Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Sci Immunol ; 4(35)2019 05 31.
Article in English | MEDLINE | ID: mdl-31152090

ABSTRACT

Tissue residency is considered a defining feature of the innate lymphoid cell (ILC) populations located within mucosal and adipose tissues. ILCs are also present within all lymphoid tissues, but whether ILCs migrate between lymphoid and nonlymphoid sites and in what context is poorly understood. To determine whether migratory ILCs exist within peripheral lymph nodes (LNs), we labeled all cells within the brachial LN (bLN) of transgenic mice expressing a photoconvertible fluorescent protein by direct exposure to light. Tracking of cellular changes in the labeled LN revealed the gradual migration of new ILCs into the tissue, balanced by egress of ILCs dependent on sphingosine-1-phosphate receptors. Most of the migratory ILCs were ILC1s, entering LNs directly from the circulation in a CD62L- and CCR7-dependent manner and thus behaving like conventional natural killer (cNK) cells. Upon egress, both ILC1s and cNK cells were found to recirculate through peripheral LNs. A distinct population of migratory ILC2s were detected in the LN, but most of the ILC3s were tissue resident. Functionally, both migratory and resident ILC1s within LNs were able to rapidly produce IFN-γ to support the generation of robust TH1 T cell responses after immunization. Thus, migratory and resident ILC populations exist within peripheral LNs, with ILC1s, akin to cNK cells, able to traffic into these tissues where they can contribute to the initiation of adaptive immunity.


Subject(s)
Cell Movement/immunology , Immunity, Innate/immunology , Killer Cells, Natural/immunology , Lymph Nodes/cytology , Th1 Cells/immunology , Animals , Flow Cytometry , Interferon-gamma/metabolism , L-Selectin/metabolism , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CCR7/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Transcriptome
2.
Arthritis Rheumatol ; 70(12): 2087-2095, 2018 12.
Article in English | MEDLINE | ID: mdl-29956883

ABSTRACT

OBJECTIVE: B cells impact the progression of systemic sclerosis (SSc; scleroderma) through multiple pathogenic mechanisms. CD19 inhibition in mice reduced skin thickness, collagen production, and autoantibody levels, consistent with CD19 expression on plasma cells (PCs), the source of antibody production. PC depletion could effectively reduce collagen deposition and inflammation in SSc; therefore, we investigated the effects of PC depletion on SSc disease activity. METHODS: A PC gene signature was evaluated in SSc skin biopsy samples in 2 phase I clinical trials. We assessed microarray data from tissue from public studies of chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), dermatomyositis (DM), systemic lupus erythematosus (SLE), and atopic dermatitis, as well as blood from a phase IIb clinical trial in SLE. RESULTS: The PC signature was elevated in SSc skin specimens compared to healthy donor skin (P = 2.28 × 10-6 ) and correlated with the baseline modified Rodnan skin thickness score (MRSS) (r = 0.64, P = 0.0004). Patients with a high PC signature at baseline showed greater improvement in the MRSS (mean ± SD change 35 ± 16%; P = 6.30 × 10-4 ) following anti-CD19 treatment with inebilizumab (MEDI-551) than did patients with a low PC signature at baseline (mean ± SD change 8 ± 12%; P = 0.104). The PC signature was overexpressed in tissue from patients with SLE, DM, COPD, interstitial lung disease, and IPF relative to controls (all fold change >2; P < 0.001). The PC signature also differed significantly between SLE patients with mild-to-moderate disease and those with severe disease (SLE Disease Activity Index cutoff at 10) (fold change 1.44; P = 3.90 × 10-3 ) and correlated significantly with the degree of emphysema in COPD (r = 0.53, P = 7.55 × 10-8 ). CONCLUSION: Our results support the notion that PCs have a role in the pathogenesis of SSc and other autoimmune or pulmonary indications. An elevated pretreatment PC signature was associated with increased benefit from MEDI-551 in SSc.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Plasma Cells/metabolism , Scleroderma, Systemic/drug therapy , Severity of Illness Index , Adult , Biopsy , Double-Blind Method , Female , Humans , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/pathology , Male , Plasma Cells/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/pathology , Scleroderma, Systemic/pathology , Skin/pathology , Treatment Outcome
3.
Clin Cancer Res ; 24(16): 3857-3866, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29716923

ABSTRACT

Purpose: To identify a predictive biomarker for durvalumab, an anti-programmed death ligand 1 (PD-L1) mAb.Experimental Design: RNA sequencing of 97 advanced-stage non-small cell lung carcinoma (NSCLC) biopsies from a nonrandomized phase Ib/II clinical trial (1108/NCT01693562) were profiled to identify a predictive signature; 62 locally advanced or metastatic urothelial cancer tumors from the same study were profiled to confirm predictive utility of the signature. Thirty NSCLC patients provided pre- and posttreatment tumors for messenger RNA (mRNA) analysis. NSCLC with ≥25% tumor cells and urothelial cancer with ≥25% tumor or immune cells stained for PD-L1 at any intensity were scored PD-L1 positive (PD-L1+). Kaplan-Meier and Cox proportional hazards analyses were used to adjust for gender, age, prior therapies, histology, ECOG status, liver metastasis, and smoking. Tumor mutation burden (TMB) was calculated using data from The Cancer Genome Atlas (TCGA).Results: In the NSCLC discovery set, a four-gene IFNγ-positive (IFNγ+) signature comprising IFNγ, CD274, LAG3, and CXCL9 was associated with higher overall response rates, longer median progression-free survival, and overall survival compared with signature-low patients. IFNγ+-signature NSCLC patients had improved survival regardless of IHC PD-L1 status. These associations were replicated in a urothelial cancer cohort. The IFNγ+ signature was induced 2-fold (P = 0.003) by durvalumab after 8 weeks of therapy in patients with NSCLC, and baseline signature was associated with TMB but not survival in TCGA data.Conclusions: The IFNγ+ mRNA signature may assist in identifying patients with improved outcomes with durvalumab, independent of PD-L1 assessed by IHC. Clin Cancer Res; 24(16); 3857-66. ©2018 AACR.


Subject(s)
Antibodies, Monoclonal/administration & dosage , B7-H1 Antigen/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Interferon-gamma/genetics , Urologic Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , Biopsy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Chemokine CXCL9/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Middle Aged , Mutation , Neoplasm Metastasis , RNA, Messenger/genetics , Treatment Outcome , Urologic Neoplasms/genetics , Urologic Neoplasms/pathology , Urothelium/pathology , Lymphocyte Activation Gene 3 Protein
4.
Am J Pathol ; 185(11): 2949-68, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26348576

ABSTRACT

Remodeling of blood vessels and lymphatics are prominent features of sustained inflammation. Angiopoietin-2 (Ang2)/Tie2 receptor signaling and tumor necrosis factor-α (TNF)/TNF receptor signaling are known to contribute to these changes in airway inflammation after Mycoplasma pulmonis infection in mice. We determined whether Ang2 and TNF are both essential for the remodeling on blood vessels and lymphatics, and thereby influence the actions of one another. Their respective contributions to the initial stage of vascular remodeling and sprouting lymphangiogenesis were examined by comparing the effects of function-blocking antibodies to Ang2 or TNF, given individually or together during the first week after infection. As indices of efficacy, vascular enlargement, endothelial leakiness, venular marker expression, pericyte changes, and lymphatic vessel sprouting were assessed. Inhibition of Ang2 or TNF alone reduced the remodeling of blood vessels and lymphatics, but inhibition of both together completely prevented these changes. Genome-wide analysis of changes in gene expression revealed synergistic actions of the antibody combination over a broad range of genes and signaling pathways involved in inflammatory responses. These findings demonstrate that Ang2 and TNF are essential and synergistic drivers of remodeling of blood vessels and lymphatics during the initial stage of inflammation after infection. Inhibition of Ang2 and TNF together results in widespread suppression of the inflammatory response.


Subject(s)
Mycoplasma Infections/pathology , Mycoplasma pulmonis/physiology , Ribonuclease, Pancreatic/antagonists & inhibitors , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Female , Inflammation , Lymphangiogenesis , Lymphatic System/metabolism , Lymphatic System/pathology , Lymphatic Vessels/pathology , Mice , Mice, Inbred C57BL , Mycoplasma Infections/immunology , Oligonucleotide Array Sequence Analysis , Pericytes/pathology , Respiratory System/metabolism , Respiratory System/pathology , Ribonuclease, Pancreatic/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
J Clin Invest ; 124(12): 5337-51, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25384217

ABSTRACT

It has recently been demonstrated that memory B cells can reenter and reengage germinal center (GC) reactions, opening the possibility that multi-hit lymphomagenesis gradually occurs throughout life during successive immunological challenges. Here, we investigated this scenario in follicular lymphoma (FL), an indolent GC-derived malignancy. We developed a mouse model that recapitulates the FL hallmark t(14;18) translocation, which results in constitutive activation of antiapoptotic protein B cell lymphoma 2 (BCL2) in a subset of B cells, and applied a combination of molecular and immunofluorescence approaches to track normal and t(14;18)(+) memory B cells in human and BCL2-overexpressing B cells in murine lymphoid tissues. BCL2-overexpressing B cells required multiple GC transits before acquiring FL-associated developmental arrest and presenting as GC B cells with constitutive activation-induced cytidine deaminase (AID) mutator activity. Moreover, multiple reentries into the GC were necessary for the progression to advanced precursor stages of FL. Together, our results demonstrate that protracted subversion of immune dynamics contributes to early dissemination and progression of t(14;18)(+) precursors and shapes the systemic presentation of FL patients.


Subject(s)
B-Lymphocyte Subsets/metabolism , Cell Movement , Gene Expression Regulation, Neoplastic , Lymphoma, Follicular/metabolism , Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Animals , B-Lymphocyte Subsets/pathology , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Female , Humans , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Male , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-bcl-2/genetics
6.
Arthritis Rheumatol ; 66(4): 1022-33, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24757153

ABSTRACT

OBJECTIVE: The molecular basis of inflammatory myopathies such as dermatomyositis (DM), polymyositis, and inclusion body myositis, which share the characteristics of chronic muscle inflammation and skeletal muscle wasting, are poorly understood. As such, effective targeted treatments for these diseases are lacking, resulting in critical unmet medical needs for these devastating diseases. The purpose of this study was to identify possible new targets for drug development by exploring the mechanism by which inflammation may play a role in the pathology of the inflammatory myopathies. METHODS: We compared expression levels of inflammatory cytokines and microRNAs (miRNAs) between muscle biopsy samples from patients with inflammatory myopathies and those from donors without myositis. In vitro human and mouse model systems were then used to characterize the role of these cytokines and microRNAs on myoblast-to-myocyte differentiation. RESULTS: We observed increased expression of inflammatory cytokines, including tumor necrosis factor α (TNFα), interferon-α (IFNα), IFNß, and interleukin-1ß, in different subtypes of inflammatory myopathies. We observed decreased expression of microRNA-1 (miR-1), miR-133a, and miR-133b in all of the inflammatory myopathy subtypes we evaluated, as well as decreased expression of miR-206 in DM; these miRNAs are essential for adult skeletal muscle differentiation and maintenance. TNFα was significantly inversely correlated with decreased myogenic miRNA expression in the inflammatory myopathy subtypes. In mechanistic studies, TNFα inhibited the expression of myogenic miRNAs and suppressed the differentiation of C2C12 myoblasts to myocytes/myotubes in an NF-κB-dependent manner. This block in differentiation by TNFα was relieved by overexpression of miR-1, miR-206, or miR-133a/b. CONCLUSION: Taken together, these results provide a new mechanistic link between the action of proinflammatory cytokines and the degenerative pathology of inflammatory myopathies, and suggest therapeutic approaches for these diseases.


Subject(s)
Cytokines/metabolism , MicroRNAs/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Myositis/metabolism , Adult , Animals , Cell Differentiation/physiology , Cell Line , Disease Models, Animal , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , MicroRNAs/genetics , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Myositis/genetics , Myositis/pathology , NF-kappa B/metabolism
7.
Arthritis Rheumatol ; 66(1): 173-84, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24431284

ABSTRACT

OBJECTIVE: Production of pathogenic autoantibodies by self-reactive plasma cells (PCs) is a hallmark of autoimmune diseases. We undertook this study to investigate the prevalence of PCs and their relationship to known pathogenic pathways to increase our understanding of the role of PCs in disease progression and treatment response. METHODS: We developed a sensitive gene expression-based method to overcome the challenges of measuring PCs using flow cytometry. Whole-genome microarray analysis of sorted cellular fractions identified a panel of genes, IGHA1, IGJ, IGKC, IGKV4-1, and TNFRSF17, expressed predominantly in PCs. The sensitivity of the PC signature score created from the combined expression levels of these genes was assessed through ex vivo experiments with sorted cells. This PC gene expression signature was used for monitoring changes in PC levels following anti-CD19 therapy, for evaluating the relationship between PCs and other autoimmune disease-related genes, and for estimating PC levels in affected blood and tissue from patients with multiple autoimmune diseases. RESULTS: The PC signature was highly sensitive and capable of detecting a change in as few as 360 PCs. The PC signature was reduced more than 90% in scleroderma patients following anti-CD19 treatment, and this reduction was highly correlated (r = 0.80) with inhibition of collagen gene expression. Evaluation of multiple autoimmune diseases revealed that 30-35% of lupus and rheumatoid arthritis patients had increased levels of PCs. CONCLUSION: This newly developed PC signature provides a robust and accurate method of measuring PC levels in the clinic. Our results highlight subsets of patients across multiple autoimmune diseases who may benefit from PC-depleting therapy.


Subject(s)
Plasma Cells/metabolism , Scleroderma, Systemic/genetics , Transcriptome/genetics , Autoantibodies/biosynthesis , Autoimmune Diseases/genetics , B-Cell Maturation Antigen/genetics , Humans , Immunoglobulin J-Chains/genetics , Immunoglobulin kappa-Chains/genetics , Immunoglobulins/genetics , Tissue Array Analysis
8.
Arthritis Rheum ; 60(6): 1785-96, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19479852

ABSTRACT

OBJECTIVE: Type I interferons (IFNs) play an important role in the pathogenesis of systemic lupus erythematosus (SLE). This phase Ia trial was undertaken to evaluate the safety, pharmacokinetics, and immunogenicity of anti-IFNalpha monoclonal antibody (mAb) therapy in SLE. During the trial, we also examined whether overexpression of an IFNalpha/beta-inducible gene signature in whole blood could serve as a pharmacodynamic biomarker to evaluate IFNalpha neutralization and investigated downstream effects of neutralizing IFNalpha on BAFF and other key signaling pathways, i.e., granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-10 (IL-10), tumor necrosis factor alpha (TNFalpha), and IL-1beta, in SLE. METHODS: Affymetrix Human Genome U133 Plus 2.0 microarrays were used to profile whole blood and lesional skin of patients receiving standard therapy for mild to moderate SLE. Selected IFNalpha/beta-inducible proteins were analyzed by immunohistochemistry. RESULTS: With the study treatment, we observed anti-IFNalpha mAb-specific and dose-dependent inhibition of overexpression of IFNalpha/beta-inducible genes in whole blood and skin lesions from SLE patients, at both the transcript and the protein levels. In SLE patients with overexpression of messenger RNA for BAFF, TNFalpha, IL-10, IL-1beta, GM-CSF, and their respective inducible gene signatures in whole blood and/or skin lesions, we observed a general trend toward suppression of the expression of these genes and/or gene signatures upon treatment with anti-IFNalpha mAb. CONCLUSION: IFNalpha/beta-inducible gene signatures in whole blood are effective pharmacodynamic biomarkers to evaluate anti-IFNalpha mAb therapy in SLE. Anti-IFNalpha mAb can neutralize overexpression of IFNalpha/beta-inducible genes in whole blood and lesional skin from SLE patients and has profound effects on signaling pathways that may be downstream of IFNalpha in SLE.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Gene Expression Regulation/immunology , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , Biomarkers/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Female , Gene Expression Regulation/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interferon-alpha/metabolism , Interferon-beta/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lupus Erythematosus, Systemic/metabolism , Male , Middle Aged , Signal Transduction/drug effects , Signal Transduction/genetics , Skin/metabolism , Skin/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL