Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
1.
J Am Pharm Assoc (2003) ; : 102085, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38582383

RESUMEN

BACKGROUND: Obesity is a prevalent medical condition that can contribute to an increased risk of developing serious comorbidities, leading to an increase in hospitalizations, morbidity, and mortality. Many of these medical conditions can be improved with weight loss. OBJECTIVES: This project was designed to improve weight loss outcomes and assess the utilization of services provided by clinical pharmacists for collaborative weight loss management. METHODS: The study design was a single-center, retrospective, and quality improvement study within three outpatient clinics at Tufts Medical Center (TMC). Patients referred to the pharmacist-led weight loss service from September 1 to October 31, 2022, and continued care for up to 6 months were included. Pharmacist services included selection of weight loss medication, assistance with medication access, device teaching and dose titration, lifestyle counseling, and follow-up. The primary outcome was percent weight loss from baseline. RESULTS: Seventy-nine patients were referred to the pharmacist-led weight loss service. Mean age of patients was 51 years (SD ±13). Sixty-one patients were female (77.2 %). Median baseline weight was 105.5 kg (IQR 93.1 to 120.5 kg) and BMI 38.1 kg/m2 (IQR 33.9 to 43.5 kg/m2). The median percent weight loss from baseline through the end of the study duration was -8.0% (IQR -3.1 to -12.1%). CONCLUSION: Pharmacists were able to effectively provide weight loss care through a pharmacist-led weight loss service by aiding in medication access, providing education on devices and lifestyle management, and engaging in frequent follow-up. Future directions of this study include expansion of the pharmacist-led weight loss service to other ambulatory care clinics within TMC.

2.
Ann Rheum Dis ; 83(3): 360-371, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-37932009

RESUMEN

OBJECTIVES: To evaluate the safety and efficacy of remibrutinib in patients with moderate-to-severe Sjögren's syndrome (SjS) in a phase 2 randomised, double-blind trial (NCT04035668; LOUiSSE (LOU064 in Sjögren's Syndrome) study). METHODS: Eligible patients fulfilling 2016 American College of Rheumatology/European League Against Rheumatism (EULAR) criteria for SjS, positive for anti-Ro/Sjögren's syndrome-related antigen A antibodies, with moderate-to-severe disease activity (EULAR Sjögren's Syndrome Disease Activity Index (ESSDAI) (based on weighted score) ≥ 5, EULAR Sjögren's Syndrome Patient Reported Index (ESSPRI) ≥ 5) received remibrutinib (100 mg) either one or two times a day, or placebo for the 24-week study treatment period. The primary endpoint was change from baseline in ESSDAI at week 24. Key secondary endpoints included change from baseline in ESSDAI over time, change from baseline in ESSPRI over time and safety of remibrutinib in SjS. Key exploratory endpoints included changes to the salivary flow rate, soluble biomarkers, blood transcriptomic and serum proteomic profiles. RESULTS: Remibrutinib significantly improved ESSDAI score in patients with SjS over 24 weeks compared with placebo (ΔESSDAI -2.86, p=0.003). No treatment effect was observed in ESSPRI score (ΔESSPRI 0.17, p=0.663). There was a trend towards improvement of unstimulated salivary flow with remibrutinib compared with placebo over 24 weeks. Remibrutinib had a favourable safety profile in patients with SjS over 24 weeks. Remibrutinib induced significant changes in gene expression in blood, and serum protein abundance compared with placebo. CONCLUSIONS: These data show preliminary efficacy and favourable safety of remibrutinib in a phase 2 trial for SjS.


Asunto(s)
Pirimidinas , Síndrome de Sjögren , Humanos , Síndrome de Sjögren/tratamiento farmacológico , Síndrome de Sjögren/complicaciones , Proteómica , Anticuerpos , Índice de Severidad de la Enfermedad
3.
Eur J Drug Metab Pharmacokinet ; 48(5): 553-566, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37532923

RESUMEN

BACKGROUND AND OBJECTIVE: MHV370, a dual antagonist of human Toll-like receptors (TLR) 7 and 8, suppresses cytokines and interferon-stimulated genes in vitro and in vivo, and  has demonstrated efficacy in murine models of lupus. This first-in-human study aimed to evaluate the safety, tolerability, pharmacokinetics and pharmacodynamics of single and multiple doses of MHV370 in healthy adults, as well as the effects of food consumption on a single dose of MHV370. METHODS: This was a phase 1, randomised, placebo-controlled study conducted in three parts. In part A, participants received (3:1) a single ascending dose (SAD) of 1, 3, 10, 20, 40, 80, 160, 320, 640 and 1000 mg MHV370 or placebo. In part B, participants received (3:1) multiple ascending doses (MAD) of 25, 50, 100, 200 and 400 mg MHV370 twice daily (b.i.d) or placebo for 14 days. In part C, participants received an open-label single dose of 200 mg MHV370 under fasted or fed conditions. Safety, pharmacokinetic and pharmacodynamic parameters were evaluated. RESULTS: MHV370 was well tolerated, and no safety signal was observed in the study. No dose-limiting adverse events occurred across the dose range evaluated. Plasma concentrations of MHV370 increased with dose (mean [SD] maximum plasma concentrations ranged from 0.97 [0.48] to 1670 [861.0] ng/mL for SAD of 3-1000 mg, 29.5 [7.98] to 759 [325.0] ng/mL for MAD of 25-400 mg b.i.d. on day 1). The intake of food did not have a relevant impact on the pharmacokinetics of MHV370. Pharmacodynamic data indicated time- and dose-dependent inhibition of TLR7-mediated CD69 expression on B cells (100% inhibition at 24 h post-dose starting from SAD 160 mg and MAD 50 mg b.i.d.) and TLR8-mediated TNF release after ex vivo stimulation (>90% inhibition at 24 h post-dose starting from SAD 320 mg and MAD 100 mg b.i.d.). CONCLUSION: The safety, pharmacokinetic and pharmacodynamic data support the further development of MHV370 in systemic autoimmune diseases driven by the overactivation of TLR7 and TLR8.


Asunto(s)
Receptor Toll-Like 7 , Receptor Toll-Like 8 , Humanos , Adulto , Animales , Ratones , Área Bajo la Curva , Ayuno , Administración Oral , Método Doble Ciego , Relación Dosis-Respuesta a Droga , Voluntarios Sanos
4.
RMD Open ; 9(2)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37321668

RESUMEN

OBJECTIVES: Despite several effective targeted therapies, biomarkers that predict whether a patient with psoriatic arthritis (PsA) will respond to a particular treatment are currently lacking. METHODS: We analysed proteomics data from serum samples of nearly 2000 patients with PsA in placebo-controlled phase-III clinical trials of the interleukin-17 inhibitor secukinumab. To discover predictive biomarkers of clinical response, we used statistical learning with controlled feature selection. The top candidate was validated using an ELISA and was separately assessed in a trial of almost 800 patients with PsA treated with secukinumab or the tumour necrosis factor inhibitor adalimumab. RESULTS: Serum levels of beta-defensin 2 (BD-2) at baseline were found to be robustly associated with subsequent clinical response (eg, American College of Rheumatology definition of 20%, 50% and 70% improvement) to secukinumab, but not to placebo. This finding was validated in two independent clinical studies not used for discovery. Although BD-2 is known to be associated with psoriasis severity, the predictivity of BD-2 was independent of baseline Psoriasis Area and Severity Index. The association between BD-2 and response to secukinumab was observed as early as 4 weeks and maintained up to 52 weeks. BD-2 was also found to predict response to treatment with adalimumab. Unlike in PsA, BD-2 was not predictive of response to secukinumab in rheumatoid arthritis. CONCLUSIONS: In PsA, BD-2 at baseline is quantitatively associated with clinical response to secukinumab. Patients with high levels of BD-2 at baseline reach and sustain higher rates of clinical response after treatment with secukinumab.


Asunto(s)
Artritis Psoriásica , Psoriasis , beta-Defensinas , Humanos , Artritis Psoriásica/diagnóstico , Artritis Psoriásica/tratamiento farmacológico , Adalimumab/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Interleucina-17 , beta-Defensinas/uso terapéutico , Proteómica , Resultado del Tratamiento , Psoriasis/tratamiento farmacológico , Biomarcadores
5.
Int J Toxicol ; 42(5): 379-385, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37226048

RESUMEN

Agents that act at the N-methyl-D-aspartate receptor (NMDAR), such as ketamine, have gained increasing attention as rapid-acting antidepressants; however, their use has been limited by potential neurotoxicity. Recent FDA guidance requires a demonstration of safety on histologic parameters prior to the initiation of human studies. D-cycloserine (DCS) is a partial NMDA agonist that, along with lurasidone, is being investigated as a treatment for depression. The current study was designed to investigate the neurologic safety profile of DCS. To this end, female Sprague Dawley rats (n = 106) were randomly divided into 8 study groups. Ketamine was administered via tail vein infusion. DCS and lurasidone were administered via oral gavage in escalating doses to a maximum of 2000 mg/kg DCS. To ascertain toxicity, dose escalation with 3 different doses of D-cycloserine/lurasidone was given in combination with ketamine. MK-801, a known neurotoxic NMDA antagonist, was administered as a positive control. Brain tissue was sectioned and stained with H&E, silver, and Fluoro-Jade B stains. No fatalities were observed in any group. No microscopic abnormalities were found in the brain of animal subjects given ketamine, ketamine followed by DCS/lurasidone, or DCS/lurasidone alone. Neuronal necrosis, as expected, was seen in the MK-801 (positive control) group. We conclude that NRX-101, a fixed-dose combination of DCS/lurasidone, when administered with or without prior infusion of IV ketamine was tolerated and did not induce neurotoxicity, even at supratherapeutic doses of DCS.


Asunto(s)
Ketamina , Humanos , Ratas , Animales , Femenino , Ketamina/toxicidad , Cicloserina/farmacología , Cicloserina/uso terapéutico , Clorhidrato de Lurasidona , Maleato de Dizocilpina/toxicidad , N-Metilaspartato , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/agonistas
6.
Cell Rep Med ; 4(5): 101036, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37196635

RESUMEN

Genetic and in vivo evidence suggests that aberrant recognition of RNA-containing autoantigens by Toll-like receptors (TLRs) 7 and 8 drives autoimmune diseases. Here we report on the preclinical characterization of MHV370, a selective oral TLR7/8 inhibitor. In vitro, MHV370 inhibits TLR7/8-dependent production of cytokines in human and mouse cells, notably interferon-α, a clinically validated driver of autoimmune diseases. Moreover, MHV370 abrogates B cell, plasmacytoid dendritic cell, monocyte, and neutrophil responses downstream of TLR7/8. In vivo, prophylactic or therapeutic administration of MHV370 blocks secretion of TLR7 responses, including cytokine secretion, B cell activation, and gene expression of, e.g., interferon-stimulated genes. In the NZB/W F1 mouse model of lupus, MHV370 halts disease. Unlike hydroxychloroquine, MHV370 potently blocks interferon responses triggered by specific immune complexes from systemic lupus erythematosus patient sera, suggesting differentiation from clinical standard of care. These data support advancement of MHV370 to an ongoing phase 2 clinical trial.


Asunto(s)
Enfermedades Autoinmunes , Lupus Eritematoso Sistémico , Humanos , Ratones , Animales , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 7/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/metabolismo , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Interferones
7.
Cell Rep Med ; 4(3): 100983, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36948149

RESUMEN

Lilja et al.1 explore single-cell transcriptomes across multiple organs of mice with collagen-induced arthritis. They apply network analysis to prioritize functional pathways that support or suppress inflammation and integrate findings with tissue transcriptomics in human immune-mediated inflammatory diseases.


Asunto(s)
Artritis Experimental , Inflamación , Animales , Humanos , Inflamación/metabolismo , Artritis Experimental/metabolismo
8.
JCI Insight ; 7(21)2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36345939

RESUMEN

Lupus nephritis is a serious complication of systemic lupus erythematosus, mediated by IgG immune complex (IC) deposition in kidneys, with limited treatment options. Kidney macrophages are critical tissue sentinels that express IgG-binding Fcγ receptors (FcγRs), with previous studies identifying prenatally seeded resident macrophages as major IC responders. Using single-cell transcriptomic and spatial analyses in murine and human lupus nephritis, we sought to understand macrophage heterogeneity and subset-specific contributions in disease. In lupus nephritis, the cell fate trajectories of tissue-resident (TrMac) and monocyte-derived (MoMac) kidney macrophages were perturbed, with disease-associated transcriptional states indicating distinct pathogenic roles for TrMac and MoMac subsets. Lupus nephritis-associated MoMac subsets showed marked induction of FcγR response genes, avidly internalized circulating ICs, and presented IC-opsonized antigen. In contrast, lupus nephritis-associated TrMac subsets demonstrated limited IC uptake, but expressed monocyte chemoattractants, and their depletion attenuated monocyte recruitment to the kidney. TrMacs also produced B cell tissue niche factors, suggesting a role in supporting autoantibody-producing lymphoid aggregates. Extensive similarities were observed with human kidney macrophages, revealing cross-species transcriptional disruption in lupus nephritis. Overall, our study suggests a division of labor in the kidney macrophage response in lupus nephritis, with treatment implications - TrMacs orchestrate leukocyte recruitment while MoMacs take up and present IC antigen.


Asunto(s)
Lupus Eritematoso Sistémico , Nefritis Lúpica , Ratones , Humanos , Animales , Macrófagos , Monocitos/patología , Receptores de IgG/genética , Inmunoglobulina G
9.
Cells ; 11(12)2022 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-35741037

RESUMEN

Signaling through the TNF-family receptor Fas/CD95 can trigger apoptosis or non-apoptotic cellular responses and is essential for protection from autoimmunity. Receptor clustering has been observed following interaction with Fas ligand (FasL), but the stoichiometry of Fas, particularly when triggered by membrane-bound FasL, the only form of FasL competent at inducing programmed cell death, is not known. Here we used super-resolution microscopy to study the behavior of single molecules of Fas/CD95 on the plasma membrane after interaction of Fas with FasL on planar lipid bilayers. We observed rapid formation of Fas protein superclusters containing more than 20 receptors after interactions with membrane-bound FasL. Fluorescence correlation imaging demonstrated recruitment of FADD dependent on an intact Fas death domain, with lipid raft association playing a secondary role. Flow-cytometric FRET analysis confirmed these results, and also showed that some Fas clustering can occur in the absence of FADD and caspase-8. Point mutations in the Fas death domain associated with autoimmune lymphoproliferative syndrome (ALPS) completely disrupted Fas reorganization and FADD recruitment, confirming structure-based predictions of the critical role that these residues play in Fas-Fas and Fas-FADD interactions. Finally, we showed that induction of apoptosis correlated with the ability to form superclusters and recruit FADD.


Asunto(s)
Apoptosis , Receptor fas , Apoptosis/fisiología , Análisis por Conglomerados , Proteína Ligando Fas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptor fas/metabolismo
10.
J Clin Invest ; 132(6)2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35289316

RESUMEN

Host defense and inflammation are regulated by the NF-κB essential modulator (NEMO), a scaffolding protein with a broad immune cell and tissue expression profile. Hypomorphic mutations in inhibitor of NF-κB kinase regulatory subunit gamma (IKBKG) encoding NEMO typically present with immunodeficiency. Here, we characterized a pediatric autoinflammatory syndrome in 3 unrelated male patients with distinct X-linked IKBKG germline mutations that led to overexpression of a NEMO protein isoform lacking the domain encoded by exon 5 (NEMO-Δex5). This isoform failed to associate with TANK binding kinase 1 (TBK1), and dermal fibroblasts from affected patients activated NF-κB in response to TNF but not TLR3 or RIG-I-like receptor (RLR) stimulation when isoform levels were high. By contrast, T cells, monocytes, and macrophages that expressed NEMO-Δex5 exhibited increased NF-κB activation and IFN production, and blood cells from these patients expressed a strong IFN and NF-κB transcriptional signature. Immune cells and TNF-stimulated dermal fibroblasts upregulated the inducible IKK protein (IKKi) that was stabilized by NEMO-Δex5, promoting type I IFN induction and antiviral responses. These data revealed how IKBKG mutations that lead to alternative splicing of skipping exon 5 cause a clinical phenotype we have named NEMO deleted exon 5 autoinflammatory syndrome (NDAS), distinct from the immune deficiency syndrome resulting from loss-of-function IKBKG mutations.


Asunto(s)
Enfermedades Autoinflamatorias Hereditarias , Síndromes de Inmunodeficiencia , Empalme Alternativo , Niño , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Síndromes de Inmunodeficiencia/genética , Masculino , FN-kappa B/genética , FN-kappa B/metabolismo , Fenotipo
11.
Cell Rep ; 37(6): 109977, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34758308

RESUMEN

Tumor necrosis factor (TNF) is a key driver of several inflammatory diseases, such as rheumatoid arthritis, inflammatory bowel disease, and psoriasis, in which affected tissues show an interferon-stimulated gene signature. Here, we demonstrate that TNF triggers a type-I interferon response that is dependent on the cyclic guanosine monophosphate-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. We show that TNF inhibits PINK1-mediated mitophagy and leads to altered mitochondrial function and to an increase in cytosolic mtDNA levels. Using cGAS-chromatin immunoprecipitation (ChIP), we demonstrate that cytosolic mtDNA binds to cGAS after TNF treatment. Furthermore, TNF induces a cGAS-STING-dependent transcriptional response that mimics that of macrophages from rheumatoid arthritis patients. Finally, in an inflammatory arthritis mouse model, cGAS deficiency blocked interferon responses and reduced inflammatory cell infiltration and joint swelling. These findings elucidate a molecular mechanism linking TNF to type-I interferon signaling and suggest a potential benefit for therapeutic targeting of cGAS/STING in TNF-driven diseases.


Asunto(s)
Artritis Experimental/inmunología , ADN Mitocondrial/metabolismo , Inmunidad Innata , Inflamación/inmunología , Interferón Tipo I/farmacología , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/genética , Artritis Experimental/metabolismo , ADN Mitocondrial/efectos de los fármacos , Femenino , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Macrófagos/inmunología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitofagia
12.
J Transl Med ; 19(1): 245, 2021 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-34090480

RESUMEN

In the era of precision medicine, digital technologies and artificial intelligence, drug discovery and development face unprecedented opportunities for product and business model innovation, fundamentally changing the traditional approach of how drugs are discovered, developed and marketed. Critical to this transformation is the adoption of new technologies in the drug development process, catalyzing the transition from serendipity-driven to data-driven medicine. This paradigm shift comes with a need for both translation and precision, leading to a modern Translational Precision Medicine approach to drug discovery and development. Key components of Translational Precision Medicine are multi-omics profiling, digital biomarkers, model-based data integration, artificial intelligence, biomarker-guided trial designs and patient-centric companion diagnostics. In this review, we summarize and critically discuss the potential and challenges of Translational Precision Medicine from a cross-industry perspective.


Asunto(s)
Inteligencia Artificial , Medicina de Precisión , Biomarcadores , Descubrimiento de Drogas , Humanos , Investigación Biomédica Traslacional
13.
Nat Commun ; 12(1): 2745, 2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-33980856

RESUMEN

In mice, time of day strongly influences lethality in response to LPS, with survival greatest at the beginning compared to the end of the light cycle. Here we show that feeding, rather than light, controls time-of-day dependent LPS sensitivity. Mortality following LPS administration is independent of cytokine production and the clock regulator BMAL1 expressed in myeloid cells. In contrast, deletion of BMAL1 in hepatocytes globally disrupts the transcriptional response to the feeding cycle in the liver and results in constitutively high LPS sensitivity. Using RNAseq and functional validation studies we identify hepatic farnesoid X receptor (FXR) signalling as a BMAL1 and feeding-dependent regulator of LPS susceptibility. These results show that hepatocyte-intrinsic BMAL1 and FXR signalling integrate nutritional cues to regulate survival in response to innate immune stimuli. Understanding hepatic molecular programmes operational in response to these cues could identify novel pathways for targeting to enhance endotoxemia resistance.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Conducta Alimentaria/fisiología , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Sepsis/mortalidad , Factores de Transcripción ARNTL/genética , Animales , Ritmo Circadiano/genética , Modelos Animales de Enfermedad , Resistencia a la Enfermedad , Hepatocitos/metabolismo , Hipoglucemia/metabolismo , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/toxicidad , Ratones , Ratones Noqueados , Receptores Citoplasmáticos y Nucleares/genética , Sepsis/inducido químicamente , Sepsis/genética , Sepsis/metabolismo , Transducción de Señal
14.
Rheumatology (Oxford) ; 61(1): 163-173, 2021 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-33744914

RESUMEN

OBJECTIVES: Arterial calcification due to deficiency of CD73 (ACDC) is a hereditary autosomal recessive ectopic mineralization syndrome caused by loss-of-function mutations in the ecto-5'-nucleotidase gene. Periarticular calcification has been reported but the clinical characterization of arthritis as well as the microstructure and chemical composition of periarticular calcifications and SF crystals has not been systematically investigated. METHODS: Eight ACDC patients underwent extensive rheumatological and radiological evaluation over a period of 11 years. Periarticular and synovial biopsies were obtained from four patients. Characterization of crystal composition was evaluated by compensated polarized light microscopy, Alizarin Red staining for synovial fluid along with X-ray diffraction and X-ray micro tomosynthesis scanner for periarticular calcification. RESULTS: Arthritis in ACDC patients has a clinical presentation of mixed erosive-degenerative joint changes with a median onset of articular symptoms at 17 years of age and progresses over time to the development of fixed deformities and functional limitations of small peripheral joints with, eventually, larger joint and distinct axial involvement later in life. We have identified calcium pyrophosphate and calcium hydroxyapatite (CHA) crystals in SF specimens and determined that CHA crystals are the principal component of periarticular calcifications. CONCLUSION: This is the largest study in ACDC patients to describe erosive peripheral arthropathy and axial enthesopathic calcifications over a period of 11 years and the first to identify the composition of periarticular calcifications and SF crystals. ACDC should be considered among the genetic causes of early-onset OA, as musculoskeletal disease signs may often precede vascular symptoms.


Asunto(s)
5'-Nucleotidasa/deficiencia , Calcinosis/diagnóstico por imagen , Artropatías/diagnóstico por imagen , Periartritis/diagnóstico por imagen , Enfermedades Vasculares/diagnóstico por imagen , 5'-Nucleotidasa/genética , Calcinosis/genética , Calcinosis/patología , Preescolar , Femenino , Proteínas Ligadas a GPI/genética , Humanos , Artropatías/genética , Artropatías/patología , Masculino , Persona de Mediana Edad , Periartritis/genética , Periartritis/patología , Radiografía , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología
16.
Clin Immunol ; 224: 108664, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33422677

RESUMEN

OBJECTIVE: Characterize autoantibodies and autoimmune diseases in a prospective cohort of patients with Idiopathic CD4 Lymphocytopenia (ICL) a rare immunodeficiency characterized by an absolute CD4+ T count of <300 cells/µl in the absence of HIV or HTLV infection. METHODS: Single-Center prospective study of 67 patients conducted over an 11-year period. Rheumatologic evaluation and measurement of autoantibodies were systematically conducted, and flow cytometry of immune cell subsets was performed in a subset of patients. RESULTS: 54% of referred patients had clinical evidence of autoimmunity, with 34% having at least one autoimmune disease, most commonly autoimmune thyroid disease. 19%, had autoantibodies or incomplete features of autoimmune disease. Patients with autoimmune disease had more elevated serum immunoglobulins, and more effector memory T cells than those without autoimmunity. CONCLUSIONS: Evidence of autoimmunity, including autoimmune diseases, is more prevalent in ICL than the general population, and should be considered part of this syndrome.


Asunto(s)
Autoanticuerpos/análisis , Enfermedades Autoinmunes/inmunología , Inmunofenotipificación/métodos , Linfocitopenia-T Idiopática CD4-Positiva/inmunología , Adulto , Anciano , Enfermedades Autoinmunes/complicaciones , Estudios de Cohortes , Enfermedades Transmisibles/complicaciones , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Linfocitopenia-T Idiopática CD4-Positiva/complicaciones , Adulto Joven
18.
Cancers (Basel) ; 12(9)2020 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-32825784

RESUMEN

Background: Patients with cholangiocarcinoma often have indwelling biliary stents or catheters which are prone to obstructions and/or infections; studies show that 20-40% present with fever and/or jaundice requiring urgent treatment in the outpatient setting for which there are no uniform guidelines. The goal was to develop an expert panel consensus on this topic using the modified RAND/UCLA Delphi process to rate treatment appropriateness. Methods: Thirteen expert physicians from relevant specialties, geography, and practice settings were recruited for the panel. Patient scenarios were developed and panelists rated the therapies before and after a face-to-face discussion. The appropriateness of various therapies was rated on a scale from 1-9 and classified as appropriate, inappropriate, or uncertain. Scenarios with greater than 2 (>2) ratings of 1-3 (inappropriate) and greater than 2 (>2) ratings of 7-9 (appropriate) were considered to have disagreement and were not assigned an appropriateness rating. Results: Panelists were from all US regions and the UK (8%) and had practiced for a mean 16.5 years (4-33 years). Panelists rated 480 scenarios before the meeting and re-rated 288 of the clinical scenarios after the meeting. The panelists agreed that ongoing treatment with chemotherapy did not influence decision-making and, therefore, 192 scenarios were excluded from the final list. Disagreement decreased from 37.5% before to 10.4% after the meeting. Consensus on stent/tube manipulation and inpatient antibiotic therapy was obtained and summarized in patients as "appropriate" or "maybe appropriate" based on a patient's bilirubin level at presentation. Conclusions: The Delphi process produced consensus guidelines to fill an unmet need in the urgent management of ascending cholangitis in patients with cholangiocarcinoma.

19.
J Clin Invest ; 130(10): 5326-5337, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32634122

RESUMEN

BACKGROUNDIdiopathic CD4 lymphopenia (ICL) is defined by persistently low CD4+ cell counts (<300 cells/µL) in the absence of a causal infection or immune deficiency and can manifest with opportunistic infections. Approximately 30% of ICL patients develop autoimmune disease. The prevalence and breadth of their autoantibodies, however, and their potential contribution to pathogenesis of ICL remain unclear.METHODSWe hybridized 34 and 51 ICL patients' sera to a 9,000-human-proteome array and to a 128-known-autoantigen array, respectively. Using a flow-based method, we characterized the presence of anti-lymphocyte Abs in the whole cohort of 72 patients, as well as the Ab functional capability of inducing Ab-dependent cell-mediated cytotoxicity (ADCC), complement deposition, and complement-dependent cytotoxicity (CDC). We tested ex vivo the activation of the classical complement pathway on ICL CD4+ T cells.RESULTSAll ICL patients had a multitude of autoantibodies mostly directed against private (not shared) targets and unrelated quantitatively or qualitatively to the patients' autoimmune disease status. The targets included lymphocyte intracellular and membrane antigens, confirmed by the detection by flow of IgM and IgG (mostly IgG1 and IgG4) anti-CD4+ cell Abs in 50% of the patients, with half of these cases triggering lysis of CD4+ T cells. We also detected in vivo classical complement activation on CD4+ T cells in 14% of the whole cohort.CONCLUSIONOur data demonstrate that a high prevalence of autoantibodies in ICL, some of which are specific for CD4+ T cells, may contribute to pathogenesis, and may represent a potentially novel therapeutic target.TRIAL REGISTRATIONClinicalTrials.gov NCT00867269.FUNDINGNIAID and National Institute of Arthritis and Musculoskeletal and Skin Diseases of the NIH.


Asunto(s)
Autoanticuerpos/sangre , Linfocitopenia-T Idiopática CD4-Positiva/inmunología , Adulto , Anciano , Especificidad de Anticuerpos , Citotoxicidad Celular Dependiente de Anticuerpos , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Estudios de Cohortes , Activación de Complemento , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Masculino , Persona de Mediana Edad , Linfocitopenia-T Idiopática CD4-Positiva/sangre , Linfocitopenia-T Idiopática CD4-Positiva/etiología , Adulto Joven
20.
Proc Natl Acad Sci U S A ; 117(26): 15160-15171, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32541026

RESUMEN

IgG antibodies cause inflammation and organ damage in autoimmune diseases such as systemic lupus erythematosus (SLE). We investigated the metabolic profile of macrophages isolated from inflamed tissues in immune complex (IC)-associated diseases, including SLE and rheumatoid arthritis, and following IgG Fcγ receptor cross-linking. We found that human and mouse macrophages undergo a switch to glycolysis in response to IgG IC stimulation, mirroring macrophage metabolic changes in inflamed tissue in vivo. This metabolic reprogramming was required to generate a number of proinflammatory mediators, including IL-1ß, and was dependent on mTOR and hypoxia-inducible factor (HIF)1α. Inhibition of glycolysis, or genetic depletion of HIF1α, attenuated IgG IC-induced activation of macrophages in vitro, including primary human kidney macrophages. In vivo, glycolysis inhibition led to a reduction in kidney macrophage IL-1ß and reduced neutrophil recruitment in a murine model of antibody-mediated nephritis. Together, our data reveal the molecular mechanisms underpinning FcγR-mediated metabolic reprogramming in macrophages and suggest a therapeutic strategy for autoantibody-induced inflammation, including lupus nephritis.


Asunto(s)
Reprogramación Celular/fisiología , Nefritis Lúpica/metabolismo , Animales , Células Cultivadas , Dinoprostona/genética , Dinoprostona/metabolismo , Metabolismo Energético , Regulación de la Expresión Génica , Glucólisis/fisiología , Humanos , Inmunoglobulina G/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Riñón/citología , Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especies Reactivas de Oxígeno , Receptores de IgG/genética , Receptores de IgG/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA