Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nat Immunol ; 20(7): 928-942, 2019 07.
Article in English | MEDLINE | ID: mdl-31061532

ABSTRACT

To define the cell populations that drive joint inflammation in rheumatoid arthritis (RA), we applied single-cell RNA sequencing (scRNA-seq), mass cytometry, bulk RNA sequencing (RNA-seq) and flow cytometry to T cells, B cells, monocytes, and fibroblasts from 51 samples of synovial tissue from patients with RA or osteoarthritis (OA). Utilizing an integrated strategy based on canonical correlation analysis of 5,265 scRNA-seq profiles, we identified 18 unique cell populations. Combining mass cytometry and transcriptomics revealed cell states expanded in RA synovia: THY1(CD90)+HLA-DRAhi sublining fibroblasts, IL1B+ pro-inflammatory monocytes, ITGAX+TBX21+ autoimmune-associated B cells and PDCD1+ peripheral helper T (TPH) cells and follicular helper T (TFH) cells. We defined distinct subsets of CD8+ T cells characterized by GZMK+, GZMB+, and GNLY+ phenotypes. We mapped inflammatory mediators to their source cell populations; for example, we attributed IL6 expression to THY1+HLA-DRAhi fibroblasts and IL1B production to pro-inflammatory monocytes. These populations are potentially key mediators of RA pathogenesis.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Gene Expression Profiling , Synovial Membrane/metabolism , Transcriptome , Arthritis, Rheumatoid/pathology , Autoimmunity/genetics , Biomarkers , Computational Biology/methods , Cross-Sectional Studies , Cytokines/metabolism , Fibroblasts/metabolism , Flow Cytometry , Gene Expression , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Leukocytes/immunology , Leukocytes/metabolism , Monocytes/immunology , Monocytes/metabolism , Signal Transduction , Single-Cell Analysis/methods , Synovial Membrane/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Workflow
2.
Sci Transl Med ; 11(491)2019 05 08.
Article in English | MEDLINE | ID: mdl-31068444

ABSTRACT

Macrophages tailor their function according to the signals found in tissue microenvironments, assuming a wide spectrum of phenotypes. A detailed understanding of macrophage phenotypes in human tissues is limited. Using single-cell RNA sequencing, we defined distinct macrophage subsets in the joints of patients with the autoimmune disease rheumatoid arthritis (RA), which affects ~1% of the population. The subset we refer to as HBEGF+ inflammatory macrophages is enriched in RA tissues and is shaped by resident fibroblasts and the cytokine tumor necrosis factor (TNF). These macrophages promoted fibroblast invasiveness in an epidermal growth factor receptor-dependent manner, indicating that intercellular cross-talk in this inflamed setting reshapes both cell types and contributes to fibroblast-mediated joint destruction. In an ex vivo synovial tissue assay, most medications used to treat RA patients targeted HBEGF+ inflammatory macrophages; however, in some cases, medication redirected them into a state that is not expected to resolve inflammation. These data highlight how advances in our understanding of chronically inflamed human tissues and the effects of medications therein can be achieved by studies on local macrophage phenotypes and intercellular interactions.


Subject(s)
Arthritis, Rheumatoid/pathology , Fibroblasts/pathology , Heparin-binding EGF-like Growth Factor/metabolism , Macrophages/pathology , Cell Polarity , Cell Shape , Humans , Inflammation/pathology , Joints/pathology , Single-Cell Analysis , Synovial Membrane/pathology
3.
Arthritis Rheumatol ; 71(7): 1034-1041, 2019 07.
Article in English | MEDLINE | ID: mdl-30835943

ABSTRACT

OBJECTIVE: Patients with rheumatoid arthritis (RA) in clinical remission may have subclinical synovial inflammation. This study was undertaken to determine the proportion of patients with RA in remission or with low disease activity at the time of arthroplasty who had histologic or transcriptional evidence of synovitis, and to identify clinical features that distinguished patients as having subclinical synovitis. METHODS: We compared Disease Activity Score in 28 joints (DAS28) to synovial histologic features in 135 patients with RA undergoing arthroplasty. We also compared DAS28 scores to RNA-Seq data in a subset of 35 patients. RESULTS: Fourteen percent of patients met DAS28 criteria for clinical remission (DAS28 <2.6), and another 15% met criteria for low disease activity (DAS28 <3.2). Histologic analysis of synovium revealed synovitis in 27% and 31% of samples from patients in remission and patients with low disease activity, respectively. Patients with low disease activity and synovitis also exhibited increased C-reactive protein (CRP) (P = 0.0006) and increased anti-cyclic citrullinated peptide (anti-CCP) antibody levels (P = 0.03) compared to patients without synovitis. Compared to patients with a "low inflammatory synovium" subtype, 183 genes were differentially expressed in the synovium of patients with subclinical synovitis. The majority of these genes (86%) were also differentially expressed in the synovium of patients with clinically active disease (DAS28 ≥3.2). CONCLUSION: Thirty-one percent of patients with low clinical disease activity exhibited histologic evidence of subclinical synovitis, which was associated with increased CRP and anti-CCP levels. Our findings suggest that synovial gene expression signatures of clinical synovitis are present in patients with subclinical synovitis.


Subject(s)
Arthritis, Rheumatoid/pathology , Synovial Membrane/pathology , Synovitis/pathology , Aged , Anti-Citrullinated Protein Antibodies/immunology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/surgery , Arthroplasty, Replacement, Hip , Arthroplasty, Replacement, Knee , Asymptomatic Diseases , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Remission Induction , Sequence Analysis, RNA , Synovial Membrane/metabolism , Synovitis/genetics
4.
Arthritis Res Ther ; 20(1): 139, 2018 07 11.
Article in English | MEDLINE | ID: mdl-29996944

ABSTRACT

BACKGROUND: Detailed molecular analyses of cells from rheumatoid arthritis (RA) synovium hold promise in identifying cellular phenotypes that drive tissue pathology and joint damage. The Accelerating Medicines Partnership RA/SLE Network aims to deconstruct autoimmune pathology by examining cells within target tissues through multiple high-dimensional assays. Robust standardized protocols need to be developed before cellular phenotypes at a single cell level can be effectively compared across patient samples. METHODS: Multiple clinical sites collected cryopreserved synovial tissue fragments from arthroplasty and synovial biopsy in a 10% DMSO solution. Mechanical and enzymatic dissociation parameters were optimized for viable cell extraction and surface protein preservation for cell sorting and mass cytometry, as well as for reproducibility in RNA sequencing (RNA-seq). Cryopreserved synovial samples were collectively analyzed at a central processing site by a custom-designed and validated 35-marker mass cytometry panel. In parallel, each sample was flow sorted into fibroblast, T-cell, B-cell, and macrophage suspensions for bulk population RNA-seq and plate-based single-cell CEL-Seq2 RNA-seq. RESULTS: Upon dissociation, cryopreserved synovial tissue fragments yielded a high frequency of viable cells, comparable to samples undergoing immediate processing. Optimization of synovial tissue dissociation across six clinical collection sites with ~ 30 arthroplasty and ~ 20 biopsy samples yielded a consensus digestion protocol using 100 µg/ml of Liberase™ TL enzyme preparation. This protocol yielded immune and stromal cell lineages with preserved surface markers and minimized variability across replicate RNA-seq transcriptomes. Mass cytometry analysis of cells from cryopreserved synovium distinguished diverse fibroblast phenotypes, distinct populations of memory B cells and antibody-secreting cells, and multiple CD4+ and CD8+ T-cell activation states. Bulk RNA-seq of sorted cell populations demonstrated robust separation of synovial lymphocytes, fibroblasts, and macrophages. Single-cell RNA-seq produced transcriptomes of over 1000 genes/cell, including transcripts encoding characteristic lineage markers identified. CONCLUSIONS: We have established a robust protocol to acquire viable cells from cryopreserved synovial tissue with intact transcriptomes and cell surface phenotypes. A centralized pipeline to generate multiple high-dimensional analyses of synovial tissue samples collected across a collaborative network was developed. Integrated analysis of such datasets from large patient cohorts may help define molecular heterogeneity within RA pathology and identify new therapeutic targets and biomarkers.


Subject(s)
Arthritis, Rheumatoid/pathology , Flow Cytometry/methods , High-Throughput Screening Assays/methods , Synovial Membrane/pathology , Cryopreservation , Humans
5.
Nat Commun ; 9(1): 791, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29476078

ABSTRACT

Droplet-based single-cell RNA-seq has emerged as a powerful technique for massively parallel cellular profiling. While this approach offers the exciting promise to deconvolute cellular heterogeneity in diseased tissues, the lack of cost-effective and user-friendly instrumentation has hindered widespread adoption of droplet microfluidic techniques. To address this, we developed a 3D-printed, low-cost droplet microfluidic control instrument and deploy it in a clinical environment to perform single-cell transcriptome profiling of disaggregated synovial tissue from five rheumatoid arthritis patients. We sequence 20,387 single cells revealing 13 transcriptomically distinct clusters. These encompass an unsupervised draft atlas of the autoimmune infiltrate that contribute to disease biology. Additionally, we identify previously uncharacterized fibroblast subpopulations and discern their spatial location within the synovium. We envision that this instrument will have broad utility in both research and clinical settings, enabling low-cost and routine application of microfluidic techniques.


Subject(s)
Arthritis, Rheumatoid/genetics , Microfluidics/methods , RNA/genetics , Single-Cell Analysis/methods , Arthritis, Rheumatoid/metabolism , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Microfluidics/economics , Microfluidics/instrumentation , RNA/metabolism , Single-Cell Analysis/economics , Single-Cell Analysis/instrumentation , Synovial Membrane/cytology , Synovial Membrane/metabolism
6.
Ann Rheum Dis ; 76(4): 740-747, 2017 04.
Article in English | MEDLINE | ID: mdl-28283529

ABSTRACT

OBJECTIVES: Deregulated production of interleukin (IL)-17 and IL-21 contributes to the pathogenesis of autoimmune disorders such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Production of IL-17 and IL-21 can be regulated by ROCK2, one of the two Rho kinases. Increased ROCK activation was previously observed in an SLE cohort. Here, we evaluated ROCK activity in a new SLE cohort, and an RA cohort, and assessed the ability of distinct inhibitors of the ROCK pathway to suppress production of IL-17 and IL-21 by SLE T cells or human Th17 cells. METHODS: ROCK activity in peripheral blood mononuclear cells (PBMCs) from 29 patients with SLE, 31 patients with RA and 28 healthy controls was determined by ELISA. SLE T cells or in vitro-differentiated Th17 cells were treated with Y27632 (a pan-ROCK inhibitor), KD025 (a selective ROCK2 inhibitor) or simvastatin (which inhibits RhoA, a major ROCK activator). ROCK activity and IL-17 and IL-21 production were assessed. The transcriptional profile altered by ROCK inhibitors was evaluated by NanoString technology. RESULTS: ROCK activity levels were significantly higher in patients with SLE and RA than healthy controls. Th17 cells exhibited high ROCK activity that was inhibited by Y27632, KD025 or simvastatin; each also decreased IL-17 and IL-21 production by purified SLE T cells or Th17 cells. Immune profiling revealed both overlapping and distinct effects of the different ROCK inhibitors. CONCLUSIONS: ROCK activity is elevated in PBMCs from patients with SLE and RA. Production of IL-17 and IL-21 by SLE T cells or Th17 cells can furthermore be inhibited by targeting the RhoA-ROCK pathway via both non-selective and selective approaches.


Subject(s)
Arthritis, Rheumatoid/blood , Interleukin-17/metabolism , Interleukins/metabolism , Lupus Erythematosus, Systemic/blood , T-Lymphocytes/metabolism , Th17 Cells/metabolism , Adult , Aged , Amides/pharmacology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Case-Control Studies , Cells, Cultured , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Male , Middle Aged , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Signal Transduction , Simvastatin/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , Th17 Cells/drug effects , Th17 Cells/enzymology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
7.
Annu Rev Med ; 67: 355-74, 2016.
Article in English | MEDLINE | ID: mdl-26768244

ABSTRACT

The Rho kinases, or ROCKs, are a family of serine-threonine kinases that serve as key downstream effectors for Rho GTPases. The ROCKs are increasingly recognized as critical coordinators of a tissue response to injury due to their ability to modulate a wide range of biological processes. Dysregulated ROCK activity has been implicated in several human pathophysiological conditions ranging from cardiovascular and renal disorders to fibrotic diseases. In recent years, an important role for the ROCKs in the regulation of immune responses is also being uncovered. We provide an overview of the role of the ROCKs in immune cells and discuss studies that highlight the emerging involvement of this family of kinases in the pathogenesis of autoimmune diseases. Given the potential promise of the ROCKs as therapeutic targets, we also outline the approaches that could be employed to inhibit the ROCKs in autoimmune disorders.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , T-Lymphocytes/immunology , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/immunology , rho-Associated Kinases/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Amides/therapeutic use , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , B-Lymphocytes/immunology , Cell Movement , Cell Proliferation , Cell Survival , Cytoskeleton/metabolism , Gene Expression , Giant Cell Arteritis/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Myeloid Cells/immunology , Osteoarthritis/metabolism , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/immunology , Scleroderma, Systemic/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/drug effects , rho-Associated Kinases/genetics
8.
Immunity ; 42(4): 719-30, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25902483

ABSTRACT

Within secondary lymphoid tissues, stromal reticular cells support lymphocyte function, and targeting reticular cells is a potential strategy for controlling pathogenic lymphocytes in disease. However, the mechanisms that regulate reticular cell function are not well understood. Here we found that during an immune response in lymph nodes, dendritic cells (DCs) maintain reticular cell survival in multiple compartments. DC-derived lymphotoxin beta receptor (LTßR) ligands were critical mediators, and LTßR signaling on reticular cells mediated cell survival by modulating podoplanin (PDPN). PDPN modulated integrin-mediated cell adhesion, which maintained cell survival. This DC-stromal axis maintained lymphocyte survival and the ongoing immune response. Our findings provide insight into the functions of DCs, LTßR, and PDPN and delineate a DC-stromal axis that can potentially be targeted in autoimmune or lymphoproliferative diseases.


Subject(s)
Dendritic Cells/cytology , Lymph Nodes/cytology , Lymphotoxin beta Receptor/immunology , Membrane Glycoproteins/immunology , Stromal Cells/cytology , Animals , Cell Adhesion , Cell Survival/immunology , Dendritic Cells/immunology , Gene Expression Regulation , Immunophenotyping , Lymph Nodes/immunology , Lymphocyte Depletion , Lymphotoxin beta Receptor/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Stromal Cells/immunology
9.
Nat Med ; 18(2): 260-6, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22245779

ABSTRACT

Helminths induce potent T helper 2 (TH2)-type immune responses that can mediate worm expulsion, but the role of this response in controlling the acute tissue damage caused by migrating multicellular parasites through vital tissues remains uncertain. We used a helminth infection model in which parasitic nematode larvae migrate transiently through the lung, resulting in hemorrhage and inflammation. We found that IL-17 initially contributed to inflammation and lung damage, whereas subsequent IL-4 receptor (IL-4R) signaling reduced elevations in IL-17 mRNA levels, enhanced the expression of insulin-like growth factor 1 (IGF-1) and IL-10 and stimulated the development of M2 macrophages, all of which contributed to the rapid resolution of tissue damage. These studies indicate an essential role for TH2-type immune responses in mediating acute wound healing during helminth infection.


Subject(s)
Helminthiasis, Animal/immunology , Th2 Cells/physiology , Animals , Female , Hemorrhage/immunology , Hemorrhage/parasitology , Immunity, Cellular , Inflammation/immunology , Inflammation/parasitology , Insulin-Like Growth Factor I/physiology , Interleukin-17/physiology , Interleukin-4/physiology , Lung/blood supply , Lung/immunology , Lung/parasitology , Lung Diseases, Parasitic/immunology , Lung Diseases, Parasitic/parasitology , Macrophages/immunology , Macrophages/physiology , Mice , Mice, Inbred BALB C , Neutrophil Infiltration/immunology , Neutrophils/physiology , Nippostrongylus/immunology , Strongylida Infections/immunology
10.
J Immunol ; 187(12): 6491-8, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22095717

ABSTRACT

Wear debris in joint replacements has been suggested as a cause of associated tissue-damaging inflammation. In this study, we examined whether solid titanium microparticles (mTi) of sufficient size to accumulate as wear debris could stimulate innate or adaptive immunity in vivo. mTi, administered in conjunction with OVA, promoted total and Ag-specific elevations in serum IgE and IgG1. Analysis of transferred transgenic OVA-specific naive T cells further showed that mTi acted as an adjuvant to drive Ag-specific Th2 cell differentiation in vivo. Assessment of the innate response indicated that mTi induced rapid recruitment and differentiation of alternatively activated macrophages in vivo, through IL-4- and TLR4-independent pathways. These studies suggest that solid microparticles alone can act as adjuvants to induce potent innate and adaptive Th2-type immune responses and further suggest that wear debris in joint replacements may have Th2-type inflammatory properties.


Subject(s)
Particle Size , Signal Transduction/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Titanium/administration & dosage , Toll-Like Receptor 4/physiology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adoptive Transfer , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Inflammation Mediators/administration & dosage , Inflammation Mediators/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Signal Transduction/drug effects , Th2 Cells/pathology , Titanium/adverse effects , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
11.
J Immunol ; 179(6): 3821-30, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785819

ABSTRACT

Previous studies have suggested that B cells promote Th2 cell development by inhibiting Th1 cell differentiation. To examine whether B cells are directly required for the development of IL-4-producing T cells in the lymph node during a highly polarized Th2 response, B cell-deficient and wild-type mice were inoculated with the nematode parasite, Nippostrongylus brasiliensis. On day 7, in the absence of increased IFN-gamma, IL-4 protein and gene expression from CD4 T cells in the draining lymph nodes were markedly reduced in B cell-deficient mice and could not be restored by multiple immunizations. Using a DO11.10 T cell adoptive transfer system, OVA-specific T cell IL-4 production and cell cycle progression, but not cell surface expression of early activation markers, were impaired in B cell-deficient recipient mice following immunization with N. brasiliensis plus OVA. Laser capture microdissection and immunofluorescent staining showed that pronounced IL-4 mRNA and protein secretion by donor DO11.10 T cells first occurred in the T cell:B cell zone of the lymph node shortly after inoculation of IL-4-/- recipients, suggesting that this microenvironment is critical for initial Th2 cell development. Reconstitution of B cell-deficient mice with wild-type naive B cells, or IL-4-/- B cells, substantially restored Ag-specific T cell IL-4 production. However, reconstitution with B7-1/B7-2-deficient B cells failed to rescue the IL-4-producing DO11.10 T cells. These results suggest that B cells, expressing B7 costimulatory molecules, are required in the absence of an underlying IFN-gamma-mediated response for the development of a polarized primary Ag-specific Th2 response in vivo.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Adoptive Transfer , Amino Acid Sequence , Animals , B-Lymphocytes/parasitology , B-Lymphocytes/pathology , B-Lymphocytes/transplantation , CD4-Positive T-Lymphocytes/parasitology , CD4-Positive T-Lymphocytes/transplantation , Cell Differentiation/genetics , Epitopes, T-Lymphocyte/immunology , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Mice, Transgenic , Molecular Sequence Data , Nippostrongylus/immunology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Strongylida Infections/genetics , Strongylida Infections/immunology , Strongylida Infections/metabolism , Th2 Cells/parasitology , Th2 Cells/pathology
12.
Nat Med ; 12(8): 955-60, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16892038

ABSTRACT

Although primary and memory responses against bacteria and viruses have been studied extensively, T helper type 2 (T(H)2) effector mechanisms leading to host protection against helminthic parasites remain elusive. Examination of the intestinal epithelial submucosa of mice after primary and secondary infections by a natural gastrointestinal parasite revealed a distinct immune-cell infiltrate after challenge, featuring interleukin-4-expressing memory CD4(+) T cells that induced IL-4 receptor(hi) (IL-4R(hi)) CD206(+) alternatively activated macrophages. In turn, these alternatively activated macrophages (AAMacs) functioned as important effector cells of the protective memory response contributing to parasite elimination, demonstrating a previously unknown mechanism for host protection against intestinal helminths.


Subject(s)
Macrophages/immunology , Nematoda/immunology , Nematode Infections/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Adoptive Transfer , Animals , Arginase/antagonists & inhibitors , Boronic Acids/pharmacology , CD4-Positive T-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/transplantation , Electron Transport Complex IV/metabolism , Enzyme Inhibitors/pharmacology , Female , Host-Parasite Interactions , Immunohistochemistry , Injections, Intravenous , Interleukin-4/genetics , Larva/enzymology , Larva/parasitology , Mice , Mice, Inbred BALB C , Mice, Knockout , Nematoda/physiology , Nematode Infections/parasitology , STAT6 Transcription Factor/genetics , Strongylida Infections/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL