Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 8(4)2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36633909

RESUMEN

Newborns are at high risk of developing neonatal sepsis, particularly if born prematurely. This has been linked to divergent requirements the immune system has to fulfill during intrauterine compared with extrauterine life. By transcriptomic analysis of fetal and adult neutrophils, we shed new light on the molecular mechanisms of neutrophil maturation and functional adaption during fetal ontogeny. We identified an accumulation of differentially regulated genes within the noncanonical NF-κB signaling pathway accompanied by constitutive nuclear localization of RelB and increased surface expression of TNF receptor type II in fetal neutrophils, as well as elevated levels of lymphotoxin α in fetal serum. Furthermore, we found strong upregulation of the negative inflammatory regulator A20 (Tnfaip3) in fetal neutrophils, which was accompanied by pronounced downregulation of the canonical NF-κB pathway. Functionally, overexpressing A20 in Hoxb8 cells led to reduced adhesion of these neutrophil-like cells in a flow chamber system. Conversely, mice with a neutrophil-specific A20 deletion displayed increased inflammation in vivo. Taken together, we have uncovered constitutive activation of the noncanonical NF-κB pathway with concomitant upregulation of A20 in fetal neutrophils. This offers perfect adaption of neutrophil function during intrauterine fetal life but also restricts appropriate immune responses particularly in prematurely born infants.


Asunto(s)
FN-kappa B , Infiltración Neutrófila , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Animales , Humanos , Ratones , Inflamación , Sepsis Neonatal/genética , Sepsis Neonatal/metabolismo , Infiltración Neutrófila/genética , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
2.
ACS Biomater Sci Eng ; 9(6): 2891-2901, 2023 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-33914503

RESUMEN

Oral protein delivery technologies often depend on encapsulating or enclosing the protein cargo to protect it against pH-driven degradation in the stomach or enzymatic digestion in the small intestine. An emergent methodology is to encapsulate therapeutics in microscale, asymmetric, planar microparticles, referred to as microdevices. Previous work has shown that, compared to spherical particles, planar microdevices have longer residence times in the GI tract, but it remains unclear how specific design choices (e.g., material selection, particle diameter) impact microdevice behavior in vivo. Recent advances in microdevice fabrication through picoliter printing have expanded the range of device sizes that can be fabricated in a rapid manner. However, relatively little work has explored how device size governs their behavior in the intestinal environment. In this study, we probe the impact of geometry of planar microdevices on their transit and accumulation in the murine GI tract. Additionally, we present a strategy to label, image, and quantify these distributions in intact tissue in a continuous manner, enabling a more detailed understanding of device distribution and transit kinetics than previously possible. We show that smaller particles (194.6 ± 7 µm.diameter) tend to empty from the stomach faster than midsize (293.2 ± 7 µm.diameter) and larger devices (440.9 ± 9 µm.diameter) and that larger devices distribute more broadly in the GI tract and exit slower than other geometries. In general, we observed an inverse correlation between device diameter and GI transit rate. These results inform the future design of drug delivery systems, using particle geometry as an engineering design parameter to control device accumulation and distribution in the GI tract. Additionally, our image analysis process provides greater insight into the tissue level distribution and transit of particle populations. Using this technique, we demonstrate that microdevices act and translocate independently, as opposed to transiting in one homogeneous mass, meaning that target sites will likely be exposed to devices multiple times over the course of hours post administration. This imaging technique and associated findings enable data-informed design of future particle delivery systems, allowing orthogonal control of transit and distribution kinetics in vivo independent of material and cargo selection.


Asunto(s)
Sistemas de Liberación de Medicamentos , Tracto Gastrointestinal , Ratones , Animales , Sistemas de Liberación de Medicamentos/métodos
3.
Cell Rep ; 39(9): 110891, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35649365

RESUMEN

Resident microbes in skin and gut predominantly impact local immune cell function during homeostasis. However, colitis-associated neutrophilic skin disorders suggest possible breakdown of this compartmentalization with disease. Using a model wherein neonatal skin colonization by Staphylococcus epidermidis facilitates generation of commensal-specific tolerance and CD4+ regulatory T cells (Tregs), we ask whether this response is perturbed by gut inflammation. Chemically induced colitis is accompanied by intestinal expansion of S. epidermidis and reduces gut-draining lymph node (dLN) commensal-specific Tregs. It also results in reduced commensal-specific Tregs in skin and skin-dLNs and increased skin neutrophils. Increased CD4+ circulation between gut and skin dLN suggests that the altered cutaneous response is initiated in the colon, and resistance to colitis-induced effects in Cd4creIl1r1fl/fl mice implicate interleukin (IL)-1 in mediating the altered commensal-specific response. These findings provide mechanistic insight into observed connections between inflammatory skin and intestinal diseases.


Asunto(s)
Colitis , Inmunidad , Animales , Colitis/inducido químicamente , Inflamación , Ratones , Piel , Staphylococcus epidermidis , Linfocitos T Reguladores
4.
Sci Rep ; 12(1): 5517, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35365713

RESUMEN

Fecal microbiota transplant is a promising therapy for ulcerative colitis. Parameters maximizing effectiveness and tolerability are not yet clear, and it is not known how import the transmission of donor microbes to patients is. Here (clinicaltrails.gov: NCT03006809) we have tested the effects of antibiotic pretreatment and compared two modes of maintenance dose delivery, capsules versus enema, in a randomized, pilot, open-label, 2 × 2 factorial design with 22 patients analyzed with mild to moderate UC. Clinically, the treatment was well-tolerated with favorable safety profile. Of patients who received antibiotic pretreatment, 6 of 11 experienced remission after 6 weeks of treatment, versus 2 of 11 non-pretreated patients (log odds ratio: 1.69, 95% confidence interval: -0.25 to 3.62). No significant differences were found between maintenance dosing via capsules versus enema. In exploratory analyses, microbiome turnover at both the species and strain levels was extensive and significantly more pronounced in the pretreated patients. Associations were also revealed between taxonomic turnover and changes in the composition of primary and secondary bile acids. Together these findings suggest that antibiotic pretreatment contributes to microbiome engraftment and possibly clinical effectiveness, and validate longitudinal strain tracking as a powerful way to monitor the dynamics and impact of microbiota transfer.


Asunto(s)
Colitis Ulcerosa , Microbioma Gastrointestinal , Antibacterianos/uso terapéutico , Colitis Ulcerosa/etiología , Colitis Ulcerosa/terapia , Trasplante de Microbiota Fecal , Heces , Humanos , Inducción de Remisión
5.
Cell Discov ; 8(1): 34, 2022 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-35411038

RESUMEN

Microgliosis is a hallmark of many neurological diseases, including Alzheimer's disease, stroke, seizure, traumatic brain and spinal cord injuries, and peripheral and optic nerve injuries. Recent studies have shown that the newly self-renewed microglia have specific neurological functions. However, the mechanism of adult microglia proliferation remains largely unclear. Here, with single-cell RNA sequencing, flow cytometry, and immunohistochemistry, we demonstrate that the sciatic nerve injury induced two distinct phases of microglia proliferation in mouse spinal cord, each with different gene expression profiles. We demonstrate that the transcription factor Myc was transiently upregulated in spinal cord microglia after nerve injury to mediate an early phase microglia proliferation. On the other hand, we reveal that the tumor-necrosis factor alpha-induced protein 3 (Tnfaip3) was downregulated to mediate the Myc-independent late-phase microglia proliferation. We show that cyclin dependent kinase 1, a kinase with important function in the M phase of the cell cycle, was involved only in the early phase. We reveal that although the early phase was neither necessary nor sufficient for the late phase proliferation, the late-phase suppressed the early phase microglia proliferation in the spinal cord. Finally, we demonstrate that the termination of spinal cord microglia proliferation required both Myc and Tnfaip3 to resume their baseline expression. Thus, we have delineated an interactive signaling network in the proliferation of differentiated microglia.

6.
Biochem Biophys Res Commun ; 592: 74-80, 2022 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-35032835

RESUMEN

Crohn's disease is an inflammatory disease of the gut caused by a complex interplay among genetic, microbial, and environmental factors. The intestinal tract is constantly exposed to metals and other trace elements ingested as food. Synchrotron radiation-induced X-ray fluorescence spectroscopy and X-ray absorption fine structure analysis revealed the deposition of nickel particles within Crohn's disease tissue specimens. After nickel particle stimulation, THP-1 cells showed filopodia formation and autophagic vacuoles containing lipid bodies. Nickel particles precipitated colitis in mice bearing mutations of the IBD susceptibility protein A20/TNFAIP3. Nickel particles also exacerbated dextran sulfate sodium-induced colitis in mice harboring myeloid cell-specific Atg5 deficiency. These findings illustrate that nickel particle ingestion may worsen Crohn's disease by perturbing autophagic processes in the intestine, providing new insights into environmental factors in Crohn's disease pathogenesis.


Asunto(s)
Enfermedad de Crohn/patología , Progresión de la Enfermedad , Inflamación/patología , Intestinos/patología , Níquel/toxicidad , Animales , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/metabolismo , Sulfato de Dextran , Susceptibilidad a Enfermedades , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/patología , Macrófagos/ultraestructura , Ratones Endogámicos C57BL , Células THP-1 , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
7.
J Clin Invest ; 132(5)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35077396

RESUMEN

Anti-TNF antibodies are effective for treating patients with inflammatory bowel disease (IBD), but many patients fail to respond to anti-TNF therapy, highlighting the importance of TNF-independent disease. We previously demonstrated that acute deletion of 2 IBD susceptibility genes, A20 (Tnfaip3) and Abin-1 (Tnip1), in intestinal epithelial cells (IECs) sensitized mice to both TNF-dependent and TNF-independent death. Here we show that TNF-independent IEC death after A20 and Abin-1 deletion was rescued by germ-free derivation or deletion of MyD88, while deletion of Trif provided only partial protection. Combined deletion of Ripk3 and Casp8, which inhibits both apoptotic and necroptotic death, completely protected against death after acute deletion of A20 and Abin-1 in IECs. A20- and Abin-1-deficient IECs were sensitized to TNF-independent, TNFR1-mediated death in response to lymphotoxin α (LTα) homotrimers. Blockade of LTα in vivo reduced weight loss and improved survival when combined with partial deletion of MyD88. Biopsies of inflamed colon mucosa from patients with IBD exhibited increased LTA and IL1B expression, including a subset of patients with active colitis on anti-TNF therapy. These data show that microbial signals, MyD88, and LTα all contribute to TNF-independent intestinal injury.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Linfotoxina-alfa , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis , Células Epiteliales/metabolismo , Epitelio/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/metabolismo , Linfotoxina-alfa/genética , Linfotoxina-alfa/metabolismo , Linfotoxina-alfa/farmacología , Ratones , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Inhibidores del Factor de Necrosis Tumoral
8.
Med ; 2(1): 14-15, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-35590128

RESUMEN

Single-cell transcriptomic profiling has enhanced the description of several diseases. Beyond improving our understanding of the pathophysiology of these diseases, these approaches can lead to clues about therapeutic approaches. In a recent issue of Cell, Wang et al. describe immune perturbations in livers of biliary atresia patients that implicate specific autoimmune pathways of disease.1.


Asunto(s)
Atresia Biliar , Enfermedades Raras , Atresia Biliar/metabolismo , Perfilación de la Expresión Génica , Humanos , Hígado/metabolismo , Enfermedades Raras/genética
10.
Front Immunol ; 11: 536442, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33224133

RESUMEN

Hematopoietic stem and progenitor cells (HSPC) experience a functional decline in response to chronic inflammation or aging. Haploinsufficiency of A20, or TNFAIP3, an innate immune regulator, is associated with a variety of autoimmune, inflammatory, and hematologic malignancies. Based on a prior analysis of epigenomic and transcriptomic changes during normal human aging, we find that the expression of A20 is significantly reduced in aged HSPC as compared to young HSPC. Here, we show that the partial reduction of A20 expression in young HSPC results in characteristic features of aging. Specifically, heterozygous deletion of A20 in hematopoietic cells resulted in expansion of the HSPC pool, reduced HSPC fitness, and myeloid-biased hematopoiesis. These findings suggest that altered expression of A20 in HSPC contributes to an aging-like phenotype, and that there may be a common underlying mechanism for diminished HSPC function between inflammatory states and aging.


Asunto(s)
Envejecimiento/inmunología , Hematopoyesis , Células Madre Hematopoyéticas/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/inmunología , Envejecimiento/genética , Envejecimiento/patología , Animales , Eliminación de Gen , Células Madre Hematopoyéticas/patología , Heterocigoto , Ratones , Ratones Transgénicos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética
11.
Adv Immunol ; 148: 1-48, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33190732

RESUMEN

A20/TNFAIP3 is a TNF induced gene that plays a profound role in preserving cellular and organismal homeostasis (Lee, et al., 2000; Opipari etal., 1990). This protein has been linked to multiple human diseases via genetic, epigenetic, and an emerging series of patients with mono-allelic coding mutations. Diverse cellular functions of this pleiotropically expressed protein include immune-suppressive, anti-inflammatory, and cell protective functions. The A20 protein regulates ubiquitin dependent cell signals; however, the biochemical mechanisms by which it performs these functions is surprisingly complex. Deciphering these cellular and biochemical facets of A20 dependent biology should greatly improve our understanding of murine and human disease pathophysiology as well as unveil new mechanisms of cell and tissue biology.


Asunto(s)
Inflamación/inmunología , Mutación/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Animales , Autoinmunidad , Muerte Celular , Homeostasis , Humanos , Ratones
12.
Nat Immunol ; 21(5): 535-545, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32313245

RESUMEN

Despite evidence of chronic inflammation in myelodysplastic syndrome (MDS) and cell-intrinsic dysregulation of Toll-like receptor (TLR) signaling in MDS hematopoietic stem and progenitor cells (HSPCs), the mechanisms responsible for the competitive advantage of MDS HSPCs in an inflammatory milieu over normal HSPCs remain poorly defined. Here, we found that chronic inflammation was a determinant for the competitive advantage of MDS HSPCs and for disease progression. The cell-intrinsic response of MDS HSPCs, which involves signaling through the noncanonical NF-κB pathway, protected these cells from chronic inflammation as compared to normal HSPCs. In response to inflammation, MDS HSPCs switched from canonical to noncanonical NF-κB signaling, a process that was dependent on TLR-TRAF6-mediated activation of A20. The competitive advantage of TLR-TRAF6-primed HSPCs could be restored by deletion of A20 or inhibition of the noncanonical NF-κB pathway. These findings uncover the mechanistic basis for the clonal dominance of MDS HSPCs and indicate that interfering with noncanonical NF-κB signaling could prevent MDS progression.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Inflamación/inmunología , Síndromes Mielodisplásicos/inmunología , FN-kappa B/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Anciano , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Transgénicos , Mielopoyesis , FN-kappa B/genética , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/genética , Receptores Toll-Like/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
13.
Nat Immunol ; 21(4): 422-433, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32205880

RESUMEN

A20 is an anti-inflammatory protein that is strongly linked to human disease. Here, we find that mice expressing three distinct targeted mutations of A20's zinc finger 7 (ZF7) ubiquitin-binding motif uniformly developed digit arthritis with features common to psoriatic arthritis, while mice expressing point mutations in A20's OTU or ZF4 motifs did not exhibit this phenotype. Arthritis in A20ZF7 mice required T cells and MyD88, was exquisitely sensitive to tumor necrosis factor and interleukin-17A, and persisted in germ-free conditions. A20ZF7 cells exhibited prolonged IκB kinase activity that drove exaggerated transcription of late-phase nuclear factor-κB response genes in vitro and in prediseased mouse paws in vivo. In addition, mice expressing double-mutant A20 proteins in A20's ZF4 and ZF7 motifs died perinatally with multi-organ inflammation. Therefore, A20's ZF4 and ZF7 motifs synergistically prevent inflammatory disease in a non-catalytic manner.


Asunto(s)
Artritis Psoriásica/metabolismo , Inflamación/metabolismo , Ubiquitina/metabolismo , Animales , Células Cultivadas , Interleucina-17 , Ratones , Ratones Endogámicos C57BL , Mutación/genética , FN-kappa B/metabolismo , Unión Proteica/fisiología , Transducción de Señal/fisiología , Transcripción Genética/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitinación/fisiología , Dedos de Zinc/fisiología
14.
Dig Dis Sci ; 65(4): 1099-1106, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31302808

RESUMEN

AIMS: This pilot study assessed the efficacy, safety, and microbiome dynamics of fecal microbiota transplantation (FMT) for patients with chronic pouchitis. METHODS: A prospective open-label pilot study was performed at an academic center among pouchitis patients undergoing FMT. Patients received a minimum of a single FMT by pouchoscopy from healthy, screened donors. The primary outcome was clinical improvement in pouchitis assessed by patient survey at week 4. Secondary outcomes included decrease in total Pouchitis Disease Activity Index (PDAI) Score ≥ 3 at week 4, bowel movement frequency, ESR, CRP, fecal calprotectin, abdominal pain, and PDAI subscores including endoscopic and histologic changes. Stool samples were collected at baseline and 4 weeks post-FMT to assess bacterial microbiota using V4 16S rRNA sequencing. RESULTS: Nineteen patients were enrolled; however, 1 patient was lost to follow-up. No patients had a major adverse event or escalation of therapy related to FMT. Total PDAI scores, endoscopic scores, and histologic scores did not decrease significantly post-FMT. However, there was a statistically significant improvement in bowel movement (BM) frequency (9.25-7.25 BM/day, p = 0.03) and trend for improvement in abdominal pain to improve post-FMT (p = 0.05). Bacterial microbiota profiling revealed no distinct community-level changes post-FMT, though a small number of specific bacterial taxa significantly differed in relative abundance. CONCLUSIONS: A single FMT has a tolerable short-term safety profile and may be associated with a decrease in bowel movements in patients with chronic pouchitis; however, no robust endoscopic or histologic changes were observed.


Asunto(s)
Endoscopía Gastrointestinal/métodos , Trasplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal/fisiología , Reservoritis/diagnóstico , Reservoritis/terapia , Adolescente , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Reservoritis/microbiología , Estudios Prospectivos , Adulto Joven
15.
United European Gastroenterol J ; 7(6): 807-814, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31316785

RESUMEN

Background: Emerging trials suggest fecal microbiota transplantation (FMT) is a promising treatment for ulcerative colitis; however, there is a paucity of data in Crohn disease (CD). Objective: The objectives of this article are to determine whether single-dose FMT improves clinical and endoscopic outcomes in CD patients and to identify meaningful changes in the microbiome in response to FMT. Methods: We performed a prospective, open-label, single-center study. Ten CD patients underwent FMT and were evaluated for clinical response (defined as decrease in Harvey-Bradshaw Index score ≥3 at one month post-FMT) and microbiome profile (16S ribosomal RNA sequencing) at one month post-FMT. Results: Three of 10 patients responded to FMT. Two of 10 patients had significant adverse events requiring escalation of therapy. On microbiome analysis, bacterial communities of responders had increased relative abundance of bacteria commonly found in donor gut microbiota. Conclusions: Single-dose FMT in this cohort of CD patients showed modest effect and potential for harm. Responders tended to have lower baseline alpha diversity, suggesting baseline perturbation of microbiota may be an indicator of potential responders to FMT in this patient population. Controlled trials are needed to further assess the efficacy and safety of FMT in CD and determine whether FMT is a viable option in this patient population.Clinicaltrials.gov number: NCT02460705.


Asunto(s)
Enfermedad de Crohn/terapia , Trasplante de Microbiota Fecal , Adolescente , Adulto , Anciano , Enfermedad de Crohn/etiología , Trasplante de Microbiota Fecal/efectos adversos , Trasplante de Microbiota Fecal/métodos , Femenino , Microbioma Gastrointestinal , Humanos , Masculino , Metagenómica/métodos , Persona de Mediana Edad , ARN Ribosómico 16S/genética , Resultado del Tratamiento , Adulto Joven
16.
PLoS One ; 14(7): e0218999, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31295268

RESUMEN

Microbial dysbiosis commonly occurs in patients with inflammatory bowel diseases (IBD). Exogenous causes of dysbiosis such as antibiotics and diet are well described, but host derived causes are understudied. A20 is a potent regulator of signals triggered by microbial pattern molecules, and A20 regulates susceptibility to intestinal inflammation in mice and in humans. We now report that mice lacking A20 expression in dendritic cells, A20FL/FL CD11c-Cre mice (or A20dDC mice), spontaneously develop colitogenic intestinal dysbiosis that is evident upon weaning and precedes the onset of colitis. Intestines from A20dDC mice express increased amounts of Reg3ß and Reg3γ, but not Ang4. A20 deficient DCs promote gut microbiota perturbation in the absence of adaptive lymphocytes. Moreover, A20 deficient DCs directly induce expression of Reg3ß and Reg3γ but not Ang 4 in normal intestinal epithelial cell enteroid cultures in the absence of other cell types. These findings reveal a pathophysiological pathway in which defective expression of an IBD susceptibility gene in DCs drives aberrant expression of anti-bacterial peptides and luminal dysbiosis that in turn confers host susceptibility to intestinal inflammation.


Asunto(s)
Disbiosis/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Animales , Antibacterianos/farmacología , Células Dendríticas/microbiología , Disbiosis/genética , Disbiosis/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis , Humanos , Inflamación/genética , Inflamación/microbiología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/microbiología , Intestinos/microbiología , Ratones , Ratones Noqueados , Proteínas Asociadas a Pancreatitis/genética , Péptidos/farmacología , Ribonucleasa Pancreática/genética , Simbiosis/efectos de los fármacos
17.
Cell Immunol ; 340: 103914, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31030956

RESUMEN

A20, also known as TNFAIP3, is a potent regulator of ubiquitin (Ub) dependent signals. A20 prevents multiple human diseases, indicating that the critical functions of this protein are clinically as well as biologically impactful. As revealed by mouse models, cell specific functions of A20 are linked to its ability to regulate diverse signaling pathways. Aberrant expression or functions of A20 in specific cell types underlie divergent disease outcomes. Discernment of A20's biochemical functions and their phenotypic outcomes will contribute to our understanding of how ubiquitination is regulated, how Ub mediated functions can prevent disease, and will pave the way for future therapeutic interventions.


Asunto(s)
Artritis Reumatoide/inmunología , Asma/inmunología , Lupus Eritematoso Sistémico/inmunología , Procesamiento Proteico-Postraduccional , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Animales , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Artritis Reumatoide/prevención & control , Asma/genética , Asma/patología , Asma/prevención & control , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/patología , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Lupus Eritematoso Sistémico/prevención & control , Terapia Molecular Dirigida/métodos , FN-kappa B/genética , FN-kappa B/inmunología , Transducción de Señal , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/inmunología , Ubiquitinación
18.
J Immunol ; 202(7): 2044-2056, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30760622

RESUMEN

Deregulated immune response to a dysbiotic resident microflora within the oral cavity leads to chronic periodontal disease, local tissue destruction, and various systemic complications. To preserve tissue homeostasis, inflammatory signaling pathways involved in the progression of periodontitis must be tightly regulated. A20 (TNFAIP3), a ubiquitin-editing enzyme, has emerged as one of the key regulators of inflammation. Yet, the function of A20 in the oral mucosa and the biological pathways in which A20 mitigates periodontal inflammation remain elusive. Using a combination of in vivo and ex vivo disease models, we report in this study that A20 regulates inflammatory responses to a keystone oral bacterium, Porphyromonas gingivalis, and restrains periodontal inflammation through its effect on NF-κB signaling and cytokine production. Depletion of A20 using gene editing in human macrophage-like cells (THP-1) significantly increased cytokine secretion, whereas A20 overexpression using lentivirus infection dampened the cytokine production following bacterial challenge through modulating NF-κB activity. Similar to human cells, bone marrow-derived macrophages from A20-deficient mice infected with P. gingivalis displayed increased NF-κB activity and cytokine production compared with the cells isolated from A20-competent mice. Subsequent experiments using a murine ligature-induced periodontitis model showed that even a partial loss of A20 promotes an increased inflammatory phenotype and more severe bone loss, further verifying the critical function of A20 in the oral mucosa. Collectively, to our knowledge, these findings reveal the first systematic evidence of a physiological role for A20 in the maintenance of oral tissue homeostasis as a negative regulator of inflammation.


Asunto(s)
Inflamación/inmunología , Mucosa Bucal/inmunología , FN-kappa B/inmunología , Periodontitis/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/inmunología , Animales , Células HEK293 , Humanos , Inmunidad Mucosa/inmunología , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Mucosa Bucal/metabolismo , FN-kappa B/metabolismo , Periodontitis/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
19.
J Biol Chem ; 293(47): 18285-18295, 2018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30282802

RESUMEN

OTUB1 is a deubiquitinating enzyme that cleaves Lys-48-linked polyubiquitin chains and also regulates ubiquitin signaling through a unique, noncatalytic mechanism. OTUB1 binds to a subset of E2 ubiquitin-conjugating enzymes and inhibits their activity by trapping the E2∼ubiquitin thioester and preventing ubiquitin transfer. The same set of E2s stimulate the deubiquitinating activity of OTUB1 when the E2 is not charged with ubiquitin. Previous studies have shown that, in cells, OTUB1 binds to E2-conjugating enzymes of the UBE2D (UBCH5) and UBE2E families, as well as to UBE2N (UBC13). Cellular roles have been identified for the interaction of OTUB1 with UBE2N and members of the UBE2D family, but not for interactions with UBE2E E2 enzymes. We report here a novel role for OTUB1-E2 interactions in modulating E2 protein ubiquitination. We observe that Otub1-/- knockout mice exhibit late-stage embryonic lethality. We find that OTUB1 depletion dramatically destabilizes the E2-conjugating enzyme UBE2E1 (UBCH6) in both mouse and human OTUB1 knockout cell lines. Of note, this effect is independent of the catalytic activity of OTUB1, but depends on its ability to bind to UBE2E1. We show that OTUB1 suppresses UBE2E1 autoubiquitination in vitro and in cells, thereby preventing UBE2E1 from being targeted to the proteasome for degradation. Taken together, we provide evidence that OTUB1 rescues UBE2E1 from degradation in vivo.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Secuencias de Aminoácidos , Animales , Cisteína Endopeptidasas/genética , Enzimas Desubicuitinizantes , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Estabilidad Proteica , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/química , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitinación
20.
Clin Transl Gastroenterol ; 9(7): 172, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30065242

RESUMEN

The original version of this article contained an error in Fig. 2, in which part of the text in the legend was omitted. This has now been corrected in the PDF and HTML versions of the paper.Furthermore, the figure legends were missing for the Supplementary figure files. The HTML has now been updated to include a corrected version of the Supplementary Information.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA