Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Eur J Immunol ; 52(4): 656-668, 2022 04.
Article in English | MEDLINE | ID: mdl-34962287

ABSTRACT

B cells play a major role in the pathogenesis of many autoimmune diseases like MS, rheumatoid arthritis, or systemic lupus erythematosus. Depletion of B cells with anti-CD20 antibodies is an established therapy for MS. However, total B-cell depletion will also affect regulatory B cells that are known to suppress autoimmune responses. In our studies, we describe an alternative approach based on targeting CD79b that induces only partial B-cell depletion and achieves therapeutic effects by B-cell modulation. Prophylactic and therapeutic treatment with an antibody against CD79b and also a deglycosylated variant of this antibody, lacking effector function like antibody-dependent cellular cytotoxicity or complement activation, significantly reduced the development and progression of EAE in mice. Our data show that modulation of B cells via CD79b is equally effective as almost complete B-cell depletion with anti-CD20 antibodies and may constitute an alternative approach to treat MS.


Subject(s)
Autoimmune Diseases , Encephalitis , Animals , Antibodies , Antigens, CD20 , Autoimmunity , B-Lymphocytes , Encephalitis/drug therapy , Encephalitis/pathology , Mice
2.
Front Immunol ; 12: 816509, 2021.
Article in English | MEDLINE | ID: mdl-35126373

ABSTRACT

Fibrosis is a prominent feature of chronic allograft rejection, caused by an excessive production of matrix proteins, including collagen-1. Several cell types produce collagen-1, including mesenchymal fibroblasts and cells of hematopoietic origin. Here, we sought to determine whether tissue-resident donor-derived cells or allograft-infiltrating recipient-derived cells are responsible for allograft fibrosis, and whether hematopoietic cells contribute to collagen production. A fully MHC-mismatched mouse heterotopic heart transplantation model was used, with transient depletion of CD4+ T cells to prevent acute rejection. Collagen-1 was selectively knocked out in recipients or donors. In addition, collagen-1 was specifically deleted in hematopoietic cells. Tissue-resident macrophages were depleted using anti-CSF1R antibody. Allograft fibrosis and inflammation were quantified 20 days post-transplantation. Selective collagen-1 knock-out in recipients or donors showed that tissue-resident cells from donor hearts, but not infiltrating recipient-derived cells, are responsible for production of collagen-1 in allografts. Cell-type-specific knock-out experiments showed that hematopoietic tissue-resident cells in donor hearts substantially contributed to graft fibrosis. Tissue resident macrophages, however, were not responsible for collagen-production, as their deletion worsened allograft fibrosis. Donor-derived cells including those of hematopoietic origin determine allograft fibrosis, making them attractive targets for organ preconditioning to improve long-term transplantation outcomes.


Subject(s)
Collagen Type I/biosynthesis , Graft Rejection/etiology , Graft Rejection/metabolism , Heart Transplantation/adverse effects , Tissue Donors , Animals , Biomarkers , Chronic Disease , Collagen Type I/immunology , Disease Models, Animal , Disease Susceptibility , Graft Rejection/diagnosis , Heart Transplantation/methods , Immunophenotyping , Mice , Mice, Transgenic , Transplantation, Homologous
3.
J Immunol ; 202(12): 3514-3523, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31068389

ABSTRACT

Chronic rejection is a major problem in transplantation medicine, largely resistant to therapy, and poorly understood. We have shown previously that basophil-derived IL-4 contributes to fibrosis and vasculopathy in a model of heart transplantation with depletion of CD4+ T cells. However, it is unknown how basophils are activated in the allografts and whether they play a role when cyclosporin A (CsA) immunosuppression is applied. BALB/c donor hearts were heterotopically transplanted into fully MHC-mismatched C57BL/6 recipients and acute rejection was prevented by depletion of CD4+ T cells or treatment with CsA. We found that IL-3 is significantly upregulated in chronically rejecting allografts and is the major activator of basophils in allografts. Using IL-3-deficient mice and depletion of basophils, we show that IL-3 contributes to allograft fibrosis and organ failure in a basophil-dependent manner. Also, in the model of chronic rejection involving CsA, IL-3 and basophils substantially contribute to organ remodeling, despite the almost complete suppression of IL-4 by CsA. In this study, basophil-derived IL-6 that is resistant to suppression by CsA, was largely responsible for allograft fibrosis and limited transplant survival. Our data show that IL-3 induces allograft fibrosis and chronic rejection of heart transplants, and exerts its profibrotic effects by activation of infiltrating basophils. Blockade of IL-3 or basophil-derived cytokines may provide new strategies to prevent or delay the development of chronic allograft rejection.


Subject(s)
Basophils/immunology , Graft Rejection/immunology , Heart Transplantation , Interleukin-3/metabolism , Animals , Cell Movement , Cells, Cultured , Chronic Disease , Disease Models, Animal , Humans , Interleukin-3/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Transplantation, Homologous , Up-Regulation
4.
Cell Immunol ; 334: 49-60, 2018 12.
Article in English | MEDLINE | ID: mdl-30274838

ABSTRACT

A large number of animal models revealed that IL-3 plays an important role for the development of T and B cell-mediated autoimmune diseases. However, little is known about the expression and regulation of IL-3 receptors in human T and B cells and how IL-3 modulates the activation and survival of these cells. We show that the IL-3 receptor CD123 is substantially upregulated on proliferating CD4+ and CD8+ T as well as B cells. Upregulation of CD123 differs between various activators and can be further modulated by cytokines. Exposure of human T and B cells to IL-3 enhances proliferation and survival. IL-3 also induces a shift towards secretion of proinflammatory cytokines in T and B cells and reduces the expression of IL-10 in B cells. Thus IL-3 may have proinflammatory and immunostimulatory properties also in human autoimmune diseases.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-3/immunology , Receptors, Interleukin-3/immunology , Autoimmune Diseases/immunology , Cytokines/immunology , Humans , Interleukin-3 Receptor alpha Subunit/immunology , Lymphocyte Activation/immunology , Up-Regulation/immunology
5.
J Am Soc Nephrol ; 29(7): 1859-1873, 2018 07.
Article in English | MEDLINE | ID: mdl-29777019

ABSTRACT

Background Interstitial fibrosis is associated with chronic renal failure. In addition to fibroblasts, bone marrow-derived cells and tubular epithelial cells have the capacity to produce collagen. However, the amount of collagen produced by each of these cell types and the relevance of fibrosis to renal function are unclear.Methods We generated conditional cell type-specific collagen I knockout mice and used (reversible) unilateral ureteral obstruction and adenine-induced nephropathy to study renal fibrosis and function.Results In these mouse models, hematopoietic, bone marrow-derived cells contributed to 38%-50% of the overall deposition of collagen I in the kidney. The influence of fibrosis on renal function was dependent on the type of damage. In unilateral ureteral obstruction, collagen production by resident fibroblasts was essential to preserve renal function, whereas in the chronic model of adenine-induced nephropathy, collagen production was detrimental to renal function.Conclusions Our data show that hematopoietic cells are a major source of collagen and that antifibrotic therapies need to be carefully considered depending on the type of disease and the underlying cause of fibrosis.


Subject(s)
Acute Kidney Injury/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Kidney/pathology , Renal Insufficiency, Chronic/metabolism , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Adenine , Animals , Bone Marrow Cells/metabolism , Cell Lineage , Epithelial Cells/metabolism , Female , Fibroblasts/metabolism , Fibrosis , Glomerular Filtration Rate , Hematopoiesis , Kidney/physiopathology , Kidney Tubules/cytology , Mice , Mice, Knockout , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Ureteral Obstruction/complications
SELECTION OF CITATIONS
SEARCH DETAIL