Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Exp Med ; 220(5)2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36884218

RESUMO

STAT6 (signal transducer and activator of transcription 6) is a transcription factor that plays a central role in the pathophysiology of allergic inflammation. We have identified 16 patients from 10 families spanning three continents with a profound phenotype of early-life onset allergic immune dysregulation, widespread treatment-resistant atopic dermatitis, hypereosinophilia with esosinophilic gastrointestinal disease, asthma, elevated serum IgE, IgE-mediated food allergies, and anaphylaxis. The cases were either sporadic (seven kindreds) or followed an autosomal dominant inheritance pattern (three kindreds). All patients carried monoallelic rare variants in STAT6 and functional studies established their gain-of-function (GOF) phenotype with sustained STAT6 phosphorylation, increased STAT6 target gene expression, and TH2 skewing. Precision treatment with the anti-IL-4Rα antibody, dupilumab, was highly effective improving both clinical manifestations and immunological biomarkers. This study identifies heterozygous GOF variants in STAT6 as a novel autosomal dominant allergic disorder. We anticipate that our discovery of multiple kindreds with germline STAT6 GOF variants will facilitate the recognition of more affected individuals and the full definition of this new primary atopic disorder.


Assuntos
Asma , Hipersensibilidade Alimentar , Humanos , Fator de Transcrição STAT6 , Mutação com Ganho de Função , Imunoglobulina E/genética
2.
Hematol Oncol Clin North Am ; 37(2): 301-312, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36907604

RESUMO

ß-thalassemia is caused by mutations that reduce ß-globin production, causing globin chain imbalance, ineffective erythropoiesis, and consequent anemia. Increased fetal hemoglobin (HbF) levels can ameliorate the severity of ß-thalassemia by compensating for the globin chain imbalance. Careful clinical observations paired with population studies and advances in human genetics have enabled the discovery of major regulators of HbF switching (i.e. BCL11A, ZBTB7A) and led to pharmacological and genetic therapies for treating ß-thalassemia patients. Recent functional screens using genome editing and other emerging tools have identified many new HbF regulators, which may improve therapeutic HbF induction in the future.


Assuntos
Hemoglobina Fetal , Talassemia beta , Humanos , Hemoglobina Fetal/genética , Talassemia beta/genética , Proteínas de Ligação a DNA , Linhagem Celular Tumoral , Fatores de Transcrição
3.
medRxiv ; 2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-36993312

RESUMO

Human genetic variation has enabled the identification of several key regulators of fetal-to-adult hemoglobin switching, including BCL11A, resulting in therapeutic advances. However, despite the progress made, limited further insights have been obtained to provide a fuller accounting of how genetic variation contributes to the global mechanisms of fetal hemoglobin (HbF) gene regulation. Here, we have conducted a multi-ancestry genome-wide association study of 28,279 individuals from several cohorts spanning 5 continents to define the architecture of human genetic variation impacting HbF. We have identified a total of 178 conditionally independent genome-wide significant or suggestive variants across 14 genomic windows. Importantly, these new data enable us to better define the mechanisms by which HbF switching occurs in vivo. We conduct targeted perturbations to define BACH2 as a new genetically-nominated regulator of hemoglobin switching. We define putative causal variants and underlying mechanisms at the well-studied BCL11A and HBS1L-MYB loci, illuminating the complex variant-driven regulation present at these loci. We additionally show how rare large-effect deletions in the HBB locus can interact with polygenic variation to influence HbF levels. Our study paves the way for the next generation of therapies to more effectively induce HbF in sickle cell disease and ß-thalassemia.

5.
Comput Struct Biotechnol J ; 20: 4717-4732, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36147669

RESUMO

We developed a bioinformatics-led substrate discovery workflow to expand the known substrate repertoire of MALT1. Our approach, termed GO-2-Substrates, integrates protein function information, including GO terms from known substrates, with protein sequences to rank substrate candidates by similarity. We applied GO-2-Substrates to MALT1, a paracaspase and master regulator of NF-κB signalling in adaptive immune responses. With only 12 known substrates, the evolutionarily conserved paracaspase functions and phenotypes of Malt1 -/- mice strongly implicate the existence of undiscovered substrates. We tested the ranked predictions from GO-2-Substrates of new MALT1 human substrates by co-expression of candidates transfected with the oncogenic constitutively active cIAP2-MALT1 fusion protein or CARD11/BCL10/MALT1 active signalosome. We identified seven new MALT1 substrates by the co-transfection screen: TANK, TAB3, CASP10, ZC3H12D, ZC3H12B, CILK1 and ILDR2. Using catalytically inactive cIAP2-MALT1 (Cys464Ala), a MALT1 inhibitor, MLT-748, and noncleavable P1-Arg to Ala mutant versions of each substrate in dual transfections, we validated the seven new substrates in vitro. We confirmed the cleavage of endogenous TANK and the RNase ZC3H12D in B cells by Western blotting and mining TAILS N-terminomics datasets, where we also uncovered evidence for these and 12 other candidate substrates by endogenous MALT1. Thus, protein function information improves substrate predictions. The new substrates and other high-ranked MALT1 candidate substrates should open new biological frontiers for further validation and exploration of the function of MALT1 within and beyond NF-κB regulation.

6.
Blood ; 140(17): 1858-1874, 2022 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-35789258

RESUMO

The discovery of humans with monogenic disorders has a rich history of generating new insights into biology. Here we report the first human identified with complete deficiency of nuclear factor of activated T cells 1 (NFAT1). NFAT1, encoded by NFATC2, mediates calcium-calcineurin signals that drive cell activation, proliferation, and survival. The patient is homozygous for a damaging germline NFATC2 variant (c.2023_2026delTACC; p.Tyr675Thrfs∗18) and presented with joint contractures, osteochondromas, and recurrent B-cell lymphoma. Absence of NFAT1 protein in chondrocytes caused enrichment in prosurvival and inflammatory genes. Systematic single-cell-omic analyses in PBMCs revealed an environment that promotes lymphomagenesis with accumulation of naïve B cells (enriched for oncogenic signatures MYC and JAK1), exhausted CD4+ T cells, impaired T follicular helper cells, and aberrant CD8+ T cells. This work highlights the pleiotropic role of human NFAT1, will empower the diagnosis of additional patients with NFAT1 deficiency, and further defines the detrimental effects associated with long-term use of calcineurin inhibitors.


Assuntos
Contratura , Leucemia de Células B , Osteocondroma , Humanos , Calcineurina/genética , Leucemia de Células B/genética , Leucemia de Células B/metabolismo , Recidiva Local de Neoplasia , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Linfoma de Células B/genética , Linfoma de Células B/metabolismo
7.
Front Immunol ; 12: 788278, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34887873

RESUMO

B-cell lymphoma/leukemia 11B (BCL11B) is a C2H2 zinc finger transcription factor that is critically important for regulating the development and function of a variety of systems including the central nervous system, the skin, and the immune system. Germline heterozygous variants are associated with a spectrum of clinical disorders, including severe combined immunodeficiency as well as neurological, craniofacial, and dermal defects. Of these individuals, ~50% present with severe allergic disease. Here, we report the detailed clinical and laboratory workup of one of the most severe BCL11B-dependent atopic cases to date. Leveraging a zebrafish model, we were able to confirm a strong T-cell defect in the patient. Based on these data, we classify germline BCL11B-dependent atopic disease as a novel primary atopic disorder.


Assuntos
Mutação em Linhagem Germinativa , Hipersensibilidade/genética , Doenças da Imunodeficiência Primária/genética , Proteínas Repressoras/genética , Linfócitos T/imunologia , Proteínas Supressoras de Tumor/genética , Adolescente , Animais , Análise Mutacional de DNA , Feminino , Predisposição Genética para Doença , Heterozigoto , Humanos , Hipersensibilidade/diagnóstico , Hipersensibilidade/imunologia , Fenótipo , Doenças da Imunodeficiência Primária/diagnóstico , Doenças da Imunodeficiência Primária/imunologia , Doenças da Imunodeficiência Primária/metabolismo , Proteínas Repressoras/metabolismo , Índice de Gravidade de Doença , Linfócitos T/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
8.
Front Immunol ; 12: 749794, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34721419

RESUMO

Nuclear factor kappa B (NF-κB) is a critical transcription factor involved in regulating cell activation, inflammation, and survival. The linear ubiquitin chain assembly complex (LUBAC) which consists of HOIL1, HOIP, and SHARPIN, catalyzes the linear ubiquitination of target proteins-a post-translational modification that is essential for NF-κB activation. Human germline pathogenic variants that dysregulate linear ubiquitination and NF-κB signaling are associated with immunodeficiency and/or autoinflammation including dermatitis, recurrent fevers, systemic inflammation and enteropathy. We previously identified MALT1 paracaspase as a novel negative regulator of LUBAC by proteolytic cleavage of HOIL1. To directly investigate the impact of HOIL1 cleavage activity on the inflammatory response, we employed a stable transduction system to express and directly compare non-cleavable HOIL1 with wild-type HOIL1 in primary HOIL1-deficient patient skin fibroblasts. We discovered that non-cleavable HOIL1 resulted in enhanced NF-κB signaling in response to innate stimuli. Transcriptomics revealed enrichment of inflammation and proinflammatory cytokine-related pathways after stimulation. Multiplexed cytokine assays confirmed a 'hyperinflammatory' phenotype in these cells. This work highlights the physiological importance of MALT1-dependent cleavage and modulation of HOIL1 on NF-κB signaling and inflammation, provides a mechanism for the autoinflammation observed in MALT1-deficient patients, and will inform the development of therapeutics that target MALT1 paracaspase and LUBAC function in treating autoinflammatory skin diseases.


Assuntos
Fibroblastos/imunologia , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/imunologia , NF-kappa B/imunologia , Fatores de Transcrição/imunologia , Ubiquitina-Proteína Ligases/imunologia , Citocinas/imunologia , Humanos , Inflamação/imunologia , Transdução de Sinais , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/genética
9.
J Allergy Clin Immunol ; 148(5): 1130-1139, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34428518

RESUMO

Inborn errors of immunity are traditionally best known for enhancing susceptibility to infections. However, allergic inflammation, among other types of immune dysregulation, occurs frequently in patients with inborn errors of immunity. As such, the term primary atopic disorders (PADs) was recently coined to describe the group of heritable monogenic allergic disorders. It is becoming increasingly important for clinicians to recognize that allergic diseases such as food allergy, atopic dermatitis, and allergic asthma are expressions of misdirected immunity, and in patients who present with severe, early-onset, or coexisting allergic conditions, these can be indications of an underlying PAD. Identifying monogenic allergic disease through next-generation sequencing can dramatically improve outcomes by allowing the use of precision-based therapy targeting the patient's underlying molecular defect. It is therefore imperative that clinicians recognize PADs to be able to provide informed therapeutic options and improve patient outcomes. Here, we summarize the clinical features commonly seen with each of the currently known PADs, identify clinical warning signs that warrant assessment for PADs, and lastly, discuss the benefits of timely diagnosis and management of these conditions.


Assuntos
Predisposição Genética para Doença , Hipersensibilidade Imediata/epidemiologia , Hipersensibilidade Imediata/etiologia , Imunidade/genética , Gerenciamento Clínico , Suscetibilidade a Doenças , Estudos de Associação Genética , Humanos , Hipersensibilidade Imediata/diagnóstico , Hipersensibilidade Imediata/terapia , Fenótipo
10.
J Allergy Clin Immunol ; 148(6): 1559-1574.e13, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33872653

RESUMO

BACKGROUND: Germline pathogenic variants impairing the caspase recruitment domain family member 11 (CARD11)-B cell chronic lymphocytic leukemia/lymphoma 10 (BCL10)-MALT1 paracaspase (MALT1) (CBM) complex are associated with diverse human diseases including combined immunodeficiency (CID), atopy, and lymphoproliferation. However, the impact of CARD11 deficiency on human B-cell development, signaling, and function is incompletely understood. OBJECTIVES: This study sought to determine the cellular, immunological, and biochemical basis of disease for 2 unrelated patients who presented with profound CID associated with viral and fungal respiratory infections, interstitial lung disease, and severe colitis. METHODS: Patients underwent next-generation sequencing, immunophenotyping by flow cytometry, signaling assays by immunoblot, and transcriptome profiling by RNA-sequencing. RESULTS: Both patients carried identical novel pathogenic biallelic loss-of-function variants in CARD11 (c.2509C>T; p.Arg837∗) leading to undetectable protein expression. This variant prevented CBM complex formation, severely impairing the activation of nuclear factor-κB, c-Jun N-terminal kinase, and MALT1 paracaspase activity in B and T cells. This functional defect resulted in a developmental block in B cells at the naive and type 1 transitional B-cell stage and impaired circulating T follicular helper cell (cTFH) development, which was associated with impaired antibody responses and absent germinal center structures on lymph node histology. Transcriptomics indicated that CARD11-dependent signaling is essential for immune signaling pathways involved in the development of these cells. Both patients underwent hematopoietic stem cell transplantations, which led to functional normalization. CONCLUSIONS: Complete human CARD11 deficiency causes profound CID by impairing naive/type 1 B-cell and cTFH cell development and abolishing activation of MALT1 paracaspase, NF-κB, and JNK activity. Hematopoietic stem cell transplantation functionally restores impaired signaling pathways.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/genética , Centro Germinativo/imunologia , Guanilato Ciclase/genética , Transplante de Células-Tronco Hematopoéticas , Mutação/genética , Células Precursoras de Linfócitos B/imunologia , Doenças da Imunodeficiência Primária/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Adolescente , Proteína 10 de Linfoma CCL de Células B/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Criança , Perfilação da Expressão Gênica , Guanilato Ciclase/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imunofenotipagem , Lactente , Masculino , NF-kappa B/metabolismo , Doenças da Imunodeficiência Primária/terapia , Transdução de Sinais
11.
Curr Opin Immunol ; 72: 1-12, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33714841

RESUMO

Human germline MALT1 deficiency is an inborn error of immunity characterized by recurrent bacterial, viral, and fungal infections, periodontal disease, enteropathy, dermatitis, and failure to thrive. The number of identified MALT1-deficient patients have greatly increased in the past two years, which has significantly improved our understanding of the clinical features of this disorder. Patients frequently experience infections affecting the respiratory, skin, gastrointestinal, and blood systems. The most frequently detected pathogens are Staphylococcus aureus, Candida albicans, and cytomegalovirus. Enhanced susceptibility to S. aureus and C. albicans is likely due to impaired Th17 immunity, similar to STAT3 and IL-17 pathway deficiencies.


Assuntos
Predisposição Genética para Doença , Infecções/etiologia , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/deficiência , Alelos , Animais , Citocinas/metabolismo , Genótipo , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Síndromes de Imunodeficiência/complicações , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Infecções/diagnóstico , Infecções/metabolismo , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/genética , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/metabolismo , Mutação , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
12.
BMC Pediatr ; 21(1): 45, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33472608

RESUMO

BACKGROUND: KRAS (KRAS proto-oncogene, GTPase; OMIM: 190,070) encodes one of three small guanosine triphosphatase proteins belonging to the RAS family. This group of proteins is responsible for cell proliferation, differentiation and inhibition of apoptosis. Gain-of-function variants in KRAS are commonly found in human cancers. Non-malignant somatic KRAS variants underlie a subset of RAS-associated autoimmune leukoproliferative disorders (RALD). RALD is characterized by splenomegaly, persistent monocytosis, hypergammaglobulinemia and cytopenia, but can also include autoimmune features and lymphadenopathy. In this report, we describe a non-malignant somatic variant in KRAS with prominent clinical features of massive splenomegaly, thrombocytopenia and lymphopenia. CASE PRESENTATION: A now-11-year-old girl presented in early childhood with easy bruising and bleeding, but had an otherwise unremarkable medical history. After consulting for the first time at 5 years of age, she was discovered to have massive splenomegaly. Clinical follow-up revealed thrombocytopenia, lymphopenia and increased polyclonal immunoglobulins and C-reactive protein. The patient had an unremarkable bone marrow biopsy, flow cytometry showed no indication of expanded double negative T-cells, while malignancy and storage disorders were also excluded. When the patient was 8 years old, whole exome sequencing performed on DNA derived from whole blood revealed a heterozygous gain-of-function variant in KRAS (NM_004985.5:c.37G > T; (p.G13C)). The variant was absent from DNA derived from a buccal swab and was thus determined to be somatic. CONCLUSIONS: This case of idiopathic splenomegaly in childhood due to a somatic variant in KRAS expands our understanding of the clinical spectrum of RAS-associated autoimmune leukoproliferative disorder and emphasizes the value of securing a molecular diagnosis in children with unusual early-onset presentations with a suspected monogenic origin.


Assuntos
Transtornos Linfoproliferativos , Esplenomegalia , Biópsia , Criança , Pré-Escolar , Feminino , Citometria de Fluxo , Humanos , Mutação , Proto-Oncogene Mas , Esplenomegalia/etiologia
13.
Artigo em Inglês | MEDLINE | ID: mdl-32532880

RESUMO

The innate immune system allows for rapid recognition of pathogens. Toll-like receptor (TLR) signaling is a key aspect of the innate immune response, and interleukin-1 receptor-associated kinase 4 (IRAK4) plays a vital role in the TLR signaling cascade. Each TLR recognizes a distinct set of pathogen-associated molecular patterns (PAMPs) that encompass conserved microbial components such as lipopolysaccharides and flagellin. Upon binding of PAMPs and TLR activation, TLR intracellular domains initiate the oligomerization of the myeloid differentiation primary response 88 (MyD88), IRAK1, IRAK2, and IRAK4 signaling platform known as the Myddosome complex while also triggering the Toll/IL-1R domain-containing adaptor-inducing IFN-ß (TRIF)-dependent pathway. The Myddosome complex initiates signal transduction pathways enabling the activation of NF-κB and mitogen-activated protein kinase (MAPK) transcription factors and the subsequent production of inflammatory cytokines. Human IRAK4 deficiency is an autosomal recessive inborn error of immunity that classically presents with blunted or delayed inflammatory response to infection and susceptibility to a narrow spectrum of pyogenic bacteria, particularly Streptococcus pneumoniae, Staphylococcus aureus, and Pseudomonas aeruginosa. We describe a case of IRAK4 deficiency in an 11-mo-old boy with concurrent S. pneumoniae bacteremia and S. aureus cervical lymphadenitis with a blunted inflammatory response to invasive infection. Although initial clinical immune profiling was unremarkable, a high degree of suspicion for an innate immune defect prompted genetic sequencing. Genetic testing revealed a novel variant in the IRAK4 gene (c.1049delG, p.(Gly350Glufs*15)) predicted to be likely pathogenic. Functional testing showed a loss of IRAK4 protein expression and abolished TLR signaling, confirming the pathogenicity of this novel IRAK4 variant.


Assuntos
Alelos , Substituição de Aminoácidos , Homozigoto , Quinases Associadas a Receptores de Interleucina-1/genética , Doenças da Imunodeficiência Primária/diagnóstico , Doenças da Imunodeficiência Primária/genética , Família , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Lactente , Masculino , Modelos Biológicos , Linhagem , Fenótipo
14.
Nat Cancer ; 1(12): 1167-1175, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-35121931

RESUMO

Human leukocyte antigen (HLA)-B has been recognized as a major determinant of discrepancies in disease outcomes, and recent evidence indicates a role in immune checkpoint blockade (ICB) efficacy. The B44 supertype, which features an electropositive binding pocket that preferentially displays peptides with negatively charged amino acid anchors, is associated with improved survival in ICB-treated melanoma. Yet this effect was not seen in ICB-treated non-small-cell lung cancer (NSCLC). Here we show that mutations leading to glutamic acid substitutions occur more often in melanoma than NSCLC based on mutational landscape. We additionally show stratifying B44 based on the presence of somatic mutations that lead to negatively charged glutamic acid anchors identifies patients with NSCLC with an ICB benefit similar to that seen in melanoma. We anticipate these findings could improve assessment of HLA-related outcomes and prediction of ICB benefit in those with B44, representing approximately half of the world's population.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Melanoma , Carcinoma Pulmonar de Células não Pequenas/genética , Ácido Glutâmico/genética , Antígenos HLA-B/genética , Antígeno HLA-B44/genética , Humanos , Imunoterapia , Neoplasias Pulmonares/genética , Melanoma/genética , Mutação
15.
J Allergy Clin Immunol ; 143(5): 1661-1673, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31060714

RESUMO

Caspase recruitment domain (CARD) protein-B cell CLL/lymphoma 10 (BCL10)-MALT1 paracaspase (MALT1) [CBM] complexes are critical signaling adaptors that facilitate immune and inflammatory responses downstream of both cell surface and intracellular receptors. Germline mutations that alter the function of members of this complex (termed CBM-opathies) cause a broad array of clinical phenotypes, ranging from profound combined immunodeficiency to B-cell lymphocytosis. With an increasing number of patients being described in recent years, the clinical spectrum of diseases associated with CBM-opathies is rapidly expanding and becoming unexpectedly heterogeneous. Here we review major discoveries that have shaped our understanding of CBM complex biology, and we provide an overview of the clinical presentation, diagnostic approach, and treatment options for those carrying germline mutations affecting CARD9, CARD11, CARD14, BCL10, and MALT1.


Assuntos
Linfócitos B/fisiologia , Hipersensibilidade Imediata/genética , Síndromes de Imunodeficiência/genética , Mutação/genética , Proteína 10 de Linfoma CCL de Células B/genética , Proteína 10 de Linfoma CCL de Células B/metabolismo , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Humanos , Hipersensibilidade Imediata/metabolismo , Síndromes de Imunodeficiência/metabolismo , Inflamação , Linfocitose , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/genética , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/metabolismo , Complexos Multiproteicos/metabolismo , Fenótipo , Transdução de Sinais
16.
Acta Biomater ; 85: 203-217, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30597258

RESUMO

Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a life-threatening condition of critically-ill patients, characterized by overwhelming inflammatory responses in the lung. Multiple lines of evidence suggest that the excessive activation of Toll-like receptor 4 (TLR4) plays an important role in this detrimental lung inflammation. Recently, we developed a unique class of peptide-gold nanoparticle (GNP) hybrids that act as potent nano-inhibitors of TLR4 signaling by modulating the process of endosomal acidification. In this study, we aimed to identify the key physiochemical factors that could further strengthen the anti-inflammatory activity of these nano-inhibitors, including the nanoparticle size, the density of peptides coating the nanoparticle surface, as well as the number of the effective amino acid phenylalanine (F) residues in the peptide sequence. Among these factors, we found that the nanoparticle size could significantly affect the TLR4 inhibition. Specifically, the peptide-GNP hybrids with a GNP core of 20 nm (P12(G20)) exhibited the most potent inhibitory activity on TLR4 activation and its downstream cytokine production among those with a GNP core of 13 nm (P12(G13)) and 5 nm (P12(G5)) in THP-1 cell-derived macrophages. This size-dependent anti-inflammatory effect of the hybrid P12 was also observed in a lipopolysaccharide (LPS)-induced mouse model of ALI. It was found that P12(G20) was superior to P12(G13) in prolonging the survival of mice experiencing lethal LPS challenge, decreasing the acute lung inflammation, and alleviating diffuse alveolar damage in the lungs. Interestingly, P12(G20) could also promote long-term tolerance to endotoxin. Detailed mechanistic studies demonstrated that when compared to the smaller P12(G13), the larger P12(G20) had higher cellular uptake and a stronger endosomal pH buffering capacity, contributing to its enhanced therapeutic effects on reducing TLR4 activation in vitro and in vivo. Overall, this study suggests that nanoparticle size is one key factor determining the anti-inflammatory potency of the peptide-GNP hybrids, and the hybrid P12 may serve as a promising, novel class of nanotherapeutics for modulating TLR signaling to treat ALI/ARDS. STATEMENT OF SIGNIFICANCE: We have developed a new class of nanoparticle-based inhibitors (i.e., peptide-GNP hybrids) targeting TLR4 signaling in macrophages. Through evidence-based engineering of the nanoparticle size, surface peptide ligand density and effective amino acid (phenylalanine, F) chain length, we identified a peptide-GNP hybrid, P12(G20), with enhanced anti-inflammatory activity. Specifically, P12(G20) was more potent in reducing inflammation in THP-1 cell-derived macrophages and in a LPS-induced ALI mouse model. More interestingly, P12(G20) facilitated long-term protection against lethal LPS challenge in vivo and induced endotoxin tolerance in vitro. We anticipate that these new hybrids would serve as the next generation anti-inflammatory nano-therapeutics for the treatment of ALI/ARDS or other acute and chronic inflammatory diseases.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/patologia , Anti-Inflamatórios/uso terapêutico , Ouro/química , Inflamação/patologia , Nanopartículas Metálicas/química , Tamanho da Partícula , Peptídeos/química , Animais , Anti-Inflamatórios/farmacologia , Modelos Animais de Doenças , Lipopolissacarídeos , Nanopartículas Metálicas/ultraestrutura , Fenilalanina/química
17.
Front Immunol ; 9: 2078, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30283440

RESUMO

The caspase recruitment domain family member 11 (CARD11 or CARMA1)-B cell CLL/lymphoma 10 (BCL10)-MALT1 paracaspase (MALT1) [CBM] signalosome complex serves as a molecular bridge between cell surface antigen receptor signaling and the activation of the NF-κB, JNK, and mTORC1 signaling axes. This positions the CBM complex as a critical regulator of lymphocyte activation, proliferation, survival, and metabolism. Inborn errors in each of the CBM components have now been linked to a diverse group of human primary immunodeficiency diseases termed "CBM-opathies." Clinical manifestations range from severe combined immunodeficiency to selective B cell lymphocytosis, atopic disease, and specific humoral defects. This surprisingly broad spectrum of phenotypes underscores the importance of "tuning" CBM signaling to preserve immune homeostasis. Here, we review the distinct clinical and immunological phenotypes associated with human CBM complex mutations and introduce new avenues for targeted therapeutic intervention.


Assuntos
Proteína 10 de Linfoma CCL de Células B/imunologia , Proteínas Adaptadoras de Sinalização CARD/imunologia , Guanilato Ciclase/imunologia , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/imunologia , Imunodeficiência Combinada Severa/imunologia , Transdução de Sinais/imunologia , Proteína 10 de Linfoma CCL de Células B/genética , Proteína 10 de Linfoma CCL de Células B/metabolismo , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Homeostase/genética , Homeostase/imunologia , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/genética , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/metabolismo , Mutação/imunologia , Ligação Proteica , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/metabolismo , Transdução de Sinais/genética
19.
Cell Death Dis ; 9(2): 162, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29415982

RESUMO

Proteasome inhibitors have emerged as an effective therapy for the treatment of haematological malignancies; however, their efficacy can be limited by the development of tumour resistance mechanisms. Novel combination strategies including the addition of TLR adjuvants to increase cell death and augment immune responses may help enhance their effectiveness. Although generally thought to inhibit inflammatory responses and NF-κB activation, we found that under specific conditions proteasome inhibitors can promote inflammatory responses by mediating IL-1ß maturation and secretion after TLR stimulation. This was dependent on the timing of proteasome inhibition relative to TLR stimulation where reversal of treatment order could alternatively increase or inhibit IL-1ß secretion (P < 0.001). TLR stimulation combined with proteasome inhibition enhanced cell death in vitro and delayed tumour development in vivo in NOD SCID mice (P < 0.01). However, unlike IL-1ß secretion, cell death occurred similarly regardless of treatment order and was only partially caspase dependent, possessing characteristics of both apoptosis and necrosis as indicated by activation of caspase-1, 3, 8 and RIP3 phosphorylation. Although stimulation of various TLRs was capable of driving IL-1ß production, TLR4 stimulation was the most effective at increasing cell death in THP-1 and U937 cells. TLR4 stimulation and proteasome inhibition independently activated the RIP3 necroptotic pathway and ultimately reduced the effectiveness of caspase/necroptosis inhibitors in mitigating overall levels of cell death. This strategy of combining TLR stimulation with proteasome inhibition may improve the ability of proteasome inhibitors to generate immunogenic cell death and increase anti-tumour activity.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Interleucina-1beta/biossíntese , Neoplasias/metabolismo , Neoplasias/patologia , Inibidores de Proteassoma/farmacologia , Receptores Toll-Like/agonistas , Animais , Bortezomib/farmacologia , Inibidores de Caspase/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células HEK293 , Humanos , Lipopolissacarídeos/farmacologia , Camundongos SCID , Necrose , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
20.
Sci Rep ; 7(1): 11379, 2017 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-28900238

RESUMO

Single-particle tracking (SPT) is a powerful method for exploring single-molecule dynamics in living cells with nanoscale spatiotemporal resolution. Photostability and bright fluorescence make quantum dots (Qdots) a popular choice for SPT. However, their large size could potentially alter the mobility of the molecule of interest. To test this, we labelled B cell receptors on the surface of B-lymphocytes with monovalent Fab fragments of antibodies that were either linked to Qdots via streptavidin or directly conjugated to the small organic fluorophore Cy3. Imaging of receptor mobility by total internal reflection fluorescence microscopy (TIRFM), followed by quantitative single-molecule diffusion and confinement analysis, definitively showed that Qdots sterically hinder lateral mobility regardless of the substrate to which the cells were adhered. Qdot labelling also drastically altered the frequency with which receptors transitioned between apparent slow- and fast-moving states and reduced the size of apparent confinement zones. Although we show that Qdot-labelled probes can detect large differences in receptor mobility, they fail to resolve subtle differences in lateral diffusion that are readily detectable using Cy3-labelled Fabs. Our findings highlight the utility and limitations of using Qdots for TIRFM and wide-field-based SPT, and have significant implications for interpreting SPT data.


Assuntos
Técnicas de Sonda Molecular , Sondas Moleculares , Pontos Quânticos , Receptores de Antígenos de Linfócitos B/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Transporte Proteico , Coloração e Rotulagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...