2 and F2 syntheses are derived from infiltrating microglia, whereas prostaglandin E2 synthesis is derived from neutrophils. GBMs drive the microglial synthesis of PGD2/F2 by releasing/activating complement system component C3a. GBM expression of sperm-associated heat-shock proteins appears to stimulate neutrophilic PGE2 synthesis. The tumors that generate bile and express high levels of bile receptor NR1H4 have a fetal liver phenotype and a RORC-Treg infiltration signature. The bile-generating tumors that express high levels of GPBAR1 are infiltrated with immunosuppressive microglia/macrophage/myeloid-derived suppressor cells. These findings provide insight into how GBMs generate immune privilege and may explain the failure of checkpoint inhibitor therapy and provide novel targets for treatment.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Masculino , Humanos , Prostaglandinas , Glioblastoma/metabolismo , Ácidos y Sales Biliares , Privilegio Inmunológico , Semen/metabolismo , Dinoprostona , Prostaglandinas Sintéticas , Neoplasias Encefálicas/metabolismo , Receptores Acoplados a Proteínas G/genética
15.
J Clin Invest ; 133(9)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36853799

RESUMEN

Multiple sclerosis (MS) is a complex disease of the CNS thought to require an environmental trigger. Gut dysbiosis is common in MS, but specific causative species are unknown. To address this knowledge gap, we used sensitive and quantitative PCR detection to show that people with MS were more likely to harbor and show a greater abundance of epsilon toxin-producing (ETX-producing) strains of C. perfringens within their gut microbiomes compared with individuals who are healthy controls (HCs). Isolates derived from patients with MS produced functional ETX and had a genetic architecture typical of highly conjugative plasmids. In the active immunization model of experimental autoimmune encephalomyelitis (EAE), where pertussis toxin (PTX) is used to overcome CNS immune privilege, ETX can substitute for PTX. In contrast to PTX-induced EAE, where inflammatory demyelination is largely restricted to the spinal cord, ETX-induced EAE caused demyelination in the corpus callosum, thalamus, cerebellum, brainstem, and spinal cord, more akin to the neuroanatomical lesion distribution seen in MS. CNS endothelial cell transcriptional profiles revealed ETX-induced genes that are known to play a role in overcoming CNS immune privilege. Together, these findings suggest that ETX-producing C. perfringens strains are biologically plausible pathogens in MS that trigger inflammatory demyelination in the context of circulating myelin autoreactive lymphocytes.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Microbioma Gastrointestinal , Esclerosis Múltiple , Animales , Humanos , Clostridium perfringens/genética , Esclerosis Múltiple/genética , Privilegio Inmunológico , Linfocitos
16.
J Leukoc Biol ; 113(3): 288-304, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36805720

RESUMEN

The ocular tissue microenvironment is immune-privileged and uses multiple immunosuppressive mechanisms to prevent the induction of inflammation. The retinal pigment epithelium plays an essential role in ocular immune privilege. In addition to serving as a blood barrier separating the fenestrated choriocapillaris from the retina, the retinal pigment epithelium is a source of immunosuppressive cytokines and membrane-bound negative regulators that modulate the activity of immune cells within the retina. This article reviews the current understanding of how retinal pigment epithelium cells mediate immune regulation, focusing on the changes under pathologic conditions.


Asunto(s)
Privilegio Inmunológico , Retina , Humanos , Inflamación , Células Epiteliales , Pigmentos Retinianos
17.
Cartilage ; 13(4): 119-132, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36250484

RESUMEN

The increasing prevalence of degenerative cartilage disorders in young patients is a growing public concern worldwide. Cartilage's poor innate regenerative capacity has inspired the exploration and development of cartilage replacement treatments such as tissue-engineered cartilages and osteochondral implants as potential solutions to cartilage loss. The clinical application of tissue-engineered implants is hindered by the lack of long-term follow-up demonstrating efficacy, biocompatibility, and bio-integration. The historically reported immunological privilege of cartilage tissue was based on histomorphological observations pointing out the lack of vascularity and the presence of a tight extracellular matrix. However, clinical studies in humans and animals do not unequivocally support the immune-privilege theory. More in-depth studies on cartilage immunology are needed to make clinical advances such as tissue engineering more applicable. This review analyzes the literature that supports and opposes the concept that cartilage is an immune-privileged tissue and provides insight into mechanisms conferring various degrees of immune privilege to other, more in-depth studied tissues such as testis, eyes, brain, and cancer.


Asunto(s)
Cartílago , Privilegio Inmunológico , Masculino , Animales , Humanos , Ingeniería de Tejidos , Matriz Extracelular
18.
Orphanet J Rare Dis ; 17(1): 378, 2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-36253797

RESUMEN

BACKGROUND: Retinitis pigmentosa (RP) is a hereditary retinal disease which leads to visual impairment. The onset and progression of RP has physiological consequences that affects the ocular environment. Some of the key non-genetic factors which hasten the retinal degeneration in RP include oxidative stress, hypoxia and ocular inflammation. In this study, we investigated the status of the ocular immune privilege during retinal degeneration and the effect of ocular immune changes on the peripheral immune system in RP. We assessed the peripheral blood mononuclear cell stimulation by retinal antigens and their immune response status in RP patients. Subsequently, we examined alterations in ocular immune privilege machineries which may contribute to ocular inflammation and disease progression in rd1 mouse model. RESULTS: In RP patients, we observed a suppressed anti-inflammatory response to self-retinal antigens, thereby indicating a deviated response to self-antigens. The ocular milieu in rd1 mouse model indicated a significant decrease in immune suppressive ligands and cytokine TGF-B1, and higher pro-inflammatory ocular protein levels. Further, blood-retinal-barrier breakdown due to decrease in the expression of tight junction proteins was observed. The retinal breach potentiated pro-inflammatory peripheral immune activation against retinal antigens and caused infiltration of the peripheral immune cells into the ocular tissue. CONCLUSIONS: Our studies with RP patients and rd1 mouse model suggest that immunological consequences in RP is a contributing factor in the progression of retinal degeneration. The ocular inflammation in the RP alters the ocular immune privilege mechanisms and peripheral immune response. These aberrations in turn create an auto-reactive immune environment and accelerate retinal degeneration.


Asunto(s)
Degeneración Retiniana , Retinitis Pigmentosa , Animales , Antiinflamatorios , Autoantígenos , Citocinas , Modelos Animales de Enfermedad , Privilegio Inmunológico , Inflamación , Leucocitos Mononucleares/metabolismo , Ratones , Proteínas de Uniones Estrechas
19.
Biol Reprod ; 107(6): 1565-1579, 2022 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-36001358

RESUMEN

Sertoli cells (SCs), the only somatic cells in the seminiferous tubules, facilitate the maintenance of testicular immune privilege through the formation of the blood-testis barrier (BTB) and the expression of immunoregulatory factors. Rho guanosine exchange factor 15 (ARHGEF15) is a member of the guanosine exchange factors, which are involved in cell migration, cell polarity, and cell cycle progression via activation of Rho GTPases. This study investigated the functional role of ARHGEF15 in SCs during spermatogenesis using SC-specific Arhgef15 knockout mice. The results revealed that Arhgef15 deficiency in SCs affected the localization of SC nuclei, disrupted BTB integrity, and led to premature shedding of germ cells. In Arhgef15flox/flox/Amh-Cre+ mice, the ultrastructure of the round spermatids was impaired, accompanied by acrosome degeneration, acrosomal vesicle shedding, and atrophic nuclei. Consequently, the percentage of abnormal sperm in the Arhgef15flox/flox/Amh-Cre+ epididymis was markedly elevated. RNA-sequencing analysis revealed that most of the differentially expressed genes in SCs of Arhgef15flox/flox/Amh-Cre+ mice were associated with immunity. Further study revealed that the sera of Arhgef15flox/flox/Amh-Cre+ mice showed immunoreactivity against testicular lysate of wild-type mice, indicating the production of antibodies against testicular autoantigens in Arhgef15flox/flox/Amh-Cre+ mice. In conclusion, the specific deletion of Arhgef15 in SCs of mice leads to sperm abnormality, probably by disrupting the testicular immune homeostasis.


Asunto(s)
Privilegio Inmunológico , Células de Sertoli , Masculino , Ratones , Animales , Células de Sertoli/metabolismo , Semen , Testículo/metabolismo , Espermatogénesis/fisiología , Espermatozoides/metabolismo , Ratones Noqueados , Guanosina/metabolismo
20.
Cancer Gene Ther ; 29(12): 1951-1960, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35902729

RESUMEN

Monocytes/macrophages, a plastic and heterogeneous cell population of the tumor microenvironment (TME), can constitute a major component of most solid tumors. Under the pressure of rapid proliferation of the tumor, monocytes/macrophages can be educated and foster immune tolerance via metabolic reprogramming. Our studies have shown that the activation of FABP5, a lipid-binding protein, decreases the rate of ß-oxidation causing the accumulation of lipid droplets in monocytes. We found that hepatocellular carcinoma cells (HCC) increased IL-10 secretion by monocytes, which depended on the expression of FABP5 and suppressing of the PPARα pathway. Moreover, the elevated level of IL-10 promotes PD-L1 expression on Treg cells via the JNK-STAT3 pathway activation. We also observed that elevation of FABP5 in monocytes was negatively related to HCC patients' overall survival time. Thus, FABP5 promotes monocyte/macrophage lipid accumulation, fosters immune tolerance formation, and might represent itself as a therapeutic target in both tumor-associated monocytes (TAMs) and cancer cells.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patología , Monocitos/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Interleucina-10/metabolismo , Interleucina-10/uso terapéutico , Privilegio Inmunológico , Neoplasias Hepáticas/metabolismo , Linfocitos T Reguladores/metabolismo , Macrófagos , Microambiente Tumoral , Lípidos/uso terapéutico , Línea Celular Tumoral , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Proteínas de Unión a Ácidos Grasos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA